[Title 21 CFR ]
[Code of Federal Regulations (annual edition) - April 1, 1998 Edition]
[From the U.S. Government Printing Office]


[[Page i]]

          21



          Food and Drugs



          PARTS 300 TO 499

                         Revised as of April 1, 1998

          CONTAINING
          A CODIFICATION OF DOCUMENTS
          OF GENERAL APPLICABILITY
          AND FUTURE EFFECT
          AS OF APRIL 1, 1998

          With Ancillaries
          Published by
          the Office of the Federal Register
          National Archives and Records
          Administration
          as a Special Edition of
          the Federal Register



[[Page ii]]

                                      




                     U.S. GOVERNMENT PRINTING OFFICE
                            WASHINGTON : 1998



               For sale by U.S. Government Printing Office
 Superintendent of Documents, Mail Stop: SSOP, Washington, DC 20402-9328



[[Page iii]]




                            Table of Contents



                                                                    Page
  Explanation.................................................       v

  Title 21:
    Chapter I--Food and Drug Administration, Department of 
        Health and Human Services (Continued).................       3
  Finding Aids:
    Material Approved for Incorporation by Reference..........    1063
    Table of CFR Titles and Chapters..........................    1065
    Alphabetical List of Agencies Appearing in the CFR........    1081
    List of CFR Sections Affected.............................    1091



[[Page iv]]


      


-------------------------------------------------------------------

   Cite this Code:  CFR

   To cite the regulations in this volume use title, part and
   section number. Thus, 21 CFR 300.50 refers to title 21, part
   300, section 50.

-------------------------------------------------------------------


[[Page v]]



                               EXPLANATION

    The Code of Federal Regulations is a codification of the general and 
permanent rules published in the Federal Register by the Executive 
departments and agencies of the Federal Government. The Code is divided 
into 50 titles which represent broad areas subject to Federal 
regulation. Each title is divided into chapters which usually bear the 
name of the issuing agency. Each chapter is further subdivided into 
parts covering specific regulatory areas.
    Each volume of the Code is revised at least once each calendar year 
and issued on a quarterly basis approximately as follows:

Title 1 through Title 16.................................as of January 1
Title 17 through Title 27..................................as of April 1
Title 28 through Title 41...................................as of July 1
Title 42 through Title 50................................as of October 1
    The appropriate revision date is printed on the cover of each 
volume.

LEGAL STATUS

    The contents of the Federal Register are required to be judicially 
noticed (44 U.S.C. 1507). The Code of Federal Regulations is prima facie 
evidence of the text of the original documents (44 U.S.C. 1510).

HOW TO USE THE CODE OF FEDERAL REGULATIONS

    The Code of Federal Regulations is kept up to date by the individual 
issues of the Federal Register. These two publications must be used 
together to determine the latest version of any given rule.
    To determine whether a Code volume has been amended since its 
revision date (in this case, April 1, 1998), consult the ``List of CFR 
Sections Affected (LSA),'' which is issued monthly, and the ``Cumulative 
List of Parts Affected,'' which appears in the Reader Aids section of 
the daily Federal Register. These two lists will identify the Federal 
Register page number of the latest amendment of any given rule.

EFFECTIVE AND EXPIRATION DATES

    Each volume of the Code contains amendments published in the Federal 
Register since the last revision of that volume of the Code. Source 
citations for the regulations are referred to by volume number and page 
number of the Federal Register and date of publication. Publication 
dates and effective dates are usually not the same and care must be 
exercised by the user in determining the actual effective date. In 
instances where the effective date is beyond the cut-off date for the 
Code a note has been inserted to reflect the future effective date. In 
those instances where a regulation published in the Federal Register 
states a date certain for expiration, an appropriate note will be 
inserted following the text.

OMB CONTROL NUMBERS

    The Paperwork Reduction Act of 1980 (Pub. L. 96-511) requires 
Federal agencies to display an OMB control number with their information 
collection request.

[[Page vi]]

Many agencies have begun publishing numerous OMB control numbers as 
amendments to existing regulations in the CFR. These OMB numbers are 
placed as close as possible to the applicable recordkeeping or reporting 
requirements.

OBSOLETE PROVISIONS

    Provisions that become obsolete before the revision date stated on 
the cover of each volume are not carried. Code users may find the text 
of provisions in effect on a given date in the past by using the 
appropriate numerical list of sections affected. For the period before 
January 1, 1986, consult either the List of CFR Sections Affected, 1949-
1963, 1964-1972, or 1973-1985, published in seven separate volumes. For 
the period beginning January 1, 1986, a ``List of CFR Sections 
Affected'' is published at the end of each CFR volume.

INCORPORATION BY REFERENCE

    What is incorporation by reference? Incorporation by reference was 
established by statute and allows Federal agencies to meet the 
requirement to publish regulations in the Federal Register by referring 
to materials already published elsewhere. For an incorporation to be 
valid, the Director of the Federal Register must approve it. The legal 
effect of incorporation by reference is that the material is treated as 
if it were published in full in the Federal Register (5 U.S.C. 552(a)). 
This material, like any other properly issued regulation, has the force 
of law.
    What is a proper incorporation by reference? The Director of the 
Federal Register will approve an incorporation by reference only when 
the requirements of 1 CFR part 51 are met. Some of the elements on which 
approval is based are:
    (a) The incorporation will substantially reduce the volume of 
material published in the Federal Register.
    (b) The matter incorporated is in fact available to the extent 
necessary to afford fairness and uniformity in the administrative 
process.
    (c) The incorporating document is drafted and submitted for 
publication in accordance with 1 CFR part 51.
    Properly approved incorporations by reference in this volume are 
listed in the Finding Aids at the end of this volume.
    What if the material incorporated by reference cannot be found? If 
you have any problem locating or obtaining a copy of material listed in 
the Finding Aids of this volume as an approved incorporation by 
reference, please contact the agency that issued the regulation 
containing that incorporation. If, after contacting the agency, you find 
the material is not available, please notify the Director of the Federal 
Register, National Archives and Records Administration, Washington DC 
20408, or call (202) 523-4534.

CFR INDEXES AND TABULAR GUIDES

    A subject index to the Code of Federal Regulations is contained in a 
separate volume, revised annually as of January 1, entitled CFR Index 
and Finding Aids. This volume contains the Parallel Table of Statutory 
Authorities and Agency Rules (Table I), and Acts Requiring Publication 
in the Federal Register (Table II). A list of CFR titles, chapters, and 
parts and an alphabetical list of agencies publishing in the CFR are 
also included in this volume.
    An index to the text of ``Title 3--The President'' is carried within 
that volume.
    The Federal Register Index is issued monthly in cumulative form. 
This index is based on a consolidation of the ``Contents'' entries in 
the daily Federal Register.

[[Page vii]]

    A List of CFR Sections Affected (LSA) is published monthly, keyed to 
the revision dates of the 50 CFR titles.

REPUBLICATION OF MATERIAL

    There are no restrictions on the republication of material appearing 
in the Code of Federal Regulations.

INQUIRIES

    For a legal interpretation or explanation of any regulation in this 
volume, contact the issuing agency. The issuing agency's name appears at 
the top of odd-numbered pages.
    For inquiries concerning CFR reference assistance, call 202-523-5227 
or write to the Director, Office of the Federal Register, National 
Archives and Records Administration, Washington, DC 20408.

SALES

    The Government Printing Office (GPO) processes all sales and 
distribution of the CFR. For payment by credit card, call 202-512-1800, 
M-F, 8 a.m. to 4 p.m. e.s.t. or fax your order to 202-512-2233, 24 hours 
a day. For payment by check, write to the Superintendent of Documents, 
Attn: New Orders, P.O. Box 371954, Pittsburgh, PA 15250-7954. For GPO 
Customer Service call 202-512-1803.

ELECTRONIC SERVICES

    The full text of the Code of Federal Regulations, The United States 
Government Manual, the Federal Register, Public Laws, Weekly Compilation 
of Presidential Documents and the Privacy Act Compilation are available 
in electronic format at www.access.gpo.gov/nara (``GPO Access''). For 
more information, contact Electronic Information Dissemination Services, 
U.S. Government Printing Office. Phone 202-512-1530, or 888-293-6498 
(toll-free). E-mail, [email protected].
    The Office of the Federal Register also offers a free service on the 
National Archives and Records Administration's (NARA) World Wide Web 
site for public law numbers, Federal Register finding aids, and related 
information. Connect to NARA's web site at www.nara.gov/fedreg. The NARA 
site also contains links to GPO Access.

                              Raymond A. Mosley,
                                    Director,
                          Office of the Federal Register.

April 1, 1998.



[[Page ix]]



                               THIS TITLE

    Title 21--Food and Drugs is composed of nine volumes. The parts in 
these volumes are arranged in the following order: Parts 1-99, 100-169, 
170-199, 200-299, 300-499, 500-599, 600-799, 800-1299 and 1300-end. The 
first eight volumes, containing parts 1-1299, comprise Chapter I--Food 
and Drug Administration, Department of Health and Human Services. The 
ninth volume, containing part 1300 to end, includes Chapter II--Drug 
Enforcement Administration, Department of Justice, and Chapter III--
Office of National Drug Control Policy. The contents of these volumes 
represent all current regulations codified under this title of the CFR 
as of April 1, 1998.

    Redesignation tables for Chapter I--Food and Drug Administration 
appear in the Finding Aids section for the volumes containing parts 170-
199 and 500-599.

    For this volume, Melanie L. Marcec was Chief Editor. The Code of 
Federal Regulations publication program is under the direction of 
Frances D. McDonald, assisted by Alomha S. Morris.

[[Page x]]




[[Page 1]]



                        TITLE 21--FOOD AND DRUGS




                  (This book contains parts 300 to 499)

----------------------------------------------------------------------
                                                                    Part

Chapter i--Food and Drug Administration, Department of 
  Health and Human Services (Continued).....................         300

Cross References: Food Safety and Inspection Service, Department of 
  Agriculture: See Meat and Poultry Inspection, 9 CFR chapter III.

  Federal Trade Commission: See Commercial Practices, 16 CFR chapter I.

  United States Customs Service, Department of the Treasury: See Customs 
Duties, 19 CFR chapter I.

  Internal Revenue Service, Department of the Treasury: See Internal 
Revenue, 26 CFR chapter I.

  Bureau of Alcohol, Tobacco, and Firearms, Department of the Treasury: 
See Alcohol, Tobacco Products and Firearms, 27 CFR chapter I.

[[Page 3]]



CHAPTER I--FOOD AND DRUG ADMINISTRATION, DEPARTMENT OF HEALTH AND HUMAN SERVICES--Continued




                           (Parts 300 to 499)

  --------------------------------------------------------------------

  Editorial Note:  The Food and Drug Administration published a document 
at 49 FR 41019, Oct. 19, 1984, establishing July 1, 1987, as a uniform 
effective date for compliance for regulations affecting the labeling of 
food products. At 51 FR 34085, Sept. 25, 1986, FDA established January 
1, 1989, as a new uniform effective date for compliance. The new uniform 
effective date will apply only to final FDA food labeling regulations 
published after July 1, 1986, and before January 1, 1988. At 55 FR 276, 
Jan. 4, 1990, FDA established January 1, 1993 as a new uniform effective 
date for compliance. The new uniform effective date will apply only to 
final FDA food labeling regulations published after January 1, 1990 and 
before January 1, 1992.

                    SUBCHAPTER D--DRUGS FOR HUMAN USE

Part                                                                Page
300             General.....................................           7
310             New drugs...................................           7
312             Investigational new drug application........          61
314             Applications for FDA approval to market a 
                    new drug or an antibiotic drug..........          99
316             Orphan drugs................................         179
320             Bioavailability and bioequivalence 
                    requirements............................         190
328             Over-the-counter drug products intended for 
                    oral ingestion that contain alcohol.....         205
329             Habit-forming drugs.........................         206
330             Over-the-counter (OTC) human drugs which are 
                    generally recognized as safe and 
                    effective and not misbranded............         211
331             Antacid products for over-the-counter (OTC) 
                    human use...............................         223
332             Antiflatulent products for over-the-counter 
                    human use...............................         227
333             Topical antimicrobial drug products for 
                    over-the-counter human use..............         228
336             Antiemetic drug products for over-the-
                    counter human use.......................         236

[[Page 4]]

338             Nighttime sleep-aid drug products for over-
                    the-counter human use...................         238
340             Stimulant drug products for over-the-counter 
                    human use...............................         239
341             Cold, cough, allergy, bronchodilator, and 
                    antiasthmatic drug products for over-
                    the-counter human use...................         240
344             Topical OTIC drug products for over-the-
                    counter human use.......................         256
346             Anorectal drug products for over-the-counter 
                    human use...............................         257
347             Skin protectant drug products for over-the-
                    counter human use.......................         262
348             External analgesic drug products for over-
                    the-counter human use...................         263
349             Ophthalmic drug products for over-the-
                    counter human use.......................         265
355             Anticaries drug products for over-the-
                    counter human use.......................         270
357             Miscellaneous internal drug products for 
                    over-the-counter human use..............         275
358             Miscellaneous external drug products for 
                    over-the-counter human use..............         279
361             Prescription drugs for human use generally 
                    recognized as safe and effective and not 
                    misbranded: Drugs used in research......         285
369             Interpretative statements re warnings on 
                    drugs and devices for over-the-counter 
                    sale....................................         290
429             Drugs composed wholly or partly of insulin..         299
430             Antibiotic drugs; general...................         310
431             Certification of antibiotic drugs...........         331
432             Packaging and labeling of antibiotic drugs..         339
433             Exemptions from antibiotic certification and 
                    labeling requirements...................         340
436             Tests and methods of assay of antibiotic and 
                    antibiotic-containing drugs.............         351
440             Penicillin antibiotic drugs.................         483
441             Penem antibiotic drugs......................         588
442             Cepha antibiotic drugs......................         593
443             Carbacephem antibiotic drugs................         696
444             Oligosaccharide antibiotic drugs............         699
446             Tetracycline antibiotic drugs...............         761
448             Peptide antibiotic drugs....................         826
449             Antifungal antibiotic drugs.................         865
450             Antitumor antibiotic drugs..................         890
452             Macrolide antibiotic drugs..................         911
453             Lincomycin antibiotic drugs.................         949

[[Page 5]]

455             Certain other antibiotic drugs..............         968
460             Antibiotic drugs intended for use in 
                    laboratory diagnosis of disease.........        1019
461-499

[Reserved]

  Editorial Note: For nomenclature changes to chapter I see 59 FR 14366, 
Mar. 28, 1994.

[[Page 7]]



                    SUBCHAPTER D--DRUGS FOR HUMAN USE





PART 300--GENERAL--Table of Contents




                          Subpart A  [Reserved]

                      Subpart B--Combination Drugs

Sec.
300.50  Fixed-combination prescription drugs for humans.

          Subpart C--Substances Generally Prohibited From Drugs

300.100  Chlorofluorocarbon propellants.

    Authority:  21 U.S.C. 331, 351, 352, 355, 357, 360b, 361, 371.



                          Subpart A  [Reserved]



                      Subpart B--Combination Drugs



Sec. 300.50  Fixed-combination prescription drugs for humans.

    The Food and Drug Administration's policy in administering the new-
drug, antibiotic, and other regulatory provisions of the Federal Food, 
Drug, and Cosmetic Act regarding fixed combination dosage form 
prescription drugs for humans is as follows:
    (a) Two or more drugs may be combined in a single dosage form when 
each component makes a contribution to the claimed effects and the 
dosage of each component (amount, frequency, duration) is such that the 
combination is safe and effective for a significant patient population 
requiring such concurrent therapy as defined in the labeling for the 
drug. Special cases of this general rule are where a component is added:
    (1) To enhance the safety or effectiveness of the principal active 
component; and
    (2) To minimize the potential for abuse of the principal active 
component.
    (b) If a combination drug presently the subject of an approved new-
drug application or antibiotic monograph has not been recognized as 
effective by the Commissioner of Food and Drugs based on his evaluation 
of the appropriate National Academy of Sciences-National Research 
Council panel report, or if substantial evidence of effectiveness has 
not otherwise been presented for it, then formulation, labeling, or 
dosage changes may be proposed and any resulting formulation may meet 
the appropriate criteria listed in paragraph (a) of this section.
    (c) A fixed-combination prescription drug for humans that has been 
determined to be effective for labeled indications by the Food and Drug 
Administration, based on evaluation of the NAS-NRC report on the 
combination, is considered to be in compliance with the requirements of 
this section.0
[40 FR 13496, Mar. 27, 1975]



          Subpart C--Substances Generally Prohibited From Drugs



Sec. 300.100  Chlorofluorocarbon propellants.

    The use of chlorofluorocarbons in human drugs as propellants in 
self-pressurized containers is generally prohibited except as provided 
by Sec. 2.125 of this chapter.
[43 FR 11317, Mar. 17, 1978]



PART 310--NEW DRUGS--Table of Contents




                      Subpart A--General Provisions

Sec.
310.3  Definitions and interpretations.
310.4  Biologics; products subject to license control.
310.6  Applicability of ``new drug'' or safety or effectiveness findings 
          in drug efficacy study implementation notices and notices of 
          opportunity for hearing to identical, related, and similar 
          drug products.

        Subpart B--Specific Administrative Rulings and Decisions

310.100  New drug status opinions; statement of policy.
310.103  New drug substances intended for hypersensitivity testing.

 Subpart C--New Drugs Exempted From Prescription-Dispensing Requirements

310.200  Prescription-exemption procedure.
310.201  Exemption for certain drugs limited by new drug applications to 
          prescription sale.

[[Page 8]]

                     Subpart D--Records and Reports

310.303  Continuation of long-term studies, records, and reports on 
          certain drugs for which new drug applications have been 
          approved.
310.305  Records and reports concerning adverse drug experiences on 
          marketed prescription drugs for human use without approved new 
          drug applications.

        Subpart E--Requirements for Specific New Drugs or Devices

310.500  Digoxin products for oral use; conditions for marketing.
310.501  Patient package inserts for oral contraceptives.
310.502  Certain drugs accorded new drug status through rulemaking 
          procedures.
310.503  Requirements regarding certain radioactive drugs.
310.509  Parenteral drug products in plastic containers.
310.515  Patient package inserts for estrogens.
310.516  Progestational drug products; labeling directed to the patient.
310.517  Labeling for oral hypoglycemic drugs of the sulfonylurea class.
310.518  Drug products containing iron or iron salts.
310.519  Drug products marketed as over-the-counter (OTC) daytime 
          sedatives.
310.527  Drug products containing active ingredients offered over-the-
          counter (OTC) for external use as hair growers or for hair 
          loss prevention.
310.528  Drug products containing active ingredients offered over-the-
          counter (OTC) for use as an aphrodisiac.
310.529  Drug products containing active ingredients offered over-the-
          counter (OTC) for oral use as insect repellents.
310.530  Topically applied hormone-containing drug products for over-
          the-counter (OTC) human use.
310.531  Drug products containing active ingredients offered over-the-
          counter (OTC) for the treatment of boils.
310.532  Drug products containing active ingredients offered over-the-
          counter (OTC) to relieve the symptoms of benign prostatic 
          hypertrophy.
310.533  Drug products containing active ingredients offered over-the-
          counter (OTC) for human use as an anticholinergic in cough-
          cold drug products.
310.534  Drug products containing active ingredients offered over-the-
          counter (OTC) for human use as oral wound healing agents.
310.536  Drug products containing active ingredients offered over-the-
          counter (OTC) for use as a nailbiting or thumbsucking 
          deterrent.
310.537  Drug products containing active ingredients offered over-the-
          counter (OTC) for oral administration for the treatment of 
          fever blisters and cold sores.
310.538  Drug products containing active ingredients offered over-the-
          counter (OTC) for use for ingrown toenail relief.
310.540  Drug products containing active ingredients offered over-the-
          counter (OTC) for use as stomach acidifiers.
310.541  Over-the-counter (OTC) drug products containing active 
          ingredients offered for use in the treatment of 
          hypophosphatemia.
310.542  Over-the-counter (OTC) drug products containing active 
          ingredients offered for use in the treatment of 
          hyperphosphatemia.
310.543  Drug products containing active ingredients offered over-the-
          counter (OTC) for human use in exocrine pancreatic 
          insufficiency.
310.544  Drug products containing active ingredients offered over-the-
          counter (OTC) for use as a smoking deterrent.
310.545  Drug products containing certain active ingredients offered 
          over-the-counter (OTC) for certain uses.
310.546  Drug products containing active ingredients offered over-the-
          counter (OTC) for the treatment and/or prevention of nocturnal 
          leg muscle cramps.
310.547  Drug products containing quinine offered over-the-counter (OTC) 
          for the treatment and/or prevention of malaria.

    Authority:  21 U.S.C. 321, 331, 351, 352, 353, 355, 356, 357, 360b-
360f, 360j, 361(a), 371, 374, 375, 379e; 42 U.S.C. 216, 241, 242(a), 
262, 263b-263n.



                      Subpart A--General Provisions



Sec. 310.3  Definitions and interpretations.

    As used in this part:
    (a) The term act means the Federal Food, Drug, and Cosmetic Act, as 
amended (secs. 201-902, 52 Stat. 1040 et seq., as amended; 21 U.S.C. 
321-392).
    (b) Department means the Department of Health and Human Services.

[[Page 9]]

    (c) Secretary means the Secretary of Health and Human Services.
    (d) Commissioner means the Commissioner of Food and Drugs.
    (e) The term person includes individuals, partnerships, 
corporations, and associations.
    (f) The definitions and interpretations of terms contained in 
section 201 of the act shall be applicable to such terms when used in 
the regulations in this part.
    (g) New drug substance means any substance that when used in the 
manufacture, processing, or packing of a drug, causes that drug to be a 
new drug, but does not include intermediates used in the synthesis of 
such substance.
    (h) The newness of a drug may arise by reason (among other reasons) 
of:
    (1) The newness for drug use of any substance which composes such 
drug, in whole or in part, whether it be an active substance or a 
menstruum, excipient, carrier, coating, or other component.
    (2) The newness for a drug use of a combination of two or more 
substances, none of which is a new drug.
    (3) The newness for drug use of the proportion of a substance in a 
combination, even though such combination containing such substance in 
other proportion is not a new drug.
    (4) The newness of use of such drug in diagnosing, curing, 
mitigating, treating, or preventing a disease, or to affect a structure 
or function of the body, even though such drug is not a new drug when 
used in another disease or to affect another structure or function of 
the body.
    (5) The newness of a dosage, or method or duration of administration 
or application, or other condition of use prescribed, recommended, or 
suggested in the labeling of such drug, even though such drug when used 
in other dosage, or other method or duration of administration or 
application, or different condition, is not a new drug.
    (i) [Reserved]
    (j) The term sponsor means the person or agency who assumes 
responsibility for an investigation of a new drug, including 
responsibility for compliance with applicable provisions of the act and 
regulations. The ``sponsor'' may be an individual, partnership, 
corporation, or Government agency and may be a manufacturer, scientific 
institution, or an investigator regularly and lawfully engaged in the 
investigation of new drugs.
    (k) The phrase related drug(s) includes other brands, potencies, 
dosage forms, salts, and esters of the same drug moiety, including 
articles prepared or manufactured by other manufacturers: and any other 
drug containing a component so related by chemical structure or known 
pharmacological properties that, in the opinion of experts qualified by 
scientific training and experience to evaluate the safety and 
effectiveness of drugs, it is prudent to assume or ascertain the 
liability of similar side effects and contraindications.
    (l) Special packaging as defined in section 2(4) of the Poison 
Prevention Packaging Act of 1970 means packaging that is designed or 
constructed to be significantly difficult for children under 5 years of 
age to open or obtain a toxic or harmful amount of the substance 
contained therein within a reasonable time and not difficult for normal 
adults to use properly, but does not mean packaging which all such 
children cannot open or obtain a toxic or harmful amount within a 
reasonable time.
    (m) [Reserved]
    (n) The term radioactive drug means any substance defined as a drug 
in section 201(g)(1) of the Federal Food, Drug, and Cosmetic Act which 
exhibits spontaneous disintegration of unstable nuclei with the emission 
of nuclear particles or photons and includes any nonradioactive reagent 
kit ornuclide generator which is intended to be used in the preparation 
of any such substance but does not include drugs such as carbon-
containing compounds or potassium-containing salts which contain trace 
quantities of naturally occurring radionuclides. The term ``radioactive 
drug'' includes a ``radioactive biological product'' as defined in 
Sec. 600.3(ee) of this chapter.
[39 FR 11680, Mar. 29, 1974, as amended at 39 FR 20484, June 11, 1974; 
40 FR 31307, July 25, 1975; 46 FR 8952, Jan. 27, 1981; 50 FR 7492, Feb. 
22, 1985]

[[Page 10]]



Sec. 310.4  Biologics; products subject to license control.

    (a) Except for radioactive biological products intended for human 
use, a new drug shall not be deemed to be subject to section 505 of the 
act if it is a drug licensed under the Public Health Service Act of July 
1, 1944 (58 Stat. 682, as amended (42 U.S.C. 201 et seq.)) or under the 
animal virus, serum, and toxin law of March 4, 1913 (37 Stat. 832 (21 
U.S.C. 151 et seq.)).
    (b) A radioactive biological product (as defined in Sec. 600.3(ee) 
of this chapter) intended for human use is subject to section 505 of the 
act. Any license for such a radioactive biological product which is 
issued under the Public Health Service Act of July 1, 1944 (58 Stat. 
682, as amended (42 U.S.C. 201 et seq.)) and which has not been revoked 
or suspended as of August 25, 1975 shall constitute an approved new drug 
application in effect under the same terms and conditions as set forth 
in such license and such portions of the establishment license relating 
to such product, which include data and information required under part 
314 of this chapter for a new drug application. Any such radioactive 
biological product for which licensure under the Public Health Service 
Act is pending on August 25, 1975 shall, upon determination that it is 
acceptable for licensure, be approved as a new drug application in lieu 
of issuance of a biological product license.
[40 FR 31312, July 25, 1975]



Sec. 310.6  Applicability of ``new drug'' or safety or effectiveness findings in drug efficacy study implementation notices and notices of opportunity for 
          hearing to identical, related, and similar drug products.

    (a) The Food and Drug Administration's conclusions on the 
effectiveness of drugs are currently being published in the Federal 
Register as Drug Efficacy Study Implementation (DESI) Notices and as 
Notices of Opportunity for Hearing. The specific products listed in 
these notices include only those that were introduced into the market 
through the new drug procedures from 1938-62 and were submitted for 
review by the National Academy of Sciences-National Research Council 
(NAS-NRC), Drug Efficacy Study Group. Many products which are identical 
to, related to, or similar to the products listed in these notices have 
been marketed under different names or by different firms during this 
same period or since 1962 without going through the new drug procedures 
or the Academy review. Even though these products are not listed in the 
notices, they are covered by the new drug applications reviewed and thus 
are subject to these notices. All persons with an interest in a product 
that is identical, related, or similar to a drug listed in a drug 
efficacy notice or a notice of opportunity for a hearing will be given 
the same opportunity as the applicant to submit data and information, to 
request a hearing, and to participate in any hearing. It is not feasible 
for the Food and Drug Administration to list all products which are 
covered by an NDA and thus subject to each notice. However, it is 
essential that the findings and conclusions that a drug product is a 
``new drug'' or that there is a lack of evidence to show that a drug 
product is safe or effective be applied to all identical, related, and 
similar drug products to which they are reasonably applicable. Any 
product not in compliance with an applicable drug efficacy notice is in 
violation of section 505 (new drugs) and/or section 502 (misbranding) of 
the act.
    (b)(1) An identical, related, or similar drug includes other brands, 
potencies, dosage forms, salts, and esters of the same drug moiety as 
well as of any drug moiety related in chemical structure or known 
pharmacological properties.
    (2) Where experts qualified by scientific training and experience to 
evaluate the safety and effectiveness of drugs would conclude that the 
findings and conclusions, stated in a drug efficacy notice or notice of 
opportunity for hearing, that a drug product is a ``new drug'' or that 
there is a lack of evidence to show that a drug product is safe or 
effective are applicable to an identical, related, or similar drug 
product, such product is affected by the notice. A combination drug 
product containing a drug that is identical, related, or similar to a 
drug named in a notice may also be subject to the findings and 
conclusions in a notice that a drug

[[Page 11]]

product is a ``new drug'' or that there is a lack of evidence to show 
that a drug product is safe or effective.
    (3) Any person may request an opinion on the applicability of such a 
notice to a specific product by writing to the Food and Drug 
Administration at the address shown in paragraph (e) of this section.
    (c) Manufacturers and distributors of drugs should review their 
products as drug efficacy notices are published and assure that 
identical, related, or similar products comply with all applicable 
provisions of the notices.
    (d) The published notices and summary lists of the conclusions are 
of particular interest to drug purchasing agents. These agents should 
take particular care to assure that the same purchasing policy applies 
to drug products that are identical, related, or similar to those named 
in the drug efficacy notices. The Food and Drug Administration applies 
the same regulatory policy to all such products. In many instances a 
determination can readily be made as to the applicability of a drug 
efficacy notice by an individual who is knowledgeable about drugs and 
their indications for use. Where the relationships are more subtle and 
not readily recognized, the purchasing agent may request an opinion by 
writing to the Food and Drug Administration at the address shown in 
paragraph (e) of this section.
    (e) Interested parties may submit to the Food and Drug 
Administration, Center for Drug Evaluation and Research, Office of 
Compliance, HFD-300, 5600 Fishers Lane, Rockville, MD 20857, the names 
of drug products, and of their manufacturers or distributors, that 
should be the subject of the same purchasing and regulatory policies as 
those reviewed by the Drug Efficacy Study Group. Appropriate action, 
including referral to purchasing officials of various government 
agencies, will be taken.
    (f) This regulation does not apply to OTC drugs identical, similar, 
or related to a drug in the Drug Efficacy Study unless there has been or 
is notification in the Federal Register that a drug will not be subject 
to an OTC panel review pursuant to Secs. 330.10, 330.11, and 330.5 of 
this chapter.
[39 FR 11680, Mar. 29, 1974, as amended at 48 FR 2755, Jan. 21, 1983; 50 
FR 8996, Mar. 6, 1985; 55 FR 11578, Mar. 29, 1990]



        Subpart B--Specific Administrative Rulings and Decisions



Sec. 310.100  New drug status opinions; statement of policy.

    (a) Over the years since 1938 the Food and Drug Administration has 
given informal advice to inquirers as to the new drug status of 
preparations. These drugs have sometimes been identified only by general 
statements of composition. Generally, such informal opinions were 
incorporated in letters that did not explicitly relate all of the 
necessary conditions and qualifications such as the quantitative formula 
for the drug and the conditions under which it was prescribed, 
recommended, or suggested. This has contributed to misunderstanding and 
misinterpretation of such opinions.
    (b) These informal opinions that an article is ``not a new drug'' or 
``no longer a new drug'' require reexamination under the Kefauver-Harris 
Act (Public Law 87-781; 76 Stat. 788-89). In particular, when approval 
of a new drug application is withdrawn under provisions of section 
505(e) of the Federal Food, Drug, and Cosmetic Act, a drug generally 
recognized as safe may become a ``new drug'' within the meaning of 
section 201(p) of said act as amended by the Kefauver-Harris Act on 
October 10, 1962. This is of special importance by reason of proposed 
actions to withdraw approval of new drug applications for lack of 
substantial evidence of effectiveness as a result of reports of the 
National Academy of Sciences--National Research Council on its review of 
drug effectiveness; for example, see the notice published in the Federal 
Register of January 23, 1968 (33 FR 818), regarding rutin, quercetin, et 
al.
    (c) Any marketed drug is a ``new drug'' if any labeling change made 
after October 9, 1962, recommends or suggests new conditions of use 
under which the drug is not generally recognized as safe and effective 
by qualified experts. Undisclosed or unreported side

[[Page 12]]

effects as well as the emergence of new knowledge presenting questions 
with respect to the safety or effectiveness of a drug may result in its 
becoming a ``new drug'' even though it was previously considered ``not a 
new drug.'' Any previously given informal advice that an article is 
``not a new drug'' does not apply to such an article if it has been 
changed in formulation, manufacture control, or labeling in a way that 
may significantly affect the safety of the drug.
    (d) For these reasons, all opinions previously given by the Food and 
Drug Administration to the effect that an article is ``not a new drug'' 
or is ``no longer a new drug'' are hereby revoked. This does not mean 
that all articles that were the subjects of such prior opinions will be 
regarded as new drugs. The prior opinions will be replaced by opinions 
of the Food and Drug Administration that are qualified and current on 
when an article is ``not a new drug,'' as set forth in this subchapter.
[39 FR 11680, Mar. 29, 1974]



Sec. 310.103  New drug substances intended for hypersensitivity testing.

    (a) The Food and Drug Administration is aware of the need in the 
practice of medicine for the ingredients of a new drug to be available 
for tests of hypersensitivity to such ingredients and therefore will not 
object to the shipment of a new drug substance, as defined in 
Sec. 310.3(g), for such purpose if all of the following conditions are 
met:
    (1) The shipment is made as a result of a specific request made to 
the manufacturer or distributor by a practitioner licensed by law to 
administer such drugs, and the use of such drugs for patch testing is 
not promoted by the manufacturer or distributor.
    (2) The new drug substance requested is an ingredient in a marketed 
new drug and is not one that is an ingredient solely in a new drug that 
is legally available only under the investigational drug provisions of 
this part.
    (3) The label bears the following prominently placed statements in 
lieu of adequate directions for use and in addition to complying with 
the other labeling provisions of the act:
    (i) ``Caution: Federal law prohibits dispensing without a 
prescription''; and
    (ii) ``For use only in patch testing''.
    (4) The quantity shipped is limited to an amount reasonable for the 
purpose of patch testing in the normal course of the practice of 
medicine and is used solely for such patch testing.
    (5) The new drug substance is manufactured by the same procedures 
and meets the same specifications as the component used in the finished 
dosage form.
    (6) The manufacturer or distributor maintains records of all 
shipments for this purpose for a period of 2 years after shipment and 
will make them available to the Food and Drug Administration on request.
    (b) When the requested new drug substance is intended for 
investigational use in humans or the substance is legally available only 
under the investigational drug provisions of part 312 of this chapter, 
the submission of an ``Investigational New Drug Application'' (IND) is 
required. The Food and Drug Administration will offer assistance to any 
practitioner wishing to submit an Investigational New Drug Application.
    (c) This section does not apply to drugs or their components that 
are subject to the licensing requirements of the Public Health Service 
Act of 1944, as amended. (See subchapter F--Biologics, of this chapter.)
[39 FR 11680, Mar. 29, 1974, as amended at 55 FR 11578, Mar. 29, 1990]



 Subpart C--New Drugs Exempted From Prescription-Dispensing Requirements



Sec. 310.200  Prescription-exemption procedure.

    (a) Duration of prescription requirement. Any drug limited to 
prescription use under section 503(b)(1)(C) of the act remains so 
limited until it is exempted as provided in paragraph (b) or (e) of this 
section.
    (b) Prescription-exemption procedure for drugs limited by a new drug 
application. Any drug limited to prescription use under section 
503(b)(1)(C) of the act shall be exempted from prescription-dispensing 
requirements when the Commissioner finds such requirements are not 
necessary for the protection of the public health by reason of the

[[Page 13]]

drug's toxicity or other potentiality for harmful effect, or the method 
of its use, or the collateral measures necessary to its use, and he 
finds that the drug is safe and effective for use in self-medication as 
directed in proposed labeling. A proposal to exempt a drug from the 
prescription-dispensing requirements of section 503(b)(1)(C) of the act 
may be initiated by the Commissioner or by any interested person. Any 
interested person may file a petition seeking such exemption, which 
petition may be pursuant to part 10 of this chapter, or in the form of a 
supplement to an approved new drug application.
    (c) New drug status of drugs exempted from the prescription 
requirement. A drug exempted from the prescription requirement under the 
provisions of paragraph (b) of this section is a ``new drug'' within the 
meaning of section 201(p) of the act until it has been used to a 
material extent and for a material time under such conditions except as 
provided in paragraph (e) of this section.
    (d) Prescription legend not allowed on exempted drugs. The use of 
the prescription caution statement quoted in section 503(b) (4) of the 
act, in the labeling of a drug exempted under the provisions of this 
section, constitutes misbranding. Any other statement or suggestion in 
the labeling of a drug exempted under this section, that such drug is 
limited to prescription use, may constitute misbranding.
    (e) Prescription-exemption procedure of OTC drug review. A drug 
limited to prescription use under section 503(b)(1)(C) of the act may 
also be exempted from prescription-dispensing requirements by the 
procedure set forth in Sec. 330.13 of this chapter.
[39 FR 11680, Mar. 29, 1974, as amended at 41 FR 32582, Aug. 4, 1976; 42 
FR 4714, Jan. 25, 1977; 42 FR 15674, Mar. 22, 1977]



Sec. 310.201  Exemption for certain drugs limited by new-drug applications to prescription sale.

    (a) The prescription-dispensing requirements of section 503(b)(1)(C) 
of the Federal Food, Drug, and Cosmetic Act are not necessary for the 
protection of the public health with respect to the following drugs 
subject to new drug applications:
    (1) N-Acetyl-p-aminophenol (acetaminophen, p-hydroxy-acetanilid) 
preparations meeting all the following conditions:
    (i) The N-acetyl-p-aminophenol is prepared, with or without other 
drugs, in tablet or other dosage form suitable for oral use in self-
medication, and containing no drug limited to prescription sale under 
the provisions of section 503(b)(1) of the act.
    (ii) The N-acetyl-p-aminophenol and all other components of the 
preparation meet their professed standards of identity, strength, 
quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505 (b) of the act is approved for it.
    (iv) The preparation contains not more than 0.325 gram (5 grains) of 
N-acetyl-p-aminophenol per dosage unit, or if it is in liquid form not 
more than 100 milligrams of N-acetyl-p-aminophenol per milliliter.
    (v) The preparation is labeled with adequate directions for use in 
minor conditions as a simple analgesic.
    (vi) The dosages of N-acetyl-p-aminophenol recommended or suggested 
in the labeling do not exceed: For adults, 0.65 gram (10 grains) per 
dose or 2.6 grams (40 grains) per 24-hour period: for children 6 to 12 
years of age, one-half of the maximum adult dose or dosage; for children 
3 to 6 years of age, one-fifth of the maximum adult dose or dosage.
    (vii) The labeling bears, in juxtaposition with the dosage 
recommendations, a clear warning statement against administration of the 
drug to children under 3 years of age and against use of the drug for 
more than 10 days, unless such uses are directed by a physician.
    (viii) If the article is offered for use in arthritis or rheumatism, 
the labeling prominently bears a statement that the beneficial effects 
claimed are limited to the temporary relief of minor aches and pains of 
arthritis and rheumatism and, in juxtaposition with directions for use 
in such conditions, a conspicuous warning statement, such as ``Caution: 
If pain persists for more than 10 days, or redness is present, or in 
conditions affecting children under 12 years of age, consult a physician 
immediately''.

[[Page 14]]

    (2) Sodium gentisate (sodium-2, 5-dihydroxybenzoate) preparations 
meeting all the following conditions:
    (i) The sodium gentisate is prepared, with or without other drugs, 
in tablet or other dosage form suitable for oral use in self-medication, 
and containing no drug limited to prescription sale under the provisions 
of section 503(b)(1) of the act.
    (ii) The sodium gentisate and all other components of the 
preparation meet their professed standards of identity, strength, 
quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 0.5 gram (7.7 grains) of 
anhydrous sodium gentisate per dosage unit.
    (v) The preparation is labeled with adequate directions for use in 
minor conditions as a simple analgesic.
    (vi) The dosages of sodium gentisate recommended or suggested in the 
labeling do not exceed: For adults, 0.5 gram (7.7 grains) per dose of 
2.0 grams (31 grains) per 24-hour period; for children 6 to 12 years of 
age, one-half of the maximum adult dose or dosage.
    (vii) The labeling bears, in juxtaposition with the dosage 
recommendations, a clear warning statement against administration of the 
drug to children under 6 years of age and against use of the drug for a 
prolonged period, except as such uses may be directed by a physician.
    (3) Isoamylhydrocupreine and zolamine hydrochloride (N, N-dimethyl-
N'-2-thiazolyl-N'-p-methoxybenzyl-ethyl- enediamine hydrochloride) 
preparations meeting all the following conditions:
    (i) The isoamylhydrocupreine and zolamine hydrochloride are prepared 
in dosage form suitable for self-medication as rectal suppositories or 
as an ointment and containing no drug limited to prescription sale under 
the provisions of section 503(b)(1) of the act.
    (ii) The isoamylhydrocupreine, zola-amine hydrochloride, and all 
other components of the preparation meet their professed standards of 
identity, strength, quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 0.25 percent of 
isoamylhydrocupreine and 1.0 percent of zolamine hydrochloride.
    (v) If the preparation is in suppository form, it contains not more 
than 5.0 milligrams of isoamylhydrocupreine and not more than 20.0 
milligrams of zolamine hydrochloride per suppository.
    (vi) The preparation is labeled with adequate directions for use in 
the temporary relief of local pain and itching associated with 
hemorrhoids.
    (vii) The directions provide for the use of not more than two 
suppositories or two applications of ointment in a 24-hour period.
    (viii) The labeling bears, in juxtaposition with the dosage 
recommendations, a clear warning statement against use of the 
preparation in case of rectal bleeding, as this may indicate serious 
disease.
    (4) Phenyltoloxamine dihydrogen citrate (N,N-dimethyl-(a-phenyl-O-
toloxy) ethylamine dihydrogen citrate), preparations meeting all the 
following conditions:
    (i) The phenyltoloxamine dihydrogen citrate is prepared, with or 
without other drugs, in tablet or other dosage form suitable for oral 
use in self-medication, and containing no drug limited to prescription 
sale under the provisions of section 503(b)(1) of the act.
    (ii) The phenyltoloxamine dihydrogen citrate and all other 
components of the preparation meet their professed standards of 
identity, strength, quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 88 milligrams of 
phenyltoloxamine dihydrogen citrate (equivalent to 50 milligrams of 
phenyltoloxamine) per dosage unit.
    (v) The preparation is labeled with adequate directions for use in 
the temporary relief of the symptoms of hay fever and/or the symptoms of 
other minor conditions in which it is indicated.

[[Page 15]]

    (vi) The dosages recommended or suggested in the labeling do not 
exceed: For adults, 88 milligrams of phenyltoloxamine dihydrogen citrate 
(equivalent to 50 milligrams of phenyltoloxamine) per dose or 264 
milligrams of phenyltoloxamine dihydrogen citrate (equivalent to 150 
milligrams of phenyltoloxamine) per 24-hour period; for children 6 to 12 
years of age, one-half of the maximum adult dose or dosage.
    (vii) The labeling bears, in juxtaposition with the dosage 
recommendations:
    (a) Clear warning statements against administration of the drug to 
children under 6 years of age, except as directed by a physician, and 
against driving a car or operating machinery while using the drug, since 
it may cause drowsiness.
    (b) If the article is offered for temporary relief of the symptoms 
of colds, a statement that continued administration for such use should 
not exceed 3 days, except as directed by a physician.
    (5)-(7) [Reserved]
    (8) Dicyclomine hydrochloride (1-cyclohexylhexahydrobenzoic acid. 
-diethylaminoethyl ester hydrochloride; diethylaminocarbethoxy-
bicyclohexyl hydrochloride) preparations meeting all the following 
conditions:
    (i) The dicyclomine hydrochloride is prepared with suitable antacid 
and other components, in tablet or other dosage form for oral use in 
self-medication, and containing no drug limited to prescription sale 
under the provisions of section 503(b)(1) of the act.
    (ii) The dicyclomine hydrochloride and all other components of the 
preparation meet their professed standards of identity, strength, 
quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 5 milligrams of 
dicyclomine hydrochloride per dosage unit, or if it is in liquid form 
not more than 0.5 milligram of dicyclomine hydrochloride per milliliter.
    (v) The preparation is labeled with adequate directions for use only 
by adults and children over 12 years of age, in the temporary relief of 
gastric hyperacidity.
    (vi) The dosages recommended or suggested in the directions for use 
do not exceed 10 milligrams of dicyclomine hydrochloride per dose or 30 
milligrams in a 24-hour period.
    (vii) The labeling bears, in juxtaposition with the dosage 
recommendations, clear warning statements against:
    (a) Exceeding the recommended dosage.
    (b) Prolonged use, except as directed by a physician, since 
persistent or recurring symptoms may indicate a serious disease 
requiring medical attention.
    (c) Administration to children under 12 years of age except as 
directed by a physician.
    (9)-(10) [Reserved]
    (11) Hexadenol (a mixture of tetracosanes and their oxidation 
products) preparations meeting all the following conditions:
    (i) The hexadenol is prepared and packaged, with or without other 
drugs, solvents, and propellants, in a form suitable for self-medication 
by external application to the skin as a spray, and containing no drug 
limited to prescription sale under the provisions of section 503(b)(1) 
of the act.
    (ii) The hexadenol and all other components of the preparation meet 
their professed standards of identity, strength, quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 5 percent by weight of 
hexadenol.
    (v) The preparation is labeled with adequate directions for use by 
external application in the treatment of minor burns and minor skin 
irritations.
    (vi) The labeling bears, in juxtaposition with the directions for 
use, clear warning statements against:
    (a) Use on serious burns or skin conditions or prolonged use, except 
as directed by a physician.
    (b) Spraying the preparation in the vicinity of eyes, mouth, nose, 
or ears.
    (12) Sulfur dioxide preparations meeting all the following 
conditions:
    (i) The sulfur dioxide is prepared with or without other drugs, in 
an aqueous

[[Page 16]]

solution packaged in a hermetic container suitable for use in self-
medication by external application to the skin, and containing no drug 
limited to prescription sale under the provisions of section 503(b)(1) 
of the act.
    (ii) The sulfur dioxide and all other components of the preparation 
meet their professed standards of identity, strength, quality, and 
purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 5 grams of sulfur 
dioxide per 100 milliliters of solution.
    (v) The preparation is labeled with adequate directions for use by 
external application to the smooth skin in the prevention or treatment 
of minor conditions in which it is indicated.
    (vi) The directions for use recommend or suggest not more than two 
applications a day for not more than 1 week, except as directed by a 
physician.
    (13)-(15) [Reserved]
    (16) Tuaminoheptane sulfate (2-aminoheptane sulfate) preparations 
meeting all the following conditions:
    (i) The tuaminoheptane sulfate is prepared, with or without other 
drugs, in an aqueous vehicle suitable for administration in self-
medication as nose drops, and containing no drug limited to prescription 
sale under the provisions of section 503(b)(1) of the act.
    (ii) The preparation is packaged with a style of container or 
assembly suited to self-medication by the recommended route of 
administration, and delivering not more than 0.1 milliliter of the 
preparation per drop.
    (iii) The tuaminoheptane sulfate and all other components of the 
preparation meet their professed standards of identity, strength, 
quality, and purity.
    (iv) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (v) The tuaminoheptane sulfate content of the preparation does not 
exceed 10 milligrams per milliliter.
    (vi) The preparation is labeled with adequate directions for use in 
the temporary relief of nasal congestion.
    (vii) The dosages recommended or suggested in the directions for use 
do not exceed the equivalent: For adults, 5 drops of a 1 percent 
solution per nostril per dose, and 5 doses in a 24-hour period; for 
children 1 to 6 years of age, 3 drops of a 1 percent solution per 
nostril per dose, and 5 doses in a 24-hour period; for infants under 1 
year of age, 2 drops of a 1 percent solution per nostril per dose, and 5 
doses in a 24-hour period.
    (viii) The labeling bears, in juxtaposition with the dosage 
recommendations:
    (a) Clear warning statements against use of more than 5 doses daily, 
and against use longer than 4 days unless directed by a physician.
    (b) A clear warning statement to the effect that frequent use may 
cause nervousness or sleeplessness, and that individuals with high blood 
pressure, heart disease, diabetes, or thyroid disease should not use the 
preparation unless directed by a physician.
    (17) [Reserved]
    (18) Vibesate (a mixture of copolymers of hydroxy-vinyl 
chlorideacetate, sebacic acid, and modified maleic rosin ester) 
preparations meeting all the following conditions.
    (i) The vibesate is prepared and packaged, with or without other 
drugs, solvents, and propellants, in a form suitable for self-medication 
by external application to the skin as a spray, and containing no drug 
limited to prescription sale under the provisions of section 503(b)(1) 
of the act.
    (ii) The vibesate and all other components of the preparation meet 
their professed standards of identity, strength, quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 13 percent by weight of 
vibesate.
    (v) The preparation is labeled with adequate directions for use by 
external application as a dressing for minor burns, minor cuts, or other 
minor skin irritations.
    (vi) The labeling bears in juxtaposition with the directions for use 
clear warning statements against:
    (a) Use on serious burns and on infected, deep, and puncture wounds 
unless directed by a physician.

[[Page 17]]

    (b) Spraying the preparation near the eyes or other mucous 
membranes.
    (c) Inhaling the preparation.
    (d) Use near open flames.
    (e) Puncturing the container or throwing the container into fire.
    (19) Pramoxine hydrochloride (4-N-butoxyphenyl -
morpholinopropyl ether hydrochloride) preparations meeting all the 
following conditions:
    (i) The pramoxine hydrochloride is prepared, with or without other 
drugs, in a dosage form suitable for use in self-medication by external 
application to the skin, and containing no drug limited to prescription 
sale under the provisions of section 503(b)(1) of the act.
    (ii) The pramoxine hydrochloride and all other components of the 
preparation meet their professed standards of identity, strength, 
quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 1.0 percent of pramoxine 
hydrochloride.
    (v) The preparation is labeled with adequate directions for use by 
external application to the skin for the temporary relief of pain or 
itching due to minor burns and sunburn, nonpoisonous insect bites, and 
minor skin irritations.
    (vi) The directions for use recommend or suggest not more than four 
applications of the preparation per day, unless directed by a physician.
    (vii) The labeling bears, in juxtaposition with the directions for 
use, clear warning statements against:
    (a) Prolonged use.
    (b) Application to large areas of the body.
    (c) Continued use if redness, irritation, swelling, or pain persists 
or increases, unless directed by a physician.
    (d) Use in the eyes or nose.
    (20) Carbetapentane citrate (2-(2-diethylaminoethoxy)-ethyl-1-
phenyl- cyclopentyl-1-carboxylate citrate) preparations meeting all the 
following conditions:
    (i) The carbetanentane citrate is prepared, with or without other 
drugs, in tablet or other dosage form suitable for oral use in self-
medication, and containing no drug limited to prescription sale under 
the provisions of section 503(b)(1) of the act.
    (ii) The carbetapentane citrate and all other components of the 
preparation meet their professed standards of identity, strength, 
quality, and purity.
    (iii) If the preparation is a new drug, and application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 25 milligrams of 
carbetapentane citrate per dosage unit; or if it is in liquid form, not 
more than 1.5 milligrams of carbetapentane citrate per milliliter.
    (v) The preparation is labeled with adequate directions for use in 
the temporary relief of cough due to minor conditions in which it is 
indicated.
    (vi) The dosages recommended or suggested in the labeling do not 
exceed: For adults, 30 milligrams of carbetapentane citrate per dose or 
120 milligrams of carbetapentane citrate per 24-hour period; for 
children 4 to 12 years of age, 7.5 milligrams per dose or 30 milligrams 
per 24-hour period; for children 2 to 4 years of age, 4.0 milligrams per 
dose or 16.0 milligrams per 24-hour period.
    (vii) The label bears a conspicuous warning to keep the drug out of 
the reach of children, and the labeling bears, in juxtaposition with the 
dosage recommendations:
    (a) A clear warning statement against administration of the drug to 
children under 2 years of age, unless directed by a physician.
    (b) Clear warning statements against use of the drug in the presence 
of high fever or if cough persists, since persistent cough as well as 
high fever may indicate the presence of a serious condition.
    (21) Pamabrom (2-amino-2-methylpropanol-1-8-bromotheophyllinate) 
preparations meeting all the following conditions:
    (i) The pamabrom is prepared with appropriate amounts of a suitable 
analgesic and with or without other drugs, in tablet or other dosage 
form suitable for oral use in self-medication, and containing no drug 
limited to prescription sale under the provisions of section 503(b)(1) 
of the act.
    (ii) The pamabrom and all other components of the preparation meet 
their

[[Page 18]]

professed standards of identity, strength, quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 50 milligrams of 
pamabrom per dosage unit.
    (v) The preparation is labeled with adequate directions for use in 
the temporary relief of the minor pains and discomforts that may occur a 
few days before and during the menstrual period.
    (vi) The dosages recommended or suggested in the labeling do not 
exceed 50 milligrams of pamabrom per dose or 200 milligrams per 24-hour 
period.
    (22) Diphemanil methylsulfate (4-diphenylmethylene-1,1-dimethyl-
piperidinium methylsulfate) preparations meeting all the following 
conditions:
    (i) The diphemanil methylsulfate is prepared, with or without other 
drugs, in a dosage form suitable for use in self-medication by external 
application to the skin, and containing no drug limited to prescription 
sale under the provisions of section 503(b)(1) of the act.
    (ii) The diphemanil methylsulfate and all other components of the 
preparation meet their professed standards of identity, strength, 
quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 2.0 percent of 
diphemanil methylsulfate.
    (v) The preparation is labeled with adequate directions for use by 
external application to the skin for the relief of symptoms of mild 
poison ivy, oak, and sumac and other minor irritations and itching of 
the skin.
    (vi) The directions for use recommend or suggest not more than four 
applications of the preparation per day, unless directed by a physician.
    (vii) The labeling bears, in juxtaposition with the directions for 
use, a clear warning statement, such as: ``Caution: If redness, 
irritation, swelling, or pain persists or increases, discontinue use and 
consult physician.''
    (23) Dyclonine hydrochloride (4-butoxy-3-piperidinopropiophenone 
hydrochloride; 4-n-butoxy--piperidonopropiophenone 
hydrochloride) preparations meeting all the following conditions:
    (i) The dyclonine hydrochloride is prepared, with or without other 
drugs, in a dosage form suitable for use as a cream or ointment in self-
medication by external application to the skin, or rectally, and 
contains no drug limited to prescription sale under the provisions of 
section 503(b)(1) of the act.
    (ii) The dyclonine hydrochloride and all other components of the 
preparation meet their professed standards of identity, strength, 
quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 1.0 percent of dyclonine 
hydrochloride.
    (v) The preparation is labeled with adequate directions for use:
    (a) By external application to the skin for the temporary relief of 
pain and itching in sunburn, nonpoisonous insect bites, minor burns, 
cuts, abrasions, and other minor skin irritations.
    (b) [Reserved]
    (c) In the prevention or treatment of other minor conditions in 
which it is indicated.
    (vi) The labeling bears, in juxtaposition with the directions for 
use, clear warning statements against:
    (a) Continued use if redness, irritation, swelling, or pain persists 
or increases, unless directed by a physician.
    (b) Use in case of rectal bleeding, as this may indicate serious 
disease.
    (c) Use in the eyes.
    (d) Prolonged use.
    (e) Application to large areas of the body.
    (f) Use for deep or puncture wounds or serious burns.
    (24) Chlorothen citrate (chloromethapyrilene citrate; N,N-dimethyl-
N'-(2-pyridyl)-N '-(5-chloro-2-thenyl) ethylenediamine citrate) 
preparations meeting all the following conditions:
    (i) The chlorothen citrate is prepared, with or without other drugs, 
in tablet or other dosage form suitable for oral use in self-medication, 
and containing no drug limited to prescription sale under the provisions 
of section 503(b)(1) of the act.

[[Page 19]]

    (ii) The chlorothen citrate and all other components of the 
preparation meet their professed standards of identity, strength, 
quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 25 milligrams of 
chlorothen citrate per dosage unit.
    (v) The preparation is labeled with adequate directions for use in 
the temporary relief of the symptoms of hay fever and/or the symptoms of 
other minor conditions in which it is indicated.
    (vi) The dosages recommended or suggested in the labeling do not 
exceed: For adults, 25 milligrams of chlorothen citrate per dose or 150 
milligrams of chlorothen citrate per 24-hour period; for children 6 to 
12 years of age, one-half of the maximum adult dose or dosage.
    (vii) The labeling bears, in juxtaposition with the dosage 
recommendations:
    (a) Clear warning statements against administration of the drug to 
children under 6 years of age or exceeding the recommended dosage, 
unless directed by a physician, and against driving a car or operating 
machinery while using the drug, since it may cause drowsiness.
    (b) If the article is offered for the temporary relief of symptoms 
of colds, a statement that continued administration for such use should 
not exceed 3 days, unless directed by a physician.
    (25) [Reserved]
    (26) Methoxyphenamine hydrochloride (-(o-methoxyphenyl)-
isopropyl-methylamine hydrochloride; 1-(o-methoxyphenyl)-2-methylamino- 
propane hydrochloride) preparations meeting all the following 
conditions:
    (i) The methoxyphenamine hydrochloride is prepared with appropriate 
amounts of a suitable antitussive, with or without other drugs, in a 
dosage form suitable for oral use in self-medication, and containing no 
drug limited to prescription sale under the provisions of section 
503(b)(1) of the act.
    (ii) The methoxyphenamine hydrochloride and all other components of 
the preparation meet their professed standards of identity, strength, 
quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.
    (iv) The preparation contains not more than 3.5 milligrams of 
methoxyphenamine hydrochloride per milliliter.
    (v) The preparation is labeled with adequate directions for use in 
the temporary relief of cough due to minor conditions in which it is 
indicated.
    (vi) The dosages recommended or suggested in the labeling do not 
exceed: For adults, 35 milligrams of methoxyphenamine hydrochloride per 
dose or 140 milligrams of methoxyphenamine hydrochloride per 24-hour 
period; for children 6 to 12 years of age, one-half of the maximum adult 
dose or dosage.
    (vii) The label bears a conspicuous warning to keep the drug out of 
the reach of children, and the labeling bears, in juxtaposition with the 
dosage recommendations:
    (a) A clear warning statement against administration of the drug to 
children under 6 years of age, unless directed by a physician.
    (b) A clear warning statement to the effect that frequent or 
prolonged use may cause nervousness, restlessness, or drowsiness, and 
that individuals with high blood pressure, heart disease, diabetes, or 
thyroid disease should not use the preparation unless directed by a 
physician.
    (c) A clear warning statement against use of the drug in the 
presence of high fever or if cough persists, since persistent cough as 
well as high fever may indicate the presence of a serious condition.
    (27) Biphenamine hydrochloride (-diethylaminoethyl-3-
phenyl-2-hydroxy-benzoate hydrochloride) preparations meeting all the 
following conditions:
    (i) The biphenamine hydrochloride is prepared in a form suitable for 
use as a shampoo and contains no drug limited to prescription sale under 
the provisions of section 503(b)(1) of the act.
    (ii) The biphenamine hydrochloride meets its professed standards of 
identity, strength, quality, and purity.
    (iii) If the preparation is a new drug, an application pursuant to 
section 505(b) of the act is approved for it.

[[Page 20]]

    (iv) The preparation contains not more than 1 percent of biphenamine 
hydrochloride.
    (v) The preparation is labeled with adequate directions for use for 
the temporary relief of itching and scaling due to dandruff.
    (vi) The label bears a conspicuous warning to keep the drug out of 
the reach of children.
    (28) Tyloxapol (an alkylarylpolyether alcohol) and benzalkonium 
chloride ophthalmic preparations meeting all the following conditions:
    (i) The tyloxapol and benzalkonium chloride are prepared, with other 
appropriate ingredients which are not drugs limited to prescription sale 
under the provisions of section 503(b)(1) of the act, as a sterile, 
isotonic aqueous solution suitable for use in self-medication on eye 
prostheses.
    (ii) The preparation is so packaged as to volume and type of 
container as to afford adequate protection and be suitable for self-
medication with a minimum risk of contamination of the solution during 
use. Any dispensing unit is sterile and so packaged as to maintain 
sterility until the package is opened.
    (iii) The tyloxapol, benzalkonium chloride, and other ingredients 
used to prepare the isotonic aqueous solution meet their professed 
standards of identity, strength, quality, and purity.
    (iv) An application pursuant to section 505(b) of the act is 
approved for the drug.
    (v) The preparation contains 0.25 percent of tyloxapol and 0.02 
percent of benzalkonium chloride.
    (vi) The label bears a conspicuous warning to keep the drug out of 
the reach of children and the labeling bears, in juxtaposition with the 
dosage recommendations, a clear warning that if irritation occurs, 
persists, or increases, use of the drug should be discontinued and a 
physician consulted. The labeling includes a statement that the dropper 
or other dispensing tip should not touch any surface, since this may 
contaminate the solution.
    (29) [Reserved]
    (b) [Reserved]
[39 FR 11680, Mar. 29, 1974, as amended at 42 FR 36994, July 19, 1977; 
52 FR 15892, Apr. 30, 1987; 52 FR 30055, Aug. 12, 1987; 55 FR 31779, 
Aug. 3, 1990; 57 FR 58374, Dec. 9, 1992; 58 FR 49898, Sept. 23, 1993; 59 
FR 4218, Jan. 28, 1994; 60 FR 52507, Oct. 6, 1995]



                     Subpart D--Records and Reports



Sec. 310.303  Continuation of long-term studies, records, and reports on certain drugs for which new drug applications have been approved.

    (a) A new drug may not be approved for marketing unless it has been 
shown to be safe and effective for its intended use(s). After approval, 
the applicant is required to establish and maintain records and make 
reports related to clinical experience or other data or information 
necessary to make or facilitate a determination of whether there are or 
may be grounds under section 505(e) of the act for suspending or 
withdrawing approval of the application. Some drugs, because of the 
nature of the condition for which they are intended, must be used for 
long periods of time--even a lifetime. To acquire necessary data for 
determining the safety and effectiveness of long-term use of such drugs, 
extensive animal and clinical tests are required as a condition of 
approval. Nonetheless, the therapeutic or prophylactic usefulness of 
such drugs may make it inadvisable in the public interest to delay the 
availability of the drugs for widespread clinical use pending completion 
of such long-term studies. In such cases, the Food and Drug 
Administration may approve the new drug application on condition that 
the necessary long-term studies will be conducted and the results 
recorded and reported in an organized fashion. The procedures required 
by paragraph (b) of this section will be followed in order to list such 
a drug in Sec. 310.304.
    (b) A proposal to require additional or continued studies with a 
drug for which a new drug application has been approved may be made by 
the Commissioner on his own initiative or on the petition of any 
interested person, pursuant to part 10 of this chapter. Prior

[[Page 21]]

to issuance of such a proposal, the applicant will be provided an 
opportunity for a conference with representatives of the Food and Drug 
Administration. When appropriate, investigators or other individuals may 
be invited to participate in the conference. All requirements for 
special studies, records, and reports will be published in Sec. 310.304.
[39 FR 11680, Mar. 29, 1974, as amended at 41 FR 4714, Jan. 25, 1976; 42 
FR 15674, Mar. 22, 1977]



Sec. 310.305  Records and reports concerning adverse drug experiences on marketed prescription drugs for human use without approved new drug applications.

    (a) Scope. FDA is requiring manufacturers, packers, and distributors 
of marketed prescription drug products that are not the subject of an 
approved new drug or abbreviated new drug application to establish and 
maintain records and make reports to FDA of all serious, unexpected 
adversedrug experiences associated with the use of their drug products. 
Any person subject to the reporting requirements of paragraph (c) of 
this section shall also develop written procedures for the surveillance, 
receipt, evaluation, and reporting of postmarketing adverse drug 
experiences to FDA.
    (b) Definitions. The following definitions of terms apply to this 
section:-
    Adverse drug experience. Any adverse event associated with the use 
of a drug in humans, whether or not considered drug related, including 
the following: An adverse event occurring in the course of the use of a 
drug product in professional practice; an adverse event occurring from 
drug overdose whether accidental or intentional; an adverse event 
occurring from drug abuse; an adverse event occurring from drug 
withdrawal; and any failure of expected pharmacological action.
    Disability. A substantial disruption of a person's ability to 
conduct normal life functions.
    Life-threatening adverse drug experience. Any adverse drug 
experience that places the patient, in the view of the initial reporter, 
at immediate risk of death from the adverse drug experience as it 
occurred, i.e., it does not include an adverse drug experience that, had 
it occurred in a more severe form, might have caused death.
    Serious adverse drug experience. Any adverse drug experience 
occurring at any dose that results in any of the following outcomes: 
Death, a life-threatening adverse drug experience, inpatient 
hospitalization or prolongation of existing hospitalization, a 
persistent or significant disability/incapacity, or a congenital 
anomaly/birth defect. Important medical events that may not result in 
death, be life-threatening, or require hospitalization may be considered 
a serious adverse drug experience when, based upon appropriate medical 
judgment, they may jeopardize the patient or subject and may require 
medical or surgical intervention to prevent one of the outcomes listed 
in this definition. Examples of such medical events include allergic 
bronchospasm requiring intensive treatment in an emergency room or at 
home, blood dyscrasias or convulsions that do not result in inpatient 
hospitalization, or the development of drug dependency or drug abuse.
    Unexpected adverse drug experience. Any adverse drug experience that 
is not listed in the current labeling for the drug product. This 
includes events that may be symptomatically and pathophysiologically 
related to an event listed in the labeling, but differ from the event 
because of greater severity or specificity. For example, under this 
definition, hepatic necrosis would be unexpected (by virtue of greater 
severity) if the labeling only referred to elevated hepatic enzymes or 
hepatitis. Similarly, cerebral thromboembolism and cerebral vasculitis 
would be unexpected (by virtue of greater specificity) if the labeling 
only listed cerebral vascular accidents. ``Unexpected,'' as used in this 
definition, refers to an adverse drug experience that has not been 
previously observed (i.e., included in the labeling) rather than from 
the perspective of such experience not being anticipated from the 
pharmacological properties of the pharmaceutical product.
    (c) Reporting requirements. Each person identified in paragraph 
(c)(1)(i) of this section shall report to FDA adverse drug experience 
information as

[[Page 22]]

described in this section and shall submit one copy of each report to 
the Division of Pharmacovigilance and Epidemiology (HFD-730), Center for 
Drug Evaluation and Research, Food and Drug Administration, 5600 Fishers 
Lane, Rockville, MD 20857.
    (1) Postmarketing 15-day ``Alert reports''. (i) Any person whose 
name appears on the label of a marketed prescription drug product as its 
manufacturer, packer, or distributor shall report to FDA each adverse 
drug experience received or otherwise obtained that is both serious and 
unexpected as soon as possible, but in no case later than 15 calendar 
days of initial receipt of the information by the person whose name 
appears on the label. Each report shall be accompanied by a copy of the 
current labeling for the drug product.
    (ii) A person identified in paragraph (c)(1)(i) of this section is 
not required to submit a 15-day ``Alert report'' for an adverse drug 
experience obtained from a postmarketing study (whether or not conducted 
under an investigational new drug application) unless the applicant 
concludes that there is a reasonable possibility that the drug caused 
the adverse experience.
    (2) Postmarketing 15-day ``Alert reports''--followup. Each person 
identified in paragraph (c)(1)(i) of this section shall promptly 
investigate all serious, unexpected adverse drug experiences that are 
the subject of these postmarketing 15-day Alert reports and shall submit 
followup reports within 15 calendar days of receipt of new information 
or as requested by FDA. If additional information is not obtainable, 
records should be maintained of the unsuccessful steps taken to seek 
additional information. Postmarketing 15-day Alert reports and followups 
to them shall be submitted under separate cover.
    (3) Submission of reports. To avoid unnecessary duplication in the 
submission of, and followup to, reports required in this section, a 
packer's or distributor's obligations may be met by submission of all 
reports of serious adverse drug experiences to the manufacturer of the 
drug product. If a packer or distributor elects to submit these adverse 
drug experience reports to the manufacturer rather than to FDA, it shall 
submit each report to the manufacturer within 5 calendar days of its 
receipt by the packer or distributor, and the manufacturer shall then 
comply with the requirements of this section even if its name does not 
appear on the label of the drug product. Under this circumstance, the 
packer or distributor shall maintain a record of this action which shall 
include:
    (i) A copy of each adverse drug experience report;
    (ii) The date the report was received by the packer or distributor;
    (iii) The date the report was submitted to the manufacturer; and
    (iv) The name and address of the manufacturer.
    (4) Each report submitted to FDA under this section shall bear 
prominent identification as to its contents, i.e., ``15-day Alert 
report,'' or ``15-day Alert report-followup.''
    (5) A person identified in paragraph (c)(1)(i) of this section is 
not required to resubmit to FDA adverse drug experience reports 
forwarded to that person by FDA; however, the person must submit all 
followup information on such reports to FDA.
    (d) Reporting form. (1) Except as provided in paragraph (d)(3) of 
this section, each person identified in paragraph (c)(1)(i) of this 
section shall submit each report of a serious and unexpected adverse 
drug experience on an FDA Form 3500A (foreign events may be submitted 
either on an FDA Form 3500A or, if preferred, on a CIOMS I form).
    (2) Each completed FDA Form 3500A should pertain only to an 
individual patient.
    (3) Instead of using Form FDA Form 3500A, a manufacturer, packer, or 
distributor may use a computer-generated FDA Form 3500A or other 
alternative format (e.g., a computer-generated tape or tabular listing) 
provided that:
    (i) The content of the alternative format is equivalent in all 
elements of information to those specified in FDA Form 3500A, and
    (ii) The format is agreed to in advance by MedWatch: The FDA Medical 
Products Reporting Program.
    (4) Ten copies or fewer of FDA Form 3500A and/or a copy of the 
instructions

[[Page 23]]

for completing the form may be obtained from the Division of 
Pharmacovigilance and Epidemiology (HFD-730), Center for Drug Evaluation 
and Research, Food and Drug Administration, 5600 Fishers Lane, 
Rockville, MD 20857. More than 10 copies of the form may be obtained by 
writing to the Consolidated Forms and Publications Distribution Center, 
Washington Commerce Center, 3222 Hubbard Rd., Landover, MD 20785.
    (e) Patient privacy. Manufacturers, packers, and distributors should 
not include in reports under this section the names and addresses of 
individual patients; instead, the manufacturer, packer, and distributor 
should assign a unique code number to each report, preferably not more 
than eight characters in length. The manufacturer, packer, and 
distributor should include the name of the reporter from whom the 
information was received. Names of patients, individual reporters, 
health care professionals, hospitals, and geographical identifiers in 
adverse drug experience reports are not releasable to the public under 
FDA's public information regulations in part 20 of this chapter.
    (f) Recordkeeping. (1) Each manufacturer, packer, and distributor 
shall maintain for a period of 10 years records of all adverse drug 
experiences required under this section to be reported, including raw 
data and any correspondence relating to the adverse drug experiences, 
and the records required to be maintained under paragraph (c)(4) of this 
section.
    (2) Manufacturers and packers may retain the records required in 
paragraph (f)(1) of this section as part of its complaint files 
maintained under Sec. 211.198 of this chapter.
    (3) Manufacturers, packers, and distributors shall permit any 
authorized FDA employee, at all reasonable times, to have access to and 
copy and verify the records established and maintained under this 
section.
    (g) Disclaimer. A report or information submitted by a manufacturer, 
packer, or distributor under this section (and any release by FDA of 
that report or information) does not necessarily reflect a conclusion by 
the manufacturer, packer, or distributor, or by FDA, that the report or 
information constitutes an admission that the drug caused or contributed 
to an adverse effect. The manufacturer, packer, or distributor need not 
admit, and may deny, that the report or information submitted under this 
section constitutes an admission that the drug caused or contributed to 
an adverse effect.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0210)

[51 FR 24779, July 3, 1986, as amended at 52 FR 37936, Oct. 13, 1987; 55 
FR 11578, Mar. 29, 1990; 57 FR 17980, Apr. 28, 1992; 62 FR 34167, June 
25, 1997; 62 FR 52249, Oct. 7, 1997]

    Effective Date Note: At 62 FR 52249, Oct. 7, 1997, Sec. 310.305 was 
amended by adding a new sentence at the end of paragraph (a); by 
revising paragraphs (b), (c), (d)(1), (d)(3)(ii), and (d)(4); by 
removing in paragraph (d)(2), the introductory text of paragraph (d)(3), 
and paragraph (d)(3)(i) the words ``Form FDA-1639'' or ``FDA-1639'' and 
adding in their place ``FDA Form 3500A'', effective Apr. 6, 1998. For 
the convenience of the user, the superseded text is set forth as 
follows:

Sec. 310.305  Records and reports concerning adverse drug experiences on 
          marketed prescription drugs for human use without approved new 
          drug applications.

                                * * * * *

    (b) Definitions. The following definitions of terms apply to this 
section:
    (1) FDA means the Food and Drug Administration.
    (2) Adverse drug experience means any adverse event associated with 
the use of a drug in humans, whether or not considered drug related, 
including the following: an adverse event occurring in the course of the 
use of a drug product in professional practice; an adverse event 
occurring from drug overdose, whether accidental or intentional; an 
adverse event occurring from drug abuse; an adverse event occurring from 
drug withdrawal; and any failure of expected pharmacological action.
    (3) Unexpected means an adverse drug experience that is not listed 
in the current labeling for the drug product and includes an event that 
may be symptomatically and pathophysiologically related to an event 
listed in the labeling, but differs from the event because of greater 
severity or specificity. For example, under this definition, hepatic 
necrosis would be unexpected (by virtue of greater severity) if the 
labeling only referred to elevated hepatic enzymes or hepatitis. 
Similarly, cerebral thromboembolism

[[Page 24]]

and cerebral vasculitis would be unexpected (by virtue of greater 
specificity) if the labeling only listed cerebral vascular accidents.
    (4) Serious means an adverse drug experience that is fatal or life-
threatening, is permanently disabling, requires inpatient 
hospitalization, or is a congenital anomaly, cancer, or overdose.
    (c) Reporting requirements--15-day ``Alert reports.'' (1)(i) Any 
person whose name appears on the label of a marketed prescription drug 
product as its manufacturer, packer, or distributor shall report to FDA 
each adverse drug experience received or otherwise obtained that is both 
serious and unexpected as soon as possible but in any case within 15 
working days of initial receipt of the information. Each report shall be 
accompanied by a copy of the current labeling for the drug product.
    (ii) A person identified in paragraph (c)(1)(i) of this section is 
not required to submit a 15-day ``Alert report'' for an adverse drug 
experience obtained from a postmarketing study (whether or not conducted 
under an investigational new drug application) unless the applicant 
concludes that there is a reasonable possibility that the drug caused 
the adverse experience.
    (2) Each person identified in paragraph (c)(1) of this section shall 
submit one copy of each report to the Division of Epidemiology and 
Surveillance (HFD-730), Center for Drug Evaluation and Research, Food 
and Drug Administration, 5600 Fishers Lane, Rockville, MD 20857.
    (3) Each person identified in paragraph (c)(1) of this section shall 
promptly investigate all serious, unexpected adverse drug experiences 
that are the subject of these 15-day Alert reports and shall submit 
followup reports within 15 working days of receipt of new information or 
as requested by FDA. If additional information is not obtainable, a 
followup report may be required that describes briefly the steps taken 
to seek additional information and the reasons why it could not be 
obtained.
    (4) To avoid unnecessary duplication in the submission of, and 
followup to, reports required in this section, a packer's or 
distributor's obligations may be met by submission of all reports of 
serious adverse drug experiences to the manufacturer of the drug 
product. If a packer or distributor elects to submit these adverse drug 
experience reports to the manufacturer rather than to FDA, it shall 
submit each report to the manufacturer within 3 working days of its 
receipt by the packer or distributor, and the manufacturer shall then 
comply with the requirements of this section even if its name does not 
appear on the label of the drug product. Under this circumstance, the 
packer or distributor shall maintain a record of this action which shall 
include:
    (i) A copy of each drug experience report.
    (ii) Date the report was received by the packer or distributor.
    (iii) Date the report was submitted to the manufacturer.
    (iv) Name and address of the manufacturer.
    (5) Each report submitted to FDA under this section shall bear 
prominent identification as to its contents, i.e., ``15-day Alert 
report'' or ``15-day Alert report--followup.''
    (d) Reporting form. (1) Except as provided in paragraph (d)(3) of 
this section, each person identified in paragraph (c)(1) of this section 
shall submit each report of a serious and unexpected adverse drug 
experience on a Form FDA-1639 (Adverse Reaction Report).

                                * * * * *

    (3)* * *
    (i)* * *
    (ii) The format is agreed to in advance by the Division of 
Epidemiology and Surveillance (HFD-730).
    (4) Single copies of Form FDA-1639 may be obtained from the Division 
of Epidemiology and Surveillance (HFD-730), Center for Drug Evaluation 
and Research, Food and Drug Administration, 5600 Fishers Lane, 
Rockville, MD 20857. Supplies of Form FDA-1639 may be obtained from the 
PHS Forms and Publications Distribution Center, 12100 Parklawn Dr., 
Rockville, MD 20857.

                                * * * * *



        Subpart E--Requirements for Specific New Drugs or Devices



Sec. 310.500  Digoxin products for oral use; conditions for marketing.

    (a) Studies have shown evidence of clinically significant 
differences in bio-availability in different batches of certain marketed 
digoxin products for oral use from single manufacturers as well as in 
batches of these products produced by different manufacturers. These 
differences were observed despite the fact that the products met 
compendial specifications. Other studies have shown that there is a 
sufficient correlation between bioavailability in vivo and the 
dissolution rate of digoxin tablets in vitro to make the dissolution 
test an important addition to the compendial standards. Because of the 
potential for serious risk to cardiac patients using digoxin products 
which may vary in bioavailability, the Commissioner of Food and Drugs 
has determined that immediate action must be taken to assure the 
uniformity

[[Page 25]]

of all digoxin products for oral use. The Commissioner is of the opinion 
that digoxin products for oral use are new drugs within the meaning of 
section 201(p) of the Federal Food, Drug, and Cosmetic Act for which 
approved new drug applications are required. The Commissioner has 
determined that, because of questions raised regarding the 
bioavailability of digoxin products for oral use, there is sufficient 
evidence to invoke the authority under section 505(j) of the act to 
fully investigate this question and to facilitate a determination of 
whether there is a ground for withdrawal of approval of the drug product 
under section 505(e) of the act. Marketing of these products may be 
continued only under the following conditions:
    (1) Digoxin products for oral use, other than tablets: Any person 
marketing digoxin products for oral use, other than tablets, shall 
submit to the Food and Drug Administration on or before February 21, 
1974, an abbreviated new drug application for these products. Any such 
drug product then on the market which is not the subject of an 
application submitted for the drug product shall be subject to 
regulatory procedures under section 505 of the act. In addition to the 
information specified in Sec. 314.50 of this chapter, the application 
shall contain:
    (i) A full list of the articles used as components of the digoxin 
product, specifications for components, detailed identification and 
analytical procedures used to assure that the components meet 
established specifications of identity, strength, quality, and purity 
and a complete description of the manufacturing process.
    (ii) The source of the digoxin used in the formulation including the 
name and address of the supplier.
    (iii) A statement that stability studies will be conducted to 
establish a suitable expiration date for the digoxin product in the form 
in which it is distributed.
    (iv) A statement that the product label will contain a suitable 
expiration date. In the absence of any stability test data, this 
expiration date shall be no longer than one year after the batch is 
manufactured. If the expiration date is greater than one year, 
supporting stability data shall be included in the application.
    (v) Labeling that is in compliance with all requirements of the act 
and regulations promulgated thereunder, the pertinent parts of which are 
as indicated in paragraph (e) of this section.
    (vi) A statement that the applicant will initiate recall of all 
stocks of the drug product outstanding when so requested by the Food and 
Drug Administration.
    (vii) A statement that the applicant intends to conduct in vivo 
bioavailability tests and that the applicant, under the records and 
reports provisions of section 505(k) of the act, will:
    (a) Within 30 days after the submission of the application, submit 
to the Food and Drug Administration the protocol which the applicant 
proposes to follow in conducting these in vivo bioavailability tests. 
The protocol shall contain all of the essential elements set forth in 
paragraph (d) of this section. The tests shall not be initiated prior to 
receiving notification from the Food and Drug Administration that the 
bioavailability protocol has been reviewed and either approved or its 
deficiencies delineated.
    (b) Within 180 days after receiving notification from the Food and 
Drug Administration that the bioavailability protocol has been reviewed, 
submit to the Food and Drug Administration the results of the in vivo 
bioavailability tests.
    (2) Digoxin tablets: Any person marketing digoxin tablets, in 
addition to complying with all of the requirements of paragraph (a)(1) 
of this section, shall include in their abbreviated new drug 
application:
    (i) A statement that the applicant will establish procedures to test 
each lot of digoxin tablets prior to releasing the batch for 
distribution to assure that the batch meets all of The United States 
Pharmacopeia (USP XVIII) requirements for digoxin tablets including, but 
not limited to, potency, content uniformity, and dissolution and either 
(a) that the quantity of digoxin dissolved at one hour is not more than 
95 percent of the assayed amount of digoxin or (b) that the quantity of 
digoxin dissolved at 15 minutes is not

[[Page 26]]

more than 90 percent of the assayed amount of digoxin.
    (ii) A statement that finished product specifications shall be 
established to include provisions to assure that the range of average 
one-hour dissolution values among batches of digoxin tablets does not 
exceed 20 percent.
    (3) Before releasing for distribution any batch of digoxin tablets 
manufactured after January 22, 1974, the manufacturer shall:
    (i) Test a sample of the batch to assure that the batch meets all of 
the requirements of The United States Pharmacopeia (USP XVIII) including 
but not limited to, potency, content uniformity, and dissolution and 
either (a) that the quantity of digoxin dissolved at one hour is not 
more than 95 percent of the assayed amount of digoxin or (b) that the 
quantity of digoxin dissolved at 15 minutes is not more than 90 percent 
of the assayed amount of digoxin.
    (ii) Submit a sample of the batch to the Food and Drug 
Administration according to the procedures set forth in paragraph (g) of 
this section. Results of tests conducted on the batch by or for the 
manufacturer and the batch production record shall accompany the sample.
    (iii) Withhold the batch from distribution until he is notified by 
the Food and Drug Administration that the sample was tested and found to 
meet all of the requirements in The United States Pharmacopeia (USP 
XVIII) for potency, content uniformity, and dissolution and either (a) 
that the quantity of digoxin dissolved at one hour is not more than 95 
percent of the assayed amount of digoxin or (b) that the quantity of 
digoxin dissolved at 15 minutes is not more than 90 percent of the 
assayed amount of digoxin.
    (iv) Submit a sample of each batch of digoxin tablets as provided 
for in paragraph (a)(3)(ii) of this section until he is notified by the 
Food and Drug Administration that he is released from the certification 
program. This notification will be made on the basis of sample test 
results, inspectional findings regarding compliance with current good 
manufacturing practice, and compliance with all other requirements of 
this section and any other directives issued by the Food and Drug 
Administration as a condition for release from the certification 
program.
    (4) Any manufacturer who has distributed any batch of digoxin 
tablets which does not meet the compendial requirement for dissolution, 
when tested by the method in The United States Pharmacopeia (USP XVIII), 
shall initiate recall of the subject batch when so requested by the Food 
and Drug Administration.
    (b) Failure of an applicant to submit the protocol and/or the 
results of the in vivo bioavailability tests showing adequate evidence 
of the product's bioavailability within the times specified in paragraph 
(a)(1)(vii) of this section and/or to comply with all of the 
certification requirements of paragraph (a)(3) of this section shall be 
justification for withdrawal of approval of the application under 
section 505(e) of the act.
    (c) Any product reformulation or change in manufacturing process 
will require the submission of a supplement to the approved abbreviated 
new drug application containing adequate data to demonstrate the 
bioavailability of the reformulated product. Food and Drug 
Administration approval of the supplement is required before the 
reformulated product is marketed. The Food and Drug Administration 
recommends that, where digoxin tablets are reformulated, manufacturers 
reformulate their product to achieve dissolution of 70 to 90 percent at 
one hour when tested by all three methods (i.e., the USP method, and the 
``paddle-water'' and ``paddle-acid'' methods) described in paragraph (h) 
of this section.
    (d) The protocol for the in vivo bioavailability tests required in 
paragraphs (a) and (c) of this section shall employ a three-way 
crossover design using the digoxin test product; a reference digoxin 
tablet supplied, on request, by the Food and Drug Administration; and 
bulk digoxin USP in an oral solution. Appropriate venous blood and 
urinary samples are to be collected and analyzed. The method shall be 
capable of detecting the difference between the reference tablet and the 
reference oral solution. Bioavailability of the test product shall be 
demonstrated if a mean absorption of at least 75 percent of the combined 
mean of the two

[[Page 27]]

reference standards is observed. Assistance in developing a protocol for 
a particular dosage formulation may be obtained by contacting the Food 
and Drug Administration, Center for Drug Evaluation and Research (HFD-
420), 5600 Fishers Lane, Rockville, MD 20857.
    (e) Parts of the digoxin product labeling indicated below shall be 
as follows:

                       Digoxin Labeling Guidelines

                          (adult and pediatric)

                               description

    Digoxin is one of the cardiac (or digitalis) glycosides, a closely 
related group of drugs having in common specific and powerful effects on 
the myocardium. These drugs are found in a number of plants. The term 
``digitalis'' is used to designate the whole group. Typically, the 
glycosides are composed of three portions: a steroid nucleus, a lactone 
ring, and a sugar (hence ``glycosides'').
    (This section should include a chemical and physical description of 
digoxin and the same quantitative ingredient information as that 
required on the label.)

                                 Action

    The digitalis glycosides have qualitatively the same therapeutic 
effects on the heart. They (1) increase the force of myocardial 
contraction, (2) increase the refractory period of the atrioventricular 
(A-V) node, and (3) to a lesser degree, affect the sinoatrial (S-A) node 
and conduction system via the parasympathetic and sympathetic nervous 
systems.
    Gastrointestinal absorption of digoxin is a passive process. About 
50-75 percent of digoxin in tablet form is absorbed. Digoxin is only 20-
25 percent bound to plasma proteins and is predominantly excreted by the 
kidneys unmetabolized unless there is significant renal failure. Renal 
excretion of digoxin is proportional to glomerular filtration rate and 
is largely independent of urine flow. Digoxin is not effectively removed 
from the body by dialysis, exchange transfusion, or during 
cardiopulmonary bypass, presumably because of tissue binding. In 
subjects with normal renal function, digoxin is excreted exponentially 
with an average half-life of 36 hours, resulting in the loss of 35-40 
percent of the body stores daily.
    Serum levels and pharmacokinetics are essentially unchanged by 
massive weight loss, suggesting that lean body mass should be used in 
dosage calculations. The peak blood level from oral dosing with tablets 
occurs 1-3 hours after administration. The onset of therapeutic action 
of digoxin after oral tablets is 1-2 hours, with the peak therapeutic 
effect occurring 6-8 hours after dosing.

                               indications

    1. Congestive heart failure, all degrees, is the primary indication. 
The increased cardiac output due to digoxin results in diuresis and 
general amelioration of the disturbances characteristic of right (venous 
congestion, edema) and left (dyspnea, orthopnea, cardiac asthma) heart 
failure.
    Digoxin, generally, is most effective in ``low output'' failure and 
less effective in ``high output'' (bronchopulmonary insufficiency, 
infection, hyperthyroidism) heart failure.
    Digoxin should be continued after heart failure is abolished unless 
some known precipitating factor is corrected.
    2. Atrial fibrillation, especially when the ventricular rate is 
elevated. Digoxin rapidly reduces ventricular rates and eliminates the 
pulse deficit. Palpitation, precordial distress or weakness are relieved 
and any concomitant congestive failure ameliorated.
    Digoxin should be continued in doses necessary to maintain the 
desired ventricular rate and other clinical effects.
    3. Atrial flutter. Digoxin slows the heart and regular sinus rhythm 
may appear. Frequently the flutter is converted to atrial fibrillation 
with a slow ventricular rate. Stopping digoxin at this point may be 
followed by restoration of sinus rhythm, especially if the flutter was 
of the paroxysmal type. It is preferable, however, to continue digoxin 
if failure ensues or if atrial flutter is a frequent occurrence.
    4. Paroxysmal atrial tachycardia. Oral digoxin may be used, 
especially if the condition is resistant to lesser measures. Depending 
on the urgency, a more rapid acting parenteral preparation may be 
preferable to initiate digitalization, although if heart failure has 
ensued or paroxysms recur frequently, digoxin should be maintained by 
oral administration.
    Digoxin is not indicated in sinus tachycardia unless due to heart 
failure.
    5. Cardiogenic shock. The drug is often employed, especially when 
the condition is accompanied by pulmonary edema. Digoxin seems to affect 
adversely shock due to septicemia from gram negative bacteria.

                            contraindications

    The presence of toxic effects (See ADVERSE REACTIONS section) 
induced by any digitalis preparation is a contraindication to all of the 
gylcosides.
    Allergy, though rare, does occur. It may not extend to all 
preparations, and another may be tried.
    Ventricular fibrillation.

                                Warnings

Digitalis alone or with other drugs has been promoted for use in the 
treatment of obesity. This use of digoxin or other

[[Page 28]]

digitalis glycosides is unwarranted. Moreover, since they may cause 
potentially fatal arrhythmias or other adverse effects, the use of these 
drugs in the treatment of obesity is dangerous.

    Many of the arrhythmias for which digoxin is advised closely 
resemble those reflecting digoxin intoxication. If the possibility of 
digoxin intoxication cannot be excluded, cardiac glycosides should be 
temporarily withheld if permitted by the clinical situation.
    The patient with congestive heart failure may complain of nausea and 
vomiting. These symptoms may also be indications on digoxin 
intoxication. A clinical determination of the cause of these symptoms 
must be attempted before further drug administration.
    Patients with renal insufficiency require smaller than usual doses 
of digoxin. See ACTION section for mechanism.

                               precautions

    Atrial arrhythmias associated with hypermetabolic states are 
particularly resistant to digoxin treatment. Care must be taken to avoid 
digoxin toxicity if digoxin is used to help the arrhythmia.
    Digoxin is not indicated for the treatment of ventricular 
tachycardia unless congestive heart failure supervenes after a 
protracted episode not itself due to digoxin.
    Potassium depletion sensitizes the myocardium to digoxin, and 
toxicity may develop even with the usual dosage. Hypokalemia may also 
alter the rate of onset and intensity of the positive inotropic effect 
of digoxin. Therefore, it is desirable to maintain normal serum 
potassium levels in patients being treated with digoxin.
    Potassium wastage may result from diuretic or corticosteriod 
therapy, hemodialysis, and from suction of gastrointestinal secretions. 
It may accompany malnutrition, diarrhea, prolonged vomiting, old age, 
and long-standing congestive heart failure. In general, rapid changes in 
serum potassium or other electrolytes are to be avoided, and intravenous 
treatment with potassium should be reserved only for special 
circumstances as described below (see TREATMENT OF ARRHYTHMIAS PRODUCED 
BY OVERDOSAGES section).
    Patients with acute myocardial infarction, severe pulmonary disease, 
or far advanced heart failure may be more sensitive to digoxin and more 
prone to disturbances of rhythm.
    Calcium affects contractility and excitability of the heart in a 
manner similar to that of digoxin. Calcium may produce serious 
arrhythmias in digitalized patients.
    In myxedema the digoxin requirements are less because excretion rate 
is decreased and blood levels are significantly higher.
    In incomplete A-V block, especially in patients subject to Stokes-
Adams attacks, advanced or complete heart block may develop if digoxin 
is given. Heart failure in these patients can usually be controlled by 
other measures and by increasing the heart rate.
    Patients with chronic constructive pericarditis may respond 
unfavorably to digoxin.
    Patients with idiopathic hypertrophic subaortic stenosis must be 
managed extremely carefully. Unless cardiac failure is severe, it is 
doubtful whether digoxin should be employed.
    Renal insufficiency delays the excretion of digoxin, and dosage must 
be adjusted accordingly in patients with renal disease. Note: This 
applies also to potassium administration should it become necessary.
    Electrical conversion of arrhythmias may require reduction of 
digoxin dosage.

                            adverse reactions

    Gynecomastia, uncommon.
    Overdosage or toxic effects.
    Gastrointestinal: Anorexia, nausea, vomiting, diarrhea are the most 
common early symptoms of overdosages in the adult (but rarely 
conspicuous in infants). Uncontrolled heart failure may also produce 
such symptoms.
    Central nervous system: Visual disturbances (blurred vision, yellow 
vision), headache, weakness, apathy.
    Cardiac disturbances (arrhythmias): Ventricular premature beats are 
the most common, except in infants and young children. Paroxysmal and 
nonparoxysmal nodal rhythms, atrioventricular (interference) 
disassociation and paroxysmal atrial tachycardia (PAT) with block are 
also common arrhythmias due to digoxin overdosage. Conduction 
disturbances: Excessive slowing of the pulse is a clinical sign of 
digoxin overdosage. Atrioventricular block of increasing degree may 
proceed to complete heart block. Note: The electrocardiogram is 
fundamental in determining the presence and nature of these cardiac 
toxic disturbances. Digoxin may also induce other changes (as of the ST 
segment), but these provide no measure of the degree of digitalization.

            treatment of arrhythmias produced by overdosages

    Digoxin should be discontinued until all signs of toxicity are 
abolished. Discontinuation may be all that is necessary if toxic 
manifestations are not severe and appear after the time for peak effect 
of the drug.
    Potassium salts are commonly used. Potassium chloride in divided 
oral doses totaling 4-6 grams for adults (see PEDIATRIC INFORMATION 
section for pediatric dosage) may be given provided renal function is 
adequate.
    When correction of the arrhythmia is urgent and the serum potassium 
level is low or normal, potassium should be administered

[[Page 29]]

intravenously in a solution of 5 percent dextrose in water. A total of 
40-100 milliequivalents (30 milliequivalents per 500 milliliters) is 
given at the rate of 20 milliequivalents per hour unless limited by pain 
due to local irritation.
    Additional amounts may be given if the arrhythmia is uncontrolled 
and the potassium well tolerated.
    Continuous electrocardiographic monitoring should be performed to 
watch for any evidence of potassium toxicity, e.g., peaking of T waves, 
and to observe the effect on the arrhythmia so that the infusion may be 
promptly stopped when the desired effect is achieved.
    Caution: Potassium should not be used and may be dangerous for 
severe or complete heart block due to digoxin and not related to any 
tachycardia.
    Other agents that have been approved for the treatment of digoxin 
intoxication include procainamide, lidocaine, and propranolol.

                        dosage and administration

    Oral digoxin is administered slowly or rapidly as required until the 
desired therapeutic effect is obtained without symptoms of overdosage. 
The amount can be predicted approximately from the lean body mass of the 
patient with allowances made for excretion during the time taken to 
induce digitalization.
    Subsequent maintenance dosage is also determined tentatively by the 
amount necessary to sustain the desired therapeutic effect.
    Recommended dosages are practical average figures that may require 
considerable modification as dictated by individual sensitivity or 
associated conditions. Diminished renal function is the most important 
factor requiring modification of recommended or average doses. (See 
WARNINGS and PRECAUTIONS sections.)
    The average amount of digoxin that patients must accumulate to be 
digitalized with digoxin tablets is 1.0-1.5 milligrams. Digitalization 
may be accomplished by any of several approaches that vary in dosage and 
frequency of administration, but reach the same endpoint in terms of 
total amount accumulated.
    In previously undigitalized patients, a single loading dose of 0.5-
0.75 milligram orally usually produces a detectable effect in 1-2 hours 
that becomes maximal in 6-8 hours. Additional doses of 0.25-0.5 
milligram may be given cautiously at 6-8 hour intervals to full 
digitalization.
    In previously undigitalized patients, institution of daily 
maintenance therapy (0.125-0.5 milligram, see next paragraph) without a 
loading dose results in development of a steady-state plateau 
concentrations in about 7 days in patients with normal renal function.
    The average daily oral maintenance dose is 0.125-0.5 milligram, 
usually 0.25 milligram. In the elderly patient, 0.125-0.25 milligram 
should be considered the average maintenance dose.
    In patients with renal impairment, digoxin excretion is impaired and 
serum half-life is prolonged (see ACTION section). Digitalizing and 
maintenance doses are lower than those recommended for patients with 
normal renal functions. Signs of digoxin toxicity develop sooner in 
patients with renal impairment, and it takes longer for toxic signs and 
symptoms to disappear. Because of the prolonged half-life, a longer 
period of time is required to achieve an initial or new steady-state 
plateau in patients with renal impairment than in patients with normal 
renal function.
    It cannot be overemphasized that the values given are averages and 
substantial individual variation can be expected.
    (If pediatric dosage is available, the labeling sections above 
should be expanded to include the following information.)

                          pediatric information

                                warnings

    Newborn infants display considerable variability in their tolerance 
to digoxin, depending on their degree of maturity.
    Premature and immature infants are particularly sensitive, and 
dosage must be reduced and digitalization should be even more 
individualized and cautiously approached than in more mature infants. 
Impaired renal function must also be carefully taken into consideration.
    Congestive heart failure accompanying acute glomerulonephritis 
requires extreme care in digitalization. A relatively low total dose 
administered in divided doses and concomitant use of antihypertensive 
drugs has been recommended. ECG monitoring is essential. Digoxin should 
be discontinued as soon as possible.
    Patients with idiopathic hypertrophic subaortic stenosis must be 
managed extremely carefully. Unless cardiac failure is severe, it is 
doubtful whether digoxin should be employed.
    Patients with rheumatic carditis, especially when severe, are 
unusually sensitive to digoxin and prone to disturbances of rhythm. If 
heart failure develops, digitalization may be initiated with relatively 
low doses; then it can be cautiously increased until a beneficial effect 
is obtained. If a therapeutic trial does not result in improvement, the 
drug should be considered ineffective and be discontinued.
    Note: Digitalis glycosides are an important cause of accidental 
poisoning in children.

[[Page 30]]

                               precautions

    Dosage must be carefully titrated and differences in the 
bioavailability of parenteral preparations, elixirs, and tablets should 
be taken into account when switching patients from one preparation to 
another.
    Electrocardiographic monitoring may be necessary to avoid 
intoxication.
    Premonitory signs of toxicity in the newborn are undue slowing of 
the sinus rate, sinoatrial arrest, and prolongation of PR interval.

                            adverse reactions

    Toxic signs differ from the adult in a number of respects. Cardiac 
arrhythmias are the more reliable and frequent signs of toxicity.
    Vomiting and diarrhea, neurologic and visual disturbances are rare 
as initial signs.
    Premature ventricular systoles are rarely seen; nodal and atrial 
systoles are more frequent.
    Atrial arrhythmias, atrial ectopic rhythms, and paroxysmal atrial 
tachycardia with A-V block particularly are more common manifestations 
of toxicity in children. Ventricular arrhythmias are rare.

            treatment of arrhythmias produced by overdosages

    (See adult section for other recommendations for the treatment of 
arrhythmias produced by overdosages and for additional recommendations 
and cautions regarding the use of potassium.) Potassium preparations may 
be given orally in divided doses totaling 1-1.5 milliequivalents/
kilogram (1 gram K contains 13.4 milliequivalents). When correction of 
the arrhythmia is urgent, approximately 0.5 milliequivalents/kilogram of 
potassium per hour may be given, with careful electrocardiographic 
monitoring, as a solution of 20 milliequivalents or less per 500 
milliliters in 5 percent dextrose in water. The total dose should 
generally not exceed 2 milliequivalents of potassium/kilogram.

                        dosage and administration

    Digitalization must be individualized. Generally, premature and 
immature infants are particularly sensitive, requiring reduced dosage 
that must be determined by careful titration.
    Oral Dosage. Beyond the immediate newborn period, children require 
proportionally greater doses than adults on the basis of body weight or 
surface area. The recommended oral digitalizing dosages in children with 
normal renal function are:
    Newborn infants (normal), up to 1 month, require 40-60 micrograms/
kilogram.
    Infants, 1 month to 2 years, require approximately 60-80 micrograms/
kilogram.
    Children 2 years to 10 years, require 40-60 micrograms/kilogram.
    Children, over 10 years of age, require adult dosages in proportion 
to their body weight.
    Maintenance therapy is 20-30 percent of the digitalizing dose 
administered each day.
    Long term use of digoxin is indicated in almost all infants who have 
been digitalized for acute congestive heart failure unless the cause is 
transient. Many favor maintaining digoxin until at least 2 years of age 
in all infants with paroxysmal atrial tachycardia or in those who show 
either definite or latent failure.
    Many children with severe inoperable congenital defects need digoxin 
throughout childhood and often for life.

    (f) Abbreviated new drug applications shall be submitted to the Food 
and Drug Administration, Center for Drug Evaluation and Research, Office 
of Generic Drugs, 5600 Fishers Lane, Rockville, MD 20857.
    (g) All samples of digoxin tablets required by paragraph (a)(3) of 
this section to be submitted to the Food and Drug Administration shall 
be handled as follows:
    (1) The sample shall consist of 6 subsamples of 1000 tablets each 
collected at random from throughout the manufacturing run. Each of the 6 
subsamples shall be identified with the name of the product, the labeled 
potency, the date of manufacture, the batch number, and the name and 
address of the manufacturer.
    (2) The sample together with the batch production record and results 
of all tests conducted by or for the manufacturer to determine the 
product's identity, strength, quality, and purity, content uniformity 
and dissolution shall be submitted to the Department of Health and Human 
Services, Public Health Service, FDA National Center for Drug Analysis, 
1114 Market St., St. Louis, MO 63101. The outer wrapper shall be 
identified ``SAMPLE--DIGOXIN CERTIFICATION.''
    (h) The Food and Drug Administration is aware of data with two in 
vitro methods, in addition to that described in The United States 
Pharmacopeia (USP XVIII), developed to measure digoxin tablets 
dissolution. These two methods, the so-called ``paddle-water'' and 
``paddle-acid'' methods, are described below and are identical with the 
exception of the nature of the dissolution medium used in the procedures 
(i.e., distilled or deionized water

[[Page 31]]

vs. dilute hydrochloric acid (0.6 percent volume/volume)). The 
dissolution apparatus used in these two methods differs significantly 
from the apparatus described in the method in the compendium. The Food 
and Drug Administration is aware that the three methods (i.e., USP, 
``paddle-water,'' and ``paddle-acid'') show significant differences in 
dissolution in comparative tests on some formulations. Definitive 
bioavailability data to compare the relative value of each of these 
methods to predict bioavailability of the few formulations where the 
methods show significant differences in dissolution rate are not now 
available. Manufacturers who conduct research utilizing the ``paddle-
water'' and ``paddle-acid'' methods, particularly in comparison with the 
method in The United States Pharmacopeia, shall submit any data obtained 
using these methods to the Food and Drug Administration pursuant to 
section 505(k) of the act.
    (1) Dissolution apparatus.

    (Note: Throughout this procedure use scrupulously clean glassware, 
which previously has been rinsed with dilute hydrochloric acid, 
distilled or deionized water, then with alcohol, and carefully dried. 
Take precautions to prevent contamination from airborne, fluorescent 
particles and from metal and rubber surfaces.) The apparatus consists of 
a suitable water bath, a 1000 milliliter glass vessel (Kimble Glass No. 
26220 or equivalent), a motor, and a polytetrafluoroethylene stirring 
blade (Sargent S-76637, Size B, 3 inch length; or equivalent) on a glass 
stirring shaft (Sargent 5-76636, 14.5 inch length; or equivalent). The 
water bath may be of any convenient size that permits keeping the water 
temperature uniformly at 37 deg. C. plus-minus0.5 deg. C. 
throughout the test. The vessel is spherical, and is provided with three 
ports at the top, one of which is centered. The lower half of the vessel 
is 65 millimeters in inside radius and the vessel's nominal capacity is 
1000 milliliters. The glass stirring shaft from the motor is placed in 
the center port, and one of the outer ports may be used for insertion of 
a thermometer. Samples may be removed for analysis through the other 
port. The motor is fitted with a speed-regulating device that allows the 
motor speed to be held at 50 rpm plus-minus2 rpm. The motor 
is suspended above the vessel in such a way that it may be raised or 
lowered to position the stirring blade. The glass stirring shaft is 10 
millimeters in diameter and about 37 centimeters in length. It must run 
true on the motor axis without perceptible wobble. The 
polytetrafluoroethylene stirring blade is 4 millimeters thick and forms 
a section of a circle, whose diameter is 83 millimeters and which is 
subtended by parallel chords of 42 and 77 millimeters. The blade is 
positioned horizontally, with the 42-millimeter edge down, 2.5 
centimeters plus-minus0.2 centimeter above the lowest inner 
surface of the vessel.

    (2) Reagents--(i) Dissolution medium. For ``paddle-water,'' use 
distilled or deionized water. For ``paddle-acid,'' use dilute 
hydrochloric acid (0.6 percent volume/volume). Use the same batch of 
dissolution medium throughout the test.
    (ii) Standard solutions. Accurately weigh approximately 25 
milligrams of The United States Pharmacopeia Digoxin Reference Standard, 
dissolve in a minimum amount of 95 percent ethanol in a 500 milliliter 
volumetric flask and add 95 percent ethanol to volume and mix. Dilute 
10.0 milliliters of this first solution to 100.0 milliliters with 95 
percent ethanol and mix for the second solution. Just prior to use, 
individually dilute 1.0, 2.0, 3.0, 4.0, and 5.0 milliliter aliquots of 
the second solution with dissolution medium to 50.0 milliliters. These 
solutions are equivalent to 20, 40, 60, 80, and 100 percent of 
dissolution, respectively, for a 0.25 milligram digoxin tablet.
    (iii) Extraction solvent. Prepare a solvent containing 6 volumes of 
chloroform, analytical reagent grade, with 1 volume of n-propyl alcohol, 
analytical reagent grade.
    (iv) Ascorbic acid-methanol solution. Prepare a solution containing 
2 milligrams of ascorbic acid, analytical reagent grade, per 1 
milliliter of methanol, absolute, analytical reagent grade.
    (v) Hydrochloric acid, concentrated reagent grade.
    (vi) Hydrogen peroxide-methanol solution. On the day of use, dilute 
2.0 milliliters of recently assayed 30 percent hydrogen peroxide, 
reagent grade, with methanol, absolute, analytical reagent grade to 
100.0 milliliters. Store in a refrigerator. Just prior to use, dilute 
2.0 milliliters of this solution with methanol to 100.0 milliliters.
    (3) Procedure--(i) Dissolution. Place 500 milliliters of dissolution 
medium in the vessel, immerse it in the constant-temperature bath set at 
37 deg.C.plus-minus0.5 deg.C., and allow the dissolution 
medium to assume the temperature of the bath.

[[Page 32]]

Position the shaft so that there is a distance of 2.5 centimeters 
plus-minus0.2 centimeter between the midpoint of the bottom 
of the blade and the bottom of the vessel. With the stirrer operating at 
a speed of 50 rpmplus-minus2 rpm, place 1 tablet into the 
flask. After 60 minutes, accurately timed, withdraw 25 milliliters, 
using a glass syringe connected to a glass sampling tube, of solution 
from a point midway between the stirring shaft and the wall of the 
vessel, and approximately midway in depth. Filter the solution promptly 
after withdrawal, using a suitable membrane filter of not greater than 
0.8 micron porosity (Millipore AAWP 025 00, or equivalent), mounted in a 
suitable holder (Millipore Swinnex SX00 025 00, or equivalent), 
discarding the first 100 milliliters of filtrate. This is the test 
solution. Repeat the dissolution procedure on 5 additional tablets.
    (ii) Extraction. Transfer 10.0 milliliters of each of the six 
filtrates, 10.0 milliliters of each of the five standard solutions, and 
10.0 milliliters of dissolution medium, to provide a blank, in separate 
60-milliliter separators. Extract each solution with two 10-milliliter 
portions of extraction solvent. Combine the extracts of each solution in 
separate, glass-stoppered, 50-milliliter conical flasks, and evaporate 
on a steam bath with the aid of a stream of nitrogen to dryness, rinsing 
the sides of the flasks with extraction solvent. Take care to ensure 
that all traces of solvent are removed, but avoid prolonged heating. For 
convenience the residues may be stored in a vacuum desiccator overnight.
    (iii) Measurement of fluorescence. Begin with the standard 
solutions, and keep all flasks in the same sequence throughout, so that 
the elapsed time from addition of reagents to reading of fluorescence is 
the same for each. Carry the test solutions, standard solutions, and the 
blank through the determination in one group. Add the following three 
reagents in as rapid a sequence as possible, swirling after each 
addition, treating 1 flask at a time, in the order named: 1.0 milliliter 
of ascorbic acid-methanol solution, 3.0 milliliters of concentrated 
hydrochloric acid, and 1.0 milliliter of hydrogen peroxide-methanol 
solution. Insert the stoppers in the flasks, and after 2 hours, measure 
the fluorescence at about 485 millimicrons, using excitation at about 
372 millimicrons. In order to provide a check on the stability of the 
fluorometer, reread one or more standard solutions. Correct each reading 
for the blank and plot a standard curve of fluorescence versus 
precentage dissolution. Determine the percentage dissolution of digoxin 
in the test solutions by reading from the standard graph.
    (iv) Digoxin tablets formulated so that the quantity of digoxin 
dissolved at one hour, when tested by the method in The United States 
Pharmacopeia (USP XVIII), is greater than 95 percent of the assayed 
amount of digoxin and so that the quantity of digoxin dissolved at 15 
minutes is greater than 90 percent of the assayed amount of digoxin are 
new drugs which may be marketed only with an approved full new drug 
application as provided for in Sec. 314.50 of this chapter. The 
application shall include, but not be limited to, clinical studies 
establishing significantly greater bioavailability than digoxin tablets 
meeting compendial requirements and dosage recommendations based on 
clinical studies establishing the safe and effective use of the 
bioavailable digoxin product. Marketing of these digoxin products will 
be allowed only under a proprietary or trade name, established name, and 
labeling which differs from that used for digoxin tablets that meet all 
of the requirements in The United States Pharmacopeia (USP XVIII) and 
that are formulated so that either (a) the quantity of digoxin dissolved 
at one hour is not more than 95 percent of the assayed amount of digoxin 
or (b) the quantity of digoxin dissolved at 15 minutes is not more than 
90 percent of the assayed amount of digoxin. New drug applications for 
these digoxin products shall be submitted to the Food and Drug 
Administration, Center for Drug Evaluation and Research, Office of Drug 
Evaluation I (HFD-100), 5600 Fishers Lane, Rockville, MD 20857.
[39 FR 11680, Mar. 29, 1974, as amended at 41 FR 43137, Sept. 30, 1976; 
41 FR 49482, Nov. 3, 1976; 50 FR 8996, Mar. 6, 1985; 55 FR 11578, Mar. 
29, 1990]

[[Page 33]]



Sec. 310.501  Patient package inserts for oral contraceptives.

    (a) Requirement for a patient package insert. The safe and effective 
use of oral contraceptive drug products requires that patients be fully 
informed of the benefits and the risks involved in their use. An oral 
contraceptive drug product that does not comply with the requirements of 
this section is misbranded under section 502 of the Federal Food, Drug, 
and Cosmetic Act. Each dispenser of an oral contraceptive drug product 
shall provide a patient package insert to each patient (or to an agent 
of the patient) to whom the product is dispensed, except that the 
dispenser may provide the insert to the parent or legal guardian of a 
legally incompetent patient (or to the agent of either). The patient 
package insert is required to be placed in or accompany each package 
dispensed to the patient.
    (b) Distribution requirements. (1) For oral contraceptive drug 
products, the manufacturer and distributor shall provide a patient 
package insert in or with each package of the drug product that the 
manufacturer or distributor intends to be dispensed to a patient.
    (2) Patient package inserts for oral contraceptives dispensed in 
acute-care hospitals or long-term care facilities will be considered to 
have been provided in accordance with this section if provided to the 
patient before administration of the first oral contraceptive and every 
30 days thereafter, as long as the therapy continues.
    (c) Contents of patient package insert. A patient package insert for 
an oral contraceptive drug product is required to contain the following:
    (1) The name of the drug.
    (2) A summary including a statement concerning the effectiveness of 
oral contraceptives in preventing pregnancy, the contraindications to 
the drug's use, and a statement of the risks and benefits associated 
with the drug's use.
    (3) A statement comparing the effectiveness of oral contraceptives 
to other methods of contraception.
    (4) A boxed warning concerning the increased risks associated with 
cigarette smoking and oral contraceptive use.
    (5) A discussion of the contraindications to use, including 
information that the patient should provide to the prescriber before 
taking the drug.
    (6) A statement of medical conditions that are not contraindications 
to use but deserve special consideration in connection with oral 
contraceptive use and about which the patient should inform the 
prescriber.
    (7) A warning regarding the most serious side effects of oral 
contraceptives.
    (8) A statement of other serious adverse reactions and potential 
safety hazards that may result from the use of oral contraceptives.
    (9) A statement concerning common, but less serious side effects 
which may help the patient evaluate the benefits and risks from the use 
of oral contraceptives.
    (10) Information on precautions the patients should observe while 
taking oral contraceptives, including the following:
    (i) A statement of risks to the mother and unborn child from the use 
of oral contraceptives before or during early pregnancy;
    (ii) A statement concerning excretion of the drug in human milk and 
associated risks to the nursing infant;
    (iii) A statement about laboratory tests which may be affected by 
oral contraceptives; and
    (iv) A statement that identifies activities and drugs, foods, or 
other substances the patient should avoid because of their interactions 
with oral contraceptives.
    (11) Information about how to take oral contraceptives properly, 
including information about what to do if the patient forgets to take 
the product, information about becoming pregnant after discontinuing use 
of the drug, a statement that the drug product has been prescribed for 
the use of the patient and should not be used for other conditions or 
given to others, and a statement that the patient's pharmacist or 
practitioner has a more technical leaflet about the drug product that 
the patient may ask to review.
    (12) A statement of the possible benefits associated with oral 
contraceptive use.

[[Page 34]]

    (13) The following information about the drug product and the 
patient package insert:
    (i) The name and place of business of the manufacturer, packer, or 
distributor, or the name and place of business of the dispenser of the 
product.
    (ii) The date, identified as such, of the most recent revision of 
the patient package insert placed prominently immediately after the last 
section of the labeling.
    (d) Other indications. The patient package insert may identify 
indications in addition to contraception that are identified in the 
professional labeling for the drug product.
    (e) Labeling guidance texts. The Food and Drug Administration issues 
informal labeling guidance texts under Sec. 10.90(b)(9) of this chapter 
to provide assistance in meeting the requirements of this section. A 
request for a copy of the guidance texts should be directed to the 
Center for Drug Evaluation and Research, Division of Metabolism and 
Endocrine Drug Products (HFD-510), Food and Drug Administration, 5600 
Fishers Lane, Rockville, MD 20857.
    (f) Requirement to supplement approved application. Holders of 
approved applications for oral contraceptive drug products that are 
subject to the requirements of this section are required to submit 
supplements under Sec. 314.70(c) of this chapter to provide for the 
labeling required by this section. Such labeling may be put into use 
without advance approval by the Food and Drug Administration.
[54 FR 22587, May 25, 1989]



Sec. 310.502  Certain drugs accorded new drug status through rulemaking procedures.

    (a) The drugs listed in this paragraph (a) have been determined by 
rulemaking procedures to be new drugs within the meaning of section 
201(p) of the act. Except as provided in paragraph (b) of this section, 
an approved new drug application under section 505 of the act and part 
314 of this chapter is required for marketing the following drugs:
    (1) Aerosol drug products for human use containing 1,1,1-
trichloroethane.
    (2) Aerosol drug products containing zirconium.
    (3) Amphetamines (amphetamine, dextroamphetamine, and their salts, 
and levamfetamine and its salts) for human use.
    (4) Camphorated oil drug products.
    (5) Certain halogenated salicylanilides (tribromsalan (TBS, 3,4',5-
tribromosalicylanilide), dibromsalan (DBS, 4', 5-dibromosalicylanilide), 
metabromsalan (MBS, 3, 5-dibromosalicylanilide), and 3,3', 4,5'-
tetrachlorosalicylanilide (TC-SA)) as an ingredient in drug products.
    (6) Chloroform used as an ingredient (active or inactive) in drug 
products.
    (7) Cobalt preparations intended for use by man.
    (8) Intrauterine devices for human use for the purpose of 
contraception that incorporate heavy metals, drugs, or other active 
substances.
    (9) Oral prenatal drugs containing fluorides intended for human use.
    (10) Parenteral drug products in plastic containers.
    (11) Sterilization of drugs by irradiation.
    (12) Sweet spirits of nitre drug products.
    (13) Thorium dioxide for drug use.
    (14) Timed release dosage forms.
    (15) Vinyl chloride as an ingredient, including propellant, in 
aerosol drug products.
    (b) Any drug listed in paragraph (a) of this section, when composed 
wholly or partly of any antibiotic drug, must be certified under section 
507 of the act or exempted from certification under section 507 of the 
act for marketing.
[62 FR 12084, Mar. 14, 1997]



Sec. 310.503  Requirements regarding certain radioactive drugs.

    (a) On January 8, 1963 (28 FR 183), the Commissioner of Food and 
Drugs exempted investigational radioactive new drugs from part 312 of 
this chapter provided they were shipped in complete conformity with the 
regulations issued by the Nuclear Regulatory Commission. This exemption 
also applied to investigational radioactive biologics.
    (b) It is the opinion of the Nuclear Regulatory Commission, and the 
Food and Drug Administration that this exemption should not apply for 
certain specific drugs and that these drugs should be appropriately 
labeled for uses

[[Page 35]]

for which safety and effectiveness can be demonstrated by new-drug 
applications or through licensing by the Public Health Service in the 
case of biologics. Continued distribution under the investigational 
exemption when the drugs are intended for established uses will not be 
permitted.
    (c) Based on its experience in regulating investigational 
radioactive pharmaceuticals, the Nuclear Regulatory Commission has 
compiled a list of reactor-produced isotopes for which it considers that 
applicants may reasonably be expected to submit adequate evidence of 
safety and effectiveness for use as recommended in appropriate labeling. 
Such use may include, among others, the uses in this tabulation:

                                                                        
------------------------------------------------------------------------
        Isotope              Chemical form                 Use          
------------------------------------------------------------------------
Chromium 51...........  Chromate...............  Spleen scans.          
    Do................  ......do...............  Placenta localization. 
    Do................  ......do...............  Red blood cell labeling
                                                  and survival studies. 
    Do................  Labeled human serum      Gastrointestinal       
                         albumin.                 protein loss studies. 
    Do................  ......do...............  Placenta localization. 
    Do................  Labeled red blood cells      Do.                
Cobalt 58 or Cobalt 60  Labeled cyanocobalamin.  Intestinal absorption  
                                                  studies.              
Gold 198..............  Colloidal..............  Liver scans.           
    Do................  ......do...............  Intracavitary treatment
                                                  of pleural effusions  
                                                  and/or ascites.       
    Do................  ......do...............  Interstitial treatment 
                                                  of cancer.            
Iodine 131............  Iodide.................  Diagnosis of thyroid   
                                                  functions.            
    Do................  ......do...............  Thyroid scans.         
    Do................  ......do...............  Treatment of           
                                                  hyperthyroidism and/or
                                                  cardiac dysfunction.  
    Do................  ......do...............  Treatment of thyroid   
                                                  carcinoma.            
    Do................  Iodinated human serum    Blood volume           
                         albumin.                 determinations.       
    Do................  ......do...............  Cisternography.        
    Do................  ......do...............  Brain tumor            
                                                  localization.         
    Do................  ......do...............  Placenta localization. 
    Do................  ......do...............  Cardiac scans for      
                                                  determination of      
                                                  pericardial effusions.
    Do................  Rose Bengal............  Liver function studies.
    Do................  ......do...............  Liver scans.           
    Do................  Iodopyracet, sodium      Kidney function studies
                         iodohippurate, sodium    and kidney scans.     
                         diatrizoate,                                   
                         diatrizoate                                    
                         methylglucamine,                               
                         sodium diprotrizoate,                          
                         sodium acetrizoate, or                         
                         sodium iothalamate.                            
    Do................  Labeled fats and/or      Fat absorption studies.
                         fatty acids.                                   
    Do................  Cholografin............  Cardiac scans for      
                                                  determination of      
                                                  pericardial effusions.
    Do................  Macroaggregated          Lung scans.            
                         iodinated human serum                          
                         albumin.                                       
    Do................  Colloidal                Liver scans.           
                         microaggregated human                          
                         serum albumin.                                 
Iodine 125............  Iodide.................  Diagnosis of thyroid   
                                                  function.             
    Do................  Iodinated human serum    Blood volume           
                         albumin.                 determinations.       
    Do................  Rose Bengal............  Liver function studies.
    Do................  Iodopyracet, sodium      Kidney function        
                         iodohippurate, sodium    studies.              
                         diatrizoate,                                   
                         diatrizoate methyl-                            
                         glucamine, sodium                              
                         diprotrizoate, sodium                          
                         acetrizoate, or sodium                         
                         iothalamate.                                   
    Do................  Labeled fats and/or      Fat absorption studies.
                         fatty acids.                                   
Iron 59...............  Chloride, citrate and/   Iron turnover studies. 
                         or sulfate.                                    
Krypton 85............  Gas....................  Diagnosis of cardiac   
                                                  abnormalities.        
Mercury 197...........  Chlormerodrin..........  Kidney scans.          
    Do................  ......do...............  Brain scans.           
Mercury 203 \1\.......  ......do...............  Kidney scans.          
    Do................  ......do...............  Brain scans.           
Phosphorus 32.........  Soluble phosphate......  Treatment of           
                                                  polycythemia vera.    
    Do................  ......do...............  Treatment of leukemia  
                                                  and bone metastasis.  
    Do................  Colloidal chromic        Intracavitary treatment
                         phosphate.               of pleural effusions  
                                                  and/or ascites.       
    Do................  ......do...............  Interstitial treatment 
                                                  of cancer.            
Potassium 42..........  Chloride...............  Potassium space        
                                                  studies.              
Selenium 75...........  Labeled methionine.....  Pancreas scans.        
Strontium 85..........  Nitrate or chloride....  Bone scans on patients 
                                                  with diagnosed cancer.
Technetium 99m........  Pertechnetate..........  Brain scans.           
    Do................  ......do...............  Thyroid scans.         
    Do................  Sulfur colloid.........  Liver and spleen scans.
    Do................  Pertechnetate..........  Placenta localization. 
    Do................  ......do...............  Blood pool scans.      
    Do................  ......do...............  Salivary gland scans.  

[[Page 36]]

                                                                        
    Do................  Diethylenetri-amine      Kidney scans.          
                         pentaacetic acid                               
                         (DTPA).                                        
Xenon 133.............  Gas....................  Diagnosis of cardia    
                                                  abnormalities.        
                                                  Cerebral bloodflow    
                                                  studies. Pulmonary    
                                                  function studies.     
                                                  Muscle bloodflow      
                                                  studies.              
------------------------------------------------------------------------
\1\ This item has been removed from the AEC list for kidney scans but is
  included as the requirements of this order are applicable.            

    (d)(1) In view of the extent of experience with the isotopes listed 
in paragraph (c) of this section, the Nuclear Regulatory Commission and 
the Food and Drug Administration conclude that such isotopes should not 
be distributed under investigational-use labeling when they are actually 
intended for use in medical practice.
    (2) The exemption referred to in paragraph (a) of this section, as 
applied to any drug or biologic containing any of the isotopes listed in 
paragraph (c) of this section, in the ``chemical form'' and intended for 
the uses stated, is terminated on March 3, 1972, except as provided in 
paragraph (d)(3) of this section.
    (3) The exemption referred to in paragraph (a) of this section, as 
applied to any drug or biologic containing any of the isotopes listed in 
paragraph (c) of this section, in the ``chemical form'' and intended for 
the uses stated, for which drug a new drug application or a 
``Investigational New Drug Application'' was submitted prior to March 3, 
1972, or for which biologic an application for product license or 
``Investigational New Drug Application'' was submitted prior to March 3, 
1972, is terminated on August 20, 1976, unless an approvable notice was 
issued on or before August 20, 1976, in which case the exemption is 
terminated either upon the subsequent issuance of a nonapprovable notice 
for the new drug application or on November 20, 1976, whichever occurs 
first.
    (e) No exemption from section 505 of the act or from part 312 of 
this chapter is in effect or has been in effect for radioactive drugs 
prepared from accelerator-produced radioisotopes, naturally occurring 
isotopes, or nonradioactive substances used in conjunction with 
isotopes.
    (f)(1) Based on its experience in regulating investigational 
radioactive pharmaceuticals, the Nuclear Regulatory Commission has 
compiled a list of reactor-produced isotopes for which it considers that 
applicants may reasonably be expected to submit adequate evidence of 
safety and effectiveness for use as recommended in appropriate labeling; 
such use may include, among others, the uses in this tabulation:

                                                                        
------------------------------------------------------------------------
        Isotope              Chemical form                 Use          
------------------------------------------------------------------------
Fluorine 18...........  Fluoride...............  Bone imaging.          
Indium-113m...........  Diethylenetriamine       Brain imaging; kidney  
                         pentaacetic acid         imaging.              
                         (DTPA).                                        
    Do................  Chloride...............  Placenta imaging; blood
                                                  pool imaging.         
Technetium 99m........  Human serum albumin      Lung imaging.          
                         microspheres.                                  
    Do................  Diethylenetriamine       Kidney imaging; kidney 
                         pentaacetic acid (Sn).   function studies.     
    Do................  ......do...............  Brain imaging.         
    Do................  Polyphosphates.........  Bone imaging.          
    Do................  Technetated aggregated   Lung imaging.          
                         albumin (human).                               
    Do................  Disodium etidronate....  Bone imaging.          
------------------------------------------------------------------------

    (2) In view of the extent of experience with the isotopes listed in 
paragraph (f)(1) of this section, the Nuclear Regulatory Commission and 
the Food and Drug Administration conclude that they should not be 
distributed under investigational-use labeling when they are actually 
intended for use in medical practice.
    (3) Any manufacturer or distributor interested in continuing to ship 
in interstate commerce drugs containing the isotopes listed in paragraph 
(f)(1) of this section for any of the indications listed, shall submit, 
on or before August 25, 1975 to the Center for Drug Evaluation and 
Research, Food and Drug Administration, 5600 Fishers Lane, Rockville, MD 
20857, a new drug application or a ``Investigational New Drug 
Application'' for each such drug for which the manufacturer or 
distributor does not have an approved new drug application pursuant to 
section 505(b) of the act. If the drug is a biologic, a 
``Investigational New Drug Application'' or an application for a license 
under section 351 of the Public Health Service Act shall be submitted to 
the Center for Biologics Evaluation and

[[Page 37]]

Research, Food and Drug Administration, 8800 Rockville Pike, Bethesda, 
MD 20014, in lieu of any submission to the Center for Drug Evaluation 
and Research.
    (4) The exemption referred to in paragraph (a) of this section, as 
applied to any drug or biologic containing any of the isotopes listed in 
paragraph (f)(1) of this section, in the ``chemical form'' and intended 
for the uses stated, is terminated on August 26, 1975 except as provided 
in paragraph (f)(5) of this section.
    (5)(i) Except as provided in paragraph (f)(5)(ii) of this section, 
the exemption referred to in paragraph (a) of this section, as applied 
to any drug containing any of the isotopes listed in paragraph (f)(1) of 
this section, in the ``chemical form'' and intended for the uses stated, 
for which drug a new drug application or ``Investigational New Drug 
Application'' was submitted to the Center for Drug Evaluation and 
Research on or before August 25, 1975 is terminated on August 20, 1976, 
unless an approvable notice was issued on or before August 20, 1976, in 
which case the exemption is terminated either upon the subsequent 
issuance of a nonapprovable notice for the new drug application or on 
November 20, 1976, whichever occurs first.
    (ii) The exemption referred to in paragraph (a) of this section, as 
applied to any biologic containing any of the isotopes listed in 
paragraph (f)(1) of this section in the ``chemical form'' and intended 
for the uses stated, for which biologic an application for product 
license or ``Investigational New Drug Application'' was submitted to the 
Center for Biologics Evaluation and Research on or before August 25, 
1975 is terminated on October 20, 1976, unless an approvable notice was 
issued on or before October 20, 1976, in which case the exemption is 
terminated either upon the subsequent issuance of a nonapprovable notice 
for the new drug application or on January 20, 1977, whichever occurs 
first.
    (g) The exemption referred to in paragraph (a) of this section, as 
applied to any drug intended solely for investigational use as part of a 
research project, which use had been approved on or before July 25, 1975 
in accordance with 10 CFR 35.11 (or equivalent regulation of an 
Agreement State) is terminated on February 20, 1976 if the manufacturer 
of such drug or the sponsor of the investigation of such drug submits on 
or before August 25, 1975 to the Food and Drug Administration, Bureau of 
Drugs, HFD-150, 5600 Fishers Lane, Rockville, MD 20857, the following 
information:
    (1) The research project title;
    (2) A brief description of the purpose of the project;
    (3) The name of the investigator responsible;
    (4) The name and license number of the institution holding the 
specific license under 10 CFR 35.11 (or equivalent regulation of an 
Agreement State);
    (5) The name and maximum amount per subject of the radionuclide 
used;
    (6) The number of subjects involved; and
    (7) The date on which the administration of the radioactive drugs is 
expected to be completed.
    (h) The exemption referred to in paragraph (a) of this section, as 
applied to any drug not referred to in paragraphs (d), (f), and (g) of 
this section, is terminated on August 26, 1975.
[39 FR 11680, Mar. 29, 1974, as amended at 40 FR 31307, July 25, 1975; 
40 FR 44543, Sept. 29, 1975; 41 FR 35171, Aug. 20, 1976; 41 FR 42947, 
Sept. 29, 1976; 50 FR 8996, Mar. 6, 1985; 55 FR 11578, Mar. 29, 1990]



Sec. 310.509  Parenteral drug products in plastic containers.

    (a) Any parenteral drug product packaged in a plastic immediate 
container is not generally recognized as safe and effective, is a new 
drug within the meaning of section 201(p) of the act, and requires an 
approved new drug application as a condition for marketing. An 
``Investigational New Drug Application'' set forth in part 312 of this 
chapter is required for clinical investigations designed to obtain 
evidence of safety and effectiveness.
    (b) As used in this section, the term ``large volume parenteral drug 
product'' means a terminally sterilized aqueous drug product packaged in 
a single-dose container with a capacity of 100 milliliters or more and 
intended to be administered or used intravenously in a human.

[[Page 38]]

    (c) Until the results of compatibility studies are evaluated, a 
large volume parenteral drug product for intravenous use in humans that 
is packaged in a plastic immediate container on or after April 16, 1979, 
is misbranded unless its labeling contains a warning that includes the 
following information:
    (1) A statement that additives may be incompatible.
    (2) A statement that, if additive drugs are introduced into the 
parenteral system, aseptic techniques should be used and the solution 
should be thoroughly mixed.
    (3) A statement that a solution containing an additive drug should 
not be stored.
    (d) This section does not apply to a biological product licensed 
under the Public Health Service Act of July 1, 1944 (42 U.S.C. 201).
[62 FR 12084, Mar. 14, 1997]



Sec. 310.515  Patient package inserts for estrogens.

    (a) Requirement for a patient package insert. FDA concludes that the 
safe and effective use of drug products containing estrogens requires 
that patients be fully informed of the benefits and risks involved in 
the use of these drugs. Accordingly, except as provided in paragraph (e) 
of this section, each estrogen drug product restricted to prescription 
distribution, including products containing estrogens in fixed 
combinations with other drugs, shall be dispensed to patients with a 
patient package insert containing information concerning the drug's 
benefits and risks. An estrogen drug product that does not comply with 
the requirements of this section is misbranded under section 502(a) of 
the Federal Food, Drug, and Cosmetic Act.
    (b) Distribution requirements. (1) For estrogen drug products, the 
manufacturer and distributor shall provide a patient package insert in 
or with each package of the drug product that the manufacturer or 
distributor intends to be dispensed to a patient.
    (2) In the case of estrogen drug products in bulk packages intended 
for multiple dispensing, and in the case of injectables in multiple-dose 
vials, a sufficient number of patient labeling pieces shall be included 
in or with each package to assure that one piece can be included with 
each package or dose dispensed or administered to every patient. Each 
bulk package shall be labeled with instructions to the dispensor to 
include one patient labeling piece with each package dispensed or, in 
the case of injectables, with each dose administered to the patient. 
This section does not preclude the manufacturer or labeler from 
distributing additional patient labeling pieces to the dispensor.
    (3) Patient package inserts for estrogens dispensed in acute-care 
hospitals or long-term care facilities will be considered to have been 
provided in accordance with this section if provided to the patient 
before administration of the first estrogen and every 30 days 
thereafter, as long as the therapy continues.
    (c) Patient package insert contents. A patient package insert for an 
estrogen drug product is required to contain the following information:
    (1) The name of the drug.
    (2) The name and place of business of the manufacturer, packer, or 
distributor.
    (3) A statement regarding the benefits and proper uses of estrogens.
    (4) The contraindications to use, i.e., when estrogens should not be 
used.
    (5) A description of the most serious risks associated with the use 
of estrogens.
    (6) A brief summary of other side effects of estrogens.
    (7) Instructions on how a patient may reduce the risks of estrogen 
use.
    (8) The date, identified as such, of the most recent revision of the 
patient package insert.
    (d) Guidance language. The Food and Drug Administration issues 
informal labeling guidance texts under Sec. 10.90(b)(9) of this chapter 
to provide assistance in meeting the requirements of paragraph (c) of 
this section. Requests for a copy of the guidance text should be 
directed to the Center for Drug Evaluation and Research, Division of 
Metabolism and Endocrine Drug Products (HFD-510), Food and Drug 
Administration, 5600 Fishers Lane, Rockville, MD 20857.

[[Page 39]]

    (e) Exemptions. This section does not apply to estrogen-progestogen 
oral contraceptives. Labeling requirements for these products are set 
forth in Sec. 310.501.
    (f) Requirement to supplement approved application. Holders of 
approved applications for estrogen drug products that are subject to the 
requirements of this section must submit supplements under 
Sec. 314.70(c) of this chapter to provide for the labeling required by 
paragraph (a) of this section. Such labeling may be put into use without 
advance approval by the Food and Drug Administration.
[55 FR 18723, May 4, 1990]



Sec. 310.516  Progestational drug products; labeling directed to the patient.

    (a) The Commissioner of Food and Drugs concludes that the safe and 
effective use of any progestational drug product requires that patients 
be informed that there is an increased risk of birth defects in children 
whose mothers have taken this drug during the first 4 months of 
pregnancy. Accordingly, except as provided by paragraph (d) of this 
section, any progestational drug product that is the subject of a new 
drug application approved either before or after October 9, 1962 and all 
identical, related, or similar drug products as defined in Sec. 310.6, 
whether or not the subject of an approved new drug application, shall be 
dispensed to patients with labeling in lay language containing such a 
warning. The patient labeling shall be provided as a separate printed 
leaflet independent of any additional materials.
    (b) The patient labeling shall specifically include the following:
    (1) Name of the drug.
    (2) Name and place of business of the manufacturer, packer, or 
distributor.
    (3) A warning that there is an increased risk of birth defects in 
children whose mothers take this drug during the first 4 months of 
pregnancy.
    (4) A brief discussion of the nature of the risks of birth defects 
resulting from the use of these drugs during the first 4 months of 
pregnancy.
    (5) A brief statement that these drugs are no longer considered safe 
as a test for pregnancy.
    (6) A statement that the patient should inform her physician as soon 
as possible if she discovers that she was pregnant when she took the 
drug.
    (c) The patient labeling shall be printed in accordance with the 
following specifications:
    (1) The minimum letter size shall be one-sixteenth of an inch in 
height.
    (2) Letter heights pertain to the lower-case letter ``o'' or its 
equivalent that shall meet the minumim height standard.
    (3) Type used shall conform to the minimum letter height. The body 
copy shall contain 1-point leading, noncondensed type, and shall not 
contain any light-face type or small capital letters.
    (d) This section does not apply to a progestogen-containing product 
intended for contraception, which shall be labeled according to the 
requirements of Sec. 310.501.
    (e)(1) Patient labeling for each progestational drug product shall 
be provided in or with each package intended to be dispensed to the 
patient. Patient labeling for drug products dispensed in acute-care 
hospitals or long-term care facilities will be considered to have been 
provided in accordance with this section if provided to the patient 
before first administration of the drug and every 30 days thereafter, as 
long as the therapy continues.
    (2) In the case of progestational drug products in bulk packages 
intended for multiple dispensing, a sufficient number of patient-
labeling pieces shall be included in or shall accompany each bulk 
package to assure that one can be included with each package dispensed 
to every patient. Each bulk package shall be labeled with instructions 
to the dispenser to include one patient-labeling piece with each package 
dispensed to the patient. This section does not preclude the 
manufacturer or labeler from distributing additional patient-labeling 
pieces to the dispenser.
    (3) In the case of progestational drug products for injection, each 
package shall include a sufficient number of patient-labeling pieces for 
the volume of the vial, and instructions to the practitioner 
administering the drug to give one patient-labeling piece to each 
premenopausal woman, except those in

[[Page 40]]

whom childbearing is impossible, receiving the drug.
    (4) This section does not apply to oral dosage forms labeled solely 
for the treatment of advanced cancer.
    (5) Any progestational drug product, except as noted in paragraphs 
(d) and (e)(4) of this section, that is not labeled as required by this 
section and is either introduced or delivered for introduction into 
interstate commerce, or held for sale after shipment in interstate 
commerce, is misbranded under section 502 of the Federal Food, Drug, and 
Cosmetic Act. However, a progestational drug product in the possession 
of a wholesaler or retailer before December 12, 1978, is not misbranded 
if adequate numbers of copies of the patient labeling are furnished to 
the wholesaler or retailer to permit any retail purchaser after that 
date to obtain such labeling with the product. The requirement that any 
progestational drug product be dispensed with patient labeling, as 
applied to physicians who dispense or administer the drug, will not be 
effective for supplies in their possession on the effective date, but 
will apply only to supplies received thereafter.
    (f) The Food and Drug Administration has available guideline patient 
labeling for progestational drug products that includes information 
responsive to all items specified in paragraph (b) of this section. This 
labeling was published in a separate notice appearing in the Federal 
Register of January 12, 1989. Any person may rely on this labeling as 
complying with paragraph (b) of this section.
    (g) Holders of approved new drug applications for progestational 
drug products that are subject to the requirements of this section shall 
submit supplements under Sec. 314.70(c) of this chapter to provide for 
the labeling required by paragraph (a) of this section.
[43 FR 47181, Oct. 13, 1978, as amended at 46 FR 53657, Oct. 30, 1981; 
54 FR 1163, Jan. 12, 1989]



Sec. 310.517  Labeling for oral hypoglycemic drugs of the sulfonylurea class.

    (a) The University Group Diabetes Program clinical trial has 
reported an association between the administration of tolbutamide and 
increased cardiovascular mortality. The Food and Drug Administration has 
concluded that this reported association provides adequate basis for a 
warning in the labeling. In view of the similarities in chemical 
structure and mode of action, the Food and Drug Administration also 
believes it is prudent from a safety standpoint to consider that the 
possible increased risk of cardiovascular mortality from tolbutamide 
applies to all other sulfonylurea drugs as well. Therefore, the labeling 
for oral hypoglycemic drugs of the sulfonylurea class shall include a 
warning concerning the possible increased risk of cardiovascular 
mortality associated with such use, as set forth in paragraph (b) of 
this section.
    (b) Labeling for oral hypoglycemic drugs of the sulfonylurea class 
shall include in boldface type at the beginning of the ``Warnings'' 
section of the labeling the following statement:

      Special Warning on Increased Risk of Cardiovascular Mortality

    The administration of oral hypoglycemic drugs has been reported to 
be associated with increased cardiovascular mortality as compared to 
treatment with diet alone or diet plus insulin. This warning is based on 
the study conducted by the University Group Diabetes Program (UGDP), a 
long-term prospective clinical trial designed to evaluate the 
effectiveness of glucose-lowering drugs in preventing or delaying 
vascular complications in patients with non-insulin-dependent diabetes. 
The study involved 823 patients who were randomly assigned to one of 
four treatment groups (Diabetes, 19 (supp. 2): 747-830, 1970).
    UGDP reported that patients treated for 5 to 8 years with diet plus 
a fixed dose of tolbutamide (1.5 grams per day) had a rate of 
cardiovascular mortality approximately 2\1/2\ times that of patients 
treated with diet alone. A significant increase in total mortality was 
not observed, but the use of tolbutamide was discontinued based on the 
increase in cardiovascular mortality, thus limiting the opportunity for 
the study to show an increase in overall mortality. Despite controversy 
regarding the interpretation of these results, the findings of the UGDP 
study provide an adequate basis for this warning. The patient should be 
informed of the potential risks and advantages of (name of drug) and of 
alternative modes of therapy.
    Although only one drug in the sulfonylurea class (tolbutamide) was 
included in this study, it is prudent from a safety standpoint to 
consider that this warning may also apply

[[Page 41]]

to other oral hypoglycemic drugs in this class, in view of their close 
similarities in mode of action and chemical structure.
[49 FR 14331, Apr. 11, 1984]



Sec. 310.518  Drug products containing iron or iron salts.

    Drug products containing elemental iron or iron salts as an active 
ingredient in solid oral dosage form, e.g., tablets or capsules shall 
meet the following requirements:
    (a) Packaging. If the product contains 30 milligrams or more of iron 
per dosage unit, it shall be packaged in unit-dose packaging. ``Unit-
dose packaging'' means a method of packaging a product into a 
nonreusable container designed to hold a single dosage unit intended for 
administration directly from that container, irrespective of whether the 
recommended dose is one or more than one of these units. The term 
``dosage unit'' means the individual physical unit of the product, e.g., 
tablet or capsule. Iron-containing drugs that are subject to this 
regulation are also subject to child-resistant special packaging 
requirements in 16 CFR parts 1700, 1701, and 1702.
    (b) Temporary exemption. (1) Drug products offered in solid oral 
dosage form (e.g., tablets or capsules), and containing 30 milligrams or 
more of iron per dosage unit, are exempt from the provisions of 
paragraph (a) of this section until January 15, 1998, if the sole source 
of iron in the drug product is carbonyl iron that meets the 
specifications of Sec. 184.1375 of this chapter.
    (2) If this temporary exemption is not extended or made permanent, 
such drug products shall be in compliance with the provisions of 
paragraph (a) of this section on or before July 15, 1998.
    (c) Labeling. (1) The label of any drug in solid oral dosage form 
(e.g., tablets or capsules) that contains iron or iron salts for use as 
an iron source shall bear the following statement:

    WARNING: Accidental overdose of iron-containing products is a 
leading cause of fatal poisoning in children under 6. Keep this product 
out of reach of children. In case of accidental overdose, call a doctor 
or poison control center immediately.

    (2)(i) The warning statement required by paragraph (c)(1) of this 
section shall appear prominently and conspicuously on the information 
panel of the immediate container label.
    (ii) If a drug product is packaged in unit-dose packaging, and if 
the immediate container bears labeling but not a label, the warning 
statement required by paragraph (c)(1) of this section shall appear 
prominently and conspicuously on the immediate container labeling in a 
way that maximizes the likelihood that the warning is intact until all 
of the dosage units to which it applies are used.
    (3) Where the immediate container is not the retail package, the 
warning statement required by paragraph (c)(1) of this section shall 
also appear prominently and conspicuously on the information panel of 
the retail package label.
    (4) The warning statement shall appear on any labeling that contains 
warnings.
    (5) The warning statement required by paragraph (c)(1) of this 
section shall be set off in a box by use of hairlines.
    (d) The iron-containing inert tablets supplied in monthly packages 
of oral contraceptives are categorically exempt from the requirements of 
paragraphs (a) and (c) of this section.
[62 FR 2250, Jan. 15, 1997; 62 FR 15111, Mar. 31, 1997]



Sec. 310.519  Drug products marketed as over-the-counter (OTC) daytime sedatives.

    (a) Antihistamines, bromides, and scopolamine compounds, either 
singly or in combinations, have been marketed as ingredients in over-
the-counter (OTC) drug products for use as daytime sedatives. The 
following claims have been made for daytime sedative products: 
``occasional simple nervous tension,'' ``nervous irritability,'' 
``nervous tension headache,'' ``simple nervousness due to common every 
day overwork and fatigue,'' ``a relaxed feeling,'' ``calming down and 
relaxing,'' ``gently soothe away the tension,'' ``calmative,'' 
``resolving that irritability that ruins your day,'' ``helps you 
relax,'' ``restlessness,'' ``when you're under occasional stress . . . 
helps you work relaxed.'' Based on

[[Page 42]]

evidence presently available, there are no ingredients that can be 
generally recognized as safe and effective for use as OTC daytime 
sedatives.
    (b) Any OTC drug product that is labeled, represented, or promoted 
as an OTC daytime sedative (or any similar or related indication) is 
regarded as a new drug within the meaning of section 201(p) of the 
Federal Food, Drug, and Cosmetic Act for which an approved new drug 
application under section 505 of the act and part 314 of this chapter is 
required for marketing.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted as an OTC daytime 
sedative (or any similar or related indication) is safe and effective 
for the purpose intended must comply with the requirements and 
procedures governing the use of investigational new drugs set forth in 
part 312 of this chapter.
    (d) Any OTC daytime sedative drug product introduced into interstate 
commerce after December 24, 1979, that is not in compliance with this 
section is subject to regulatory action.
[44 FR 36380, June 22, 1979; 45 FR 47422, July 15, 1980, as amended at 
55 FR 11579, Mar. 29, 1990]



Sec. 310.527  Drug products containing active ingredients offered over-the-counter (OTC) for external use as hair growers or for hair loss prevention.

    (a) Amino acids, aminobenzoic acid, ascorbic acid, benzoic acid, 
biotin and all other B-vitamins, dexpanthenol, estradiol and other 
topical hormones, jojoba oil, lanolin, nucleic acids, polysorbate 20, 
polysorbate 60, sulfanilamide, sulfur 1 percent on carbon in a fraction 
of paraffinic hydrocarbons, tetracaine hydrochloride, urea, and wheat 
germ oil have been marketed as ingredients in OTC drug products for 
external use as hair growers or for hair loss prevention. There is a 
lack of adequate data to establish general recognition of the safety and 
effectiveness of these or any other ingredients intended for OTC 
external use as a hair grower or for hair loss prevention. Based on 
evidence currently available, all labeling claims for OTC hair grower 
and hair loss prevention drug products for external use are either 
false, misleading, or unsupported by scientific data. Therefore, any OTC 
drug product for external use containing an ingredient offered for use 
as a hair grower or for hair loss prevention cannot be considered 
generally recognized as safe and effective for its intended use.
    (b) Any OTC drug product that is labeled, represented, or promoted 
for external use as a hair grower or for hair loss prevention is 
regarded as a new drug within the meaning of section 201(p) of the 
Federal Food, Drug, and Cosmetic Act (the act), for which an approved 
new drug application under section 505 of the act and part 314 of this 
chapter is required for marketing. In the absence of an approved new 
drug application, such product is also misbranded under section 502 of 
the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for OTC external use as a 
hair grower or for hair loss prevention is safe and effective for the 
purpose intended must comply with the requirements and procedures 
governing the use of investigational new drugs set forth in part 312 of 
this chapter.
    (d) After January 8, 1990, any such OTC drug product initially 
introduced or initially delivered for introduction into interstate 
commerce that is not in compliance with this section is subject to 
regulatory action.
[54 FR 28777, July 7, 1989]



Sec. 310.528  Drug products containing active ingredients offered over-the-counter (OTC) for use as an aphrodisiac.

    (a) Any product that bears labeling claims that it will arouse or 
increase sexual desire, or that it will improve sexual performance, is 
an aphrodisiac drug product. Anise, cantharides, don qual, estrogens, 
fennel, ginseng, golden seal, gotu kola, Korean ginseng, licorice, 
mandrake, methyltestosterone, minerals, nux vomica, Pega Palo, 
sarsaparilla, strychnine, testosterone, vitamins, yohimbine, yohimbine 
hydrochloride, and yohimbinum have been present as ingredients in such 
drug products. Androgens (e.g., testosterone and methyltestosterone) and 
estrogens

[[Page 43]]

are powerful hormones when administered internally and are not safe for 
use except under the supervision of a physician. There is a lack of 
adequate data to establish general recognition of the safety and 
effectiveness of any of these ingredients, or any other ingredient, for 
OTC use as an aphrodisiac. Labeling claims for aphrodisiacs for OTC use 
are either false, misleading, or unsupported by scientific data. The 
following claims are examples of some that have been made for 
aphrodisiac drug products for OTC use: ``acts as an aphrodisiac;'' 
``arouses or increases sexual desire and improves sexual performance;'' 
``helps restore sexual vigor, potency, and performance;'' ``improves 
performance, staying power, and sexual potency;'' and ``builds virility 
and sexual potency.'' Based on evidence currently available, any OTC 
drug product containing ingredients for use as an aphrodisiac cannot be 
generally recognized as safe and effective.
    (b) Any OTC drug product that is labeled, represented, or prompted 
for use as an aphrodisiac is regarded as a new drug within the meaning 
of section 201(p) of the Federal Food, Drug, and Cosmetic Act, (the 
act), for which an approved new drug application under section 505 of 
the act and part 314 of this chapter is required for marketing. In the 
absence of an approved new drug application, such product is also 
misbranded under section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for OTC use as an 
aphrodisiac is safe and effective for the purpose intended must comply 
with the requirements and procedures governing the use of 
investigational new drugs set forth in part 312 of this chapter.
    (d) After January 8, 1990, any such OTC drug product initially 
introduced or initially delivered for introduction into interstate 
commerce that is not in compliance with this section is subject to 
regulatory action.
[54 FR 28786, July 7, 1989]



Sec. 310.529  Drug products containing active ingredients offered over-the-counter (OTC) for oral use as insect repellents.

    (a) Thiamine hydrochloride (vitamin B-1) has been marketed as an 
ingredient in over-the-counter (OTC) drug products for oral use as an 
insect repellent (an orally administered drug product intended to keep 
insects away). There is a lack of adequate data to establish the 
effectiveness of this, or any other ingredient for OTC oral use as an 
insect repellent. Labeling claims for OTC orally administered insect 
repellent drug products are either false, misleading, or unsupported by 
scientific data. The following claims are examples of some that have 
been made for orally administered OTC insect repellent drug products: 
``Oral mosquito repellent,'' ``mosquitos avoid you,'' ``bugs stay 
away,'' ``keep mosquitos away for 12 to 24 hours,'' and ``the newest way 
to fight mosquitos.'' Therefore, any drug product containing ingredients 
offered for oral use as an insect repellent cannot be generally 
recognized as safe and effective.
    (b) Any OTC drug product that is labeled, represented, or promoted 
for oral use as an insect repellent is regarded as a new drug within the 
meaning of section 201(p) of the Federal Food, Drug and Cosmetic Act for 
which an approved new drug application under section 505 of the act and 
part 314 of this chapter is required for marketing. In the absence of an 
approved new drug application, such product is also misbranded under 
section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted OTC for oral use as an 
insect repellent is safe and effective for the purpose intended must 
comply with the requirements and procedures governing the use of 
investigational new drugs set forth in part 312 of this chapter.
    (d) Any such drug product in interstate commerce after December 17, 
1985, that is not in compliance with

[[Page 44]]

this section is subject to regulatory action.
[40 FR 25171, June 17, 1985, as amended at 55 FR 11579, Mar. 29, 1990]



Sec. 310.530  Topically applied hormone-containing drug products for over-the-counter (OTC) human use.

    (a) The term ``hormone'' is used broadly to describe a chemical 
substance formed in some organ of the body, such as the adrenal glands 
or the pituitary, and carried to another organ or tissue, where it has a 
specific effect. Hormones include, for example, estrogens, progestins, 
androgens, anabolic steroids, and adrenal corticosteroids, and synthetic 
analogs. Estrogens, progesterone, pregnenolone, and pregnenolone acetate 
have been present as ingredients in OTC drug products marketed for 
topical use as hormone creams. However, there is a lack of adequate data 
to establish effectiveness for any OTC drug use of these ingredients. 
Therefore, with the exception of those hormones identified in paragraph 
(e) of this section, any OTC drug product containing an ingredient 
offered for use as a topically applied hormone cannot be considered 
generally recognized as safe and effective for its intended use. The 
intended use of the product may be inferred from the product's labeling, 
promotional material, advertising, and any other relevant factor. The 
use of the word ``hormone'' in the text of the labeling or in the 
ingredient statement is an implied drug claim. The claim implied by the 
use of this term is that the product will have a therapeutic or some 
other physiological effect on the body. Therefore, reference to a 
product as a ``hormone cream'' or any statement in the labeling 
indicating that ``hormones'' are present in the product, or any 
statement that features or emphasizes the presence of a hormone 
ingredient in the product, will be considered to be a therapeutic claim 
for the product, or a claim that the product will affect the structure 
or function of the body, and will consequently cause the product to be a 
drug.
    (b) Any OTC drug product that is labeled, represented, or promoted 
as a topically applied hormone-containing product for drug use, with the 
exception of those hormones identified in paragraph (e) of this section, 
is regarded as a new drug within the meaning of section 201(p) of the 
act, for which an approved application or abbreviated application under 
section 505 of the act and part 314 of this chapter is required for 
marketing. In the absence of an approved new drug application or 
abbreviated new drug application, such product is also misbranded under 
section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for OTC use as a 
topically applied hormone-containing drug product is safe and effective 
for the purpose intended must comply with the requirements and 
procedures governing the use of investigational new drugs set forth in 
part 312 of this chapter.
    (d) After March 9, 1994, any such OTC drug product initially 
introduced or initially delivered for introduction into interstate 
commerce that is not in compliance with this section is subject to 
regulatory action.
    (e) This section does not apply to hydrocortisone and hydrocortisone 
acetate labeled, represented, or promoted for OTC topical use in 
accordance with part 348 of this chapter.
[58 FR 47610, Sept. 9, 1993]



Sec. 310.531  Drug products containing active ingredients offered over-the-counter (OTC) for the treatment of boils.

    (a) Aminacrine hydrochloride, benzocaine, bismuth subnitrate, 
calomel, camphor, cholesterol, ergot fluid extract, hexachlorophene, 
ichthammol, isobutamben, juniper tar (oil of cade), lanolin, magnesium 
sulfate, menthol, methyl salicylate, oxyguinoline sulfate, petrolatum, 
phenol, pine tar, rosin, rosin cerate, sassafras oil, sulfur, thymol, 
triclosan, and zinc oxide have been present in OTC boil treatment drug 
products. There is a lack of adequate data to establish general 
recognition of the safety and effectiveness of these or any other 
ingredient for OTC use for the treatment of boils. Treatment is defined 
as reducing the size of a boil or reducing an infection related to a 
boil. Treatment has involved the use of ``drawing salves'' for

[[Page 45]]

these purposes. These ``drawing salves'' contained various ingredients. 
Based on evidence currently available, any OTC drug product offered for 
the treatment of boils cannot be considered generally recognized as safe 
and effective.
    (b) Any OTC drug product that is labeled, represented, or promoted 
for the treatment of boils is regarded as a new drug within the meaning 
of section 201(p) of the Federal Food, Drug, and Cosmetic Act (the act), 
for which an approved application or abbreviated application under 
section 505 of the act and part 314 of this chapter is required for 
marketing. In the absence of an approved new drug application or 
abbreviated new drug application, such product is also misbranded under 
section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any OTC 
boil treatment drug product is safe and effective for the purpose 
intended must comply with the requirements and procedures governing the 
use of investigational new drugs set forth in part 312 of this chapter.
    (d) After May 7, 1991, any such OTC drug product that contains 
aminacrine hydrochloride, bismuth subnitrate, calomel, camphor, 
cholesterol, ergot fluid extract, hexachlorophene, isobutamben, juniper 
tar (oil of cade), lanolin, magnesium sulfate, menthol, methyl 
salicylate, oxyguinoline sulfate, petrolatum, phenol, pine tar, rosin, 
rosin cerate, sassafras oil, thymol, or zinc oxide initially introduced 
or initially delivered for introduction into interstate commerce that is 
not in compliance with this section is subject to regulatory action.
    (e) After May 16, 1994, any such OTC drug product that contains 
benzocaine, ichthammol, sulfur, or triclosan initially introduced or 
initially delivered for introduction into interstate commerce that is 
not in compliance with this section is subject to regulatory action.
    (f) This section does not apply to drug products that contain 
benzocaine labeled, represented, or promoted for OTC topical use in 
accordance with part 348 of this chapter.
[58 FR 60336, Nov. 15, 1993]



Sec. 310.532  Drug products containing active ingredients offered over-the-counter (OTC) to relieve the symptoms of benign prostatic hypertrophy.

    (a) The amino acids glycine, alanine, and glutamic acid (alone or in 
combination) and the ingredient sabal have been present in over-the-
counter (OTC) drug products to relieve the symptoms of benign prostatic 
hypertrophy, e.g., urinary urgency and frequency, excessive urinating at 
night, and delayed urination. There is a lack of adequate data to 
establish general recognition of the safety and effectiveness of these 
or any other ingredients for OTC use in relieving the symptoms of benign 
prostatic hypertrophy. In addition, there is no definitive evidence that 
any drug product offered for the relief of the symptoms of benign 
prostatic hypertrophy would alter the obstructive or inflammatory signs 
and symptoms of this condition. Therefore, self-medication with OTC drug 
products might unnecessarily delay diagnosis and treatment of 
progressive obstruction and secondary infections. Based on evidence 
currently available, any OTC drug product containing ingredients offered 
for use in relieving the symptoms of benign prostatic hypertrophy cannot 
be generally recognized as safe and effective.
    (b) Any OTC drug product that is labeled, represented, or promoted 
to relieve the symptoms of benign prostatic hypertrophy is regarded as a 
new drug within the meaning of section 201(p) of the Federal Food, Drug, 
and Cosmetic Act (the act), for which an approved application under 
section 505 of the act and part 314 of this chapter is required for 
marketing. In the absence of an approved application, such product is 
also misbranded under section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for OTC use to relieve 
the symptoms of benign prostatic hypertrophy is safe and effective for 
the purpose intended must comply with the requirements and procedures 
governing the use of investigational new drugs set forth in part 312 of 
this chapter.

[[Page 46]]

    (d) After August 27, 1990, any such OTC drug product initially 
introduced or initially delivered for introduction into interstate 
commerce that is not in compliance with this section is subject to 
regulatory action.
[55 FR 6930, Feb. 27, 1990]



Sec. 310.533  Drug products containing active ingredients offered over-the-counter (OTC) for human use as an anticholinergic in cough-cold drug products.

    (a) Atropine sulfate, belladonna alkaloids, and belladonna alkaloids 
as contained in Atropa belladonna and Datura stramonium have been 
present as ingredients in cough-cold drug products for use as an 
anticholinergic. Anticholinergic drugs have been marketed OTC in cough-
cold drug products to relieve excessive secretions of the nose and eyes, 
symptoms that are commonly associated with hay fever, allergy, rhinitis, 
and the common cold. Atropine sulfate for oral use as an anticholinergic 
is probably safe at dosages that have been used in marketed cough-cold 
products (0.2 to 0.3 milligram); however, there are inadequate data to 
establish general recognition of the effectiveness of this ingredient. 
The belladonna alkaloids, which contain atropine (d, dl hyoscyamine) and 
scopolamine (l- hyoscine), are probably safe for oral use at dosages 
that have been used in marketed cough-cold products (0.2 milligram) but 
there are inadequate data to establish general recognition of the 
effectiveness of these ingredients as an anticholinergic for cough-cold 
use. Belladonna alkaloids for inhalation use, as contained in Atropa 
belladonna and Datura stramonium, are neither safe nor effective as an 
OTC anticholinergic. There are inadequate safety and effectiveness data 
to establish general recognition of the safety and/or effectiveness or 
any of these ingredients, or any other ingredient, for OTC use as an 
anticholinergic in cough-cold drug products.
    (b) Any OTC cough-cold drug product that is labeled, represented, or 
promoted for use as an anticholinergic is regarded as a new drug within 
the meaning of section 201(p) of the Federal Food, Drug, and Cosmetic 
Act, for which an approved new drug application under section 505 of the 
act and part 314 of this chapter is required for marketing. In the 
absence of an approved new drug application, such product is also 
misbranded under section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
cough-cold drug product labeled, represented, or promoted for OTC use as 
an anticholinergic is safe and effective for the purpose intended must 
comply with the requirements and procedures governing the use of 
investigational new drugs set forth in part 312 of this chapter.
    (d) After the effective date of the final regulation, any such OTC 
cough-cold drug product that is labeled, represented, or promoted for 
use as an anticholinergic may not be initially introduced or initially 
delivered for introduction into interstate commerce unless it is the 
subject of an approved new drug application.
[50 FR 46587, Nov. 8, 1985, as amended at 55 FR 11579, Mar. 29, 1990]



Sec. 310.534  Drug products containing active ingredients offered over-the-counter (OTC) for human use as oral wound healing agents.

    (a) Allantoin, carbamide peroxide in anhydrous glycerin, water 
soluble chlorophyllins, and hydrogen peroxide in aqueous solution have 
been present in oral mucosal injury drug products for use as oral wound 
healing agents. Oral wound healing agents have been marketed as aids in 
the healing of minor oral wounds by means other than cleansing and 
irrigating, or by serving as a protectant. Allantoin, carbamide peroxide 
in anhydrous glycerin, water soluble chlorophyllins, and hydrogen 
peroxide in aqueous solution are safe for use as oral wound healing 
agents, but there are inadequate data to establish general recognition 
of the effectiveness of these ingredients as oral wound healing agents.
    (b) Any OTC drug product that is labeled, represented, or promoted 
for use as an oral wound healing agent is regarded as a new drug within 
the meaning of section 201(p) of the Federal Food, Drug, and Cosmetic 
Act, for which an approved new drug application under section 505 of the 
act and part 314 of this chapter is required for

[[Page 47]]

marketing. In the absence of an approved new drug application, such 
product is also misbranded under section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for OTC use as an oral 
wound healing agent is safe and effective for the purpose intended must 
comply with the requirements and procedures governing the use of 
investigational new drugs set forth in part 312 of this chapter.
    (d) After the effective date of the final regulation, any OTC drug 
product that is labeled, represented, or promoted for use as an oral 
wound healing agent may not be initially introduced or initially 
delivered for introduction into interstate commerce unless it is the 
subject of an approved new drug application.
[51 FR 26114, July 18, 1986, as amended at 55 FR 11579, Mar. 29, 1990]



Sec. 310.536  Drug products containing active ingredients offered over-the-counter (OTC) for use as a nailbiting or thumbsucking deterrent.

    (a) Denatonium benzoate and sucrose octaacetate have been present in 
OTC nailbiting and thumbsucking deterrent drug products. There is a lack 
of adequate data to establish general recognition of the safety and 
effectiveness of these and any other ingredients (e.g., cayenne pepper) 
for OTC use as a nailbiting or thumbsucking deterrent. Based on evidence 
currently available, any OTC drug product containing ingredients offered 
for use as a nailbiting or thumbsucking deterrent cannot be generally 
recognized as safe and effective.
    (b) Any OTC drug product that is labeled, represented, and promoted 
as a nailbiting or thumbsucking deterrent is regarded as a new drug 
within the meaning of section 201(p) of the Federal Food, Drug, and 
Cosmetic Act (the act) for which an approved application or abbreviated 
application under section 505 of the act and part 314 of this chapter is 
required for marketing. In the absence of an approved new drug 
application or abbreviated new drug application, such product is also 
misbranded under section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for OTC use as a 
nailbiting or thumbsucking deterrent is safe and effective for the 
purpose intended must comply with the requirements and procedures 
governing the use of investigational new drugs set forth in part 312 of 
this chapter.
    (d) After March 2, 1994, any such OTC drug product initially 
introduced or initially delivered for introduction into interstate 
commerce that is not in compliance with this section is subject to 
regulatory action.
[58 FR 46754, Sept. 2, 1993]



Sec. 310.537  Drug products containing active ingredients offered over-the-counter (OTC) for oral administration for the treatment of fever blisters and cold 
          sores.

    (a) l-lysine (lysine, lysine hydrochloride), Lactobacillus 
acidophilus, and Lactobacillus bulgaricus have been present in orally 
administered OTC drug products to treat fever blisters and cold sores. 
There is a lack of adequate data to establish general recognition of the 
safety and effectiveness of these or any other orally administered 
ingredients for OTC use to treat or relieve the symptoms or discomfort 
of fever blisters and cold sores. Based on evidence currently available, 
any OTC drug product for oral administration containing ingredients 
offered for use in treating or relieving the symptoms or discomfort of 
fever blisters and cold sores cannot be generally recognized as safe and 
effective.
    (b) Any OTC drug product for oral administration that is labeled, 
represented, or promoted to treat or relieve the symptoms or discomfort 
of fever blisters and cold sores is regarded as a new drug within the 
meaning of section 201(p) of the Federal Food, Drug, and Cosmetic Act 
(the act), for which an approved application under section 505 of the 
act and part 314 of this chapter is required for marketing. In the 
absence of an approved application, such product is also misbranded 
under section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product for oral administration labeled,

[[Page 48]]

represented, or promoted for OTC use to treat or relieve the symptoms or 
discomfort of fever blisters and cold sores is safe and effective for 
the purpose intended must comply with the requirements and procedures 
governing the use of investigational new drugs set forth in part 312 of 
this chapter.
    (d) After December 30, 1992, any such OTC drug product initially 
introduced or initially delivered for introduction into interstate 
commerce that is not in compliance with this section is subject to 
regulatory action.
[57 FR 29173, June 30, 1992]



Sec. 310.538  Drug products containing active ingredients offered over-the-counter (OTC) for use for ingrown toenail relief.

    (a) Any product that bears labeling claims such as for ``temporary 
relief of discomfort from ingrown toenails,'' or ``ingrown toenail 
relief product,'' or ``ingrown toenail reliever,'' or similar claims is 
considered an ingrown toenail relief drug product. Benzocaine, 
chlorobutanol, chloroxylenol, dibucaine, sodium sulfide, tannic acid, 
and urea have been present as ingredients in such products. There is 
lack of adequate data to establish general recognition of the safety and 
effectiveness of these or any other ingredients for OTC use for ingrown 
toenail relief. Based on evidence currently available, any OTC drug 
product containing ingredients offered for use for ingrown toenail 
relief cannot be generally recognized as safe and effective.
    (b) Any OTC drug product that is labeled, represented, or promoted 
for ingrown toenail relief is regarded as a new drug within the meaning 
of section 201(p) of the Federal Food, Drug, and Cosmetic Act (the act), 
for which an approved application or abbreviated application under 
section 505 of the act and part 314 of this chapter is required for 
marketing. In the absence of an approved new drug application or 
abbreviated new drug application, such product is also misbranded under 
section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for OTC use for ingrown 
toenail relief is safe and effective for the purpose intended must 
comply with the requirements and procedures governing the use of 
investigational new drugs set forth in part 312 of this chapter.
    (d) After March 9, 1994, any such OTC drug product initially 
introduced or initially delivered for introduction into interstate 
commerce that is not in compliance with this section is subject to 
regulatory action.
[58 FR 47605, Sept. 9, 1993]



Sec. 310.540  Drug products containing active ingredients offered over-the-counter (OTC) for use as stomach acidifiers.

    (a) Betaine hydrochloride, glutamic acid hydrochloride, diluted 
hydrochloric acid, and pepsin have been present as ingredients in over-
the-counter (OTC) drug products for use as stomach acidifiers. Because 
of the lack of adequate data to establish the effectiveness of these or 
any other ingredients for use in treating achlorhydria and 
hypochlorhydria, and because such conditions are asymptomatic, any OTC 
drug product containing ingredients offered for use as a stomach 
acidifier cannot be considered generally recognized as safe and 
effective.
    (b) Any OTC drug product that is labeled, represented, or promoted 
for use as a stomach acidifier is regarded as a new drug within the 
meaning of section 201(p) of the Federal Food, Drug, and Cosmetic Act, 
for which an approved new drug application under section 505 of the act 
and part 314 of this chapter is required for marketing. In the absence 
of an approved new drug application, such product is also misbranded 
under section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted as a stomach acidifier 
for OTC use is safe and effective for the purpose intended must comply 
with the requirements and procedures governing the use of 
investigational new drugs set forth in part 312 of this chapter.
    (d) After the effective date of the final regulation, any such OTC 
drug product initially introduced or initially delivered for 
introduction into interstate commerce that is not in

[[Page 49]]

compliance with this section is subject to regulatory action.
[53 FR 31271, Aug. 17, 1988]



Sec. 310.541  Over-the-counter (OTC) drug products containing active ingredients offered for use in the treatment of hypophosphatemia.

    (a) Hypophosphatemia is a condition in which an abnormally low 
plasma level of phosphate occurs in the blood. This condition is not 
amenable to self-diagnosis or self-treatment. Treatment of this 
condition should be restricted to the supervision of a physician. For 
this reason, any drug product containing ingredients offered for OTC use 
in the treatment of hypophosphatemia cannot be considered generally 
recognized as safe and effective.
    (b) Any drug product that is labeled, represented, or promoted for 
OTC use in the treatment of hypophosphatemia is regarded as a new drug 
within the meaning of section 201(p) of the Federal Food, Drug, and 
Cosmetic Act (the act), for which an approved application under section 
505 of the act and part 314 of this chapter is required for marketing. 
In the absence of an approved application, such product is also 
misbranded under section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for OTC use in the 
treatment of hypophosphatemia is safe and effective for the purpose 
intended must comply with the requirements and procedures governing the 
use of investigational new drugs set forth in part 312 of his chapter.
    (d) After November 12, 1990, any such OTC drug product initially 
introduced or initially delivered for introduction into interstate 
commerce that is not in compliance with this section is subject to 
regulatory action.
[55 FR 19858, May 11, 1990]



Sec. 310.542  Over-the-counter (OTC) drug products containing active ingredients offered for use in the treatment of hyperphosphatemia.

    (a) Hyperphosphatemia is a condition in which an abnormally high 
plasma level of phosphate occurs in the blood. This condition in not 
amenable to self-diagnosis or self-treatment. Treatment of this 
condition should be restricted to the supervision of a physician. For 
this reason, any drug product containing ingredients offered for OTC use 
in the treatment of hyperphosphatemia cannot be considered generally 
recognized as safe and effective.
    (b) Any drug product that is labeled, represented, or promoted for 
OTC use in the treatment of hyperphosphatemia is regarded as a new drug 
within the meaning of section 201(p) of the Federal Food, Drug, and 
Cosmetic Act (the act), for which an approved application under section 
505 of the act and part 314 of this chapter is required for marketing. 
In the absence of an approved application, such product is also 
misbranded under section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for use in the treatment 
of hyperphosphatemia is safe and effective for the purpose intended must 
comply with the requirements and procedures governing use of 
investigational new drugs set forth in part 312 of this chapter.
    (d) After November 12, 1990, any such OTC drug product initially 
introduced or initially delivered for introduction into interstate 
commerce that is not in compliance with this section is subject to 
regulatory action.
[55 FR 19858, May 11, 1990]



Sec. 310.543  Drug products containing active ingredients offered over-the-counter (OTC) for human use in exocrine pancreatic insufficiency.

    (a) Hemicellulase, pancreatin, and pancrelipase have been present as 
ingredients in exocrine pancreatic insufficiency drug products. 
Pancreatin and pancrelipase are composed of enzymes: amylase, trypsin 
(protease), and lipase. Significant differences have been shown in the 
bioavailability of marketed exocrine pancreatic insufficiency drug 
products produced by different manufacturers. These differences raise a 
potential for serious risk to patients using these drug products. The 
bioavailability of pancreatic enzymes is dependent on the process used 
to manufacture the drug products. Information on this process is not 
included in an OTC drug monograph. Therefore,

[[Page 50]]

the safe and effective use of these enzymes for treating exocrine 
pancreatic insufficiency cannot be regulated adequately by an OTC drug 
monograph. Information on the product's formulation, manufacture, 
quality control procedures, and final formulation effectiveness testing 
are necessary in an approved application to ensure that a company has 
the ability to manufacture a proper bioactive formulation. In addition, 
continuous physician monitoring of patients who take these drug products 
is a collateral measure necessary to the safe and effective use of these 
enzymes, causing such products to be available by prescription only.
    (b) Any drug product that is labeled, represented, or promoted for 
OTC use in the treatment of exocrine pancreatic insufficiency is 
regarded as a new drug within the meaning of section 201(p) of the 
Federal Food, Drug, and Cosmetic Act (the act), for which an approved 
application under section 505 of the act and part 314 of this chapter is 
required for marketing. In the absence of an approved application, such 
product is also misbranded under section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for OTC use in the 
treatment of exocrine pancreatic insufficiency is safe and effective for 
the purpose intended must comply with the requirements and procedures 
governing the use of investigational new drugs set forth in part 312 of 
this chapter.
    (d) After May 7, 1991, any such OTC drug product that contains 
hemicellulase initially introduced or initially delivered for 
introduction into interstate commerce that is not in compliance with 
this section is subject to regulatory action.
    (e) After October 24, 1995, any such OTC drug product that contains 
pancreatin or pancrelipase initially introduced or initially delivered 
for introduction into interstate commerce that is not in compliance with 
this section is subject to regulatory action.
[60 FR 20165, Apr. 24, 1995]



Sec. 310.544  Drug products containing active ingredients offered over-the-counter (OTC) for use as a smoking deterrent.

    (a) Any product that bears labeling claims that it ``helps stop or 
reduce the cigarette urge,'' ``helps break the cigarette habit,'' 
``helps stop or reduce smoking,'' or similar claims is a smoking 
deterrent drug product. Cloves, coriander, eucalyptus oil, ginger 
(Jamaica), lemon oil (terpeneless), licorice root extract, lobeline (in 
the form of lobeline sulfate or natural lobelia alkaloids or Lobelia 
inflata herb), menthol, methyl salicylate, povidone-silver nitrate, 
quinine ascorbate, silver acetate, silver nitrate, and thymol have been 
present as ingredients in such drug products. There is a lack of 
adequate data to establish general recognition of the safety and 
effectiveness of these or any other ingredients for OTC use as a smoking 
deterrent. Based on evidence currently available, any OTC drug product 
containing ingredients offered for use as a smoking deterrent cannot be 
generally recognized as safe and effective.
    (b) Any OTC drug product that is labeled, represented, or promoted 
as a smoking deterrent is regarded as a new drug within the meaning of 
section 201(p) of the Federal Food, Drug, and Cosmetic Act (the act), 
for which an approved application or abbreviated application under 
section 505 of the act and part 314 of this chapter is required for 
marketing. In the absence of an approved new drug application or 
abbreviated new drug application, such product is also misbranded under 
section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for OTC use as a smoking 
deterrent is safe and effective for the purpose intended must comply 
with the requirements and procedures governing the use of 
investigational new drugs set forth in part 312 of this chapter.
    (d) After May 7, 1991, any such OTC drug product containing cloves, 
coriander, eucalyptus oil, ginger (Jamaica), lemon oil (terpeneless), 
licorice root extract, menthol, methyl salicylate, quinine ascorbate, 
silver nitrate, and/or thymol initially introduced or

[[Page 51]]

initially delivered for introduction into interstate commerce that is 
not in compliance with this section is subject to regulatory action. 
After December 1, 1993, any such OTC drug product containing lobeline 
(in the form of lobeline sulfate or natural lobelia alkaloids or Lobelia 
inflata herb), povidone-silver nitrate, silver acetate, or any other 
ingredients initially introduced or initially delivered for introduction 
into interstate commerce that is not in compliance with this section is 
subject to regulatory action.
[58 FR 31241, June 1, 1993]



Sec. 310.545  Drug products containing certain active ingredients offered over-the-counter (OTC) for certain uses.

    (a) A number of active ingredients have been present in OTC drug 
products for various uses, as described below. However, based on 
evidence currently available, there are inadequate data to establish 
general recognition of the safety and effectiveness of these ingredients 
for the specified uses:
    (1) Topical acne drug products.

Alcloxa
Alkyl isoquinolinium bromide
Aluminum chlorohydrex
Aluminum hydroxide
Benzocaine
Benzoic acid
Boric acid
Calcium polysulfide
Calcium thiosulfate
Camphor
Chloroxylenol
Cloxyquin
Coal tar
Dibenzothiophene
Estrone
Magnesium aluminum silicate
Magnesium sulfate
Phenol
Phenolate sodium
Phenyl salicylate
Povidone-iodine
Pyrilamine maleate
Resorcinol (as single ingredient)
Resorcinol monoacetate (as single ingredient)
Salicylic acid (over 2 up to 5 percent)
Sodium borate
Sodium thiosulfate
Tetracaine hydrochloride
Thymol
Vitamin E
Zinc oxide
Zinc stearate
Zinc sulfide

    (2) Anticaries drug products--(i) Approved as of May 7, 1991.

Hydrogen fluoride
Sodium carbonate
Sodium monofluorophosphate (6 percent rinse)
Sodium phosphate

    (ii) Approved as of October 7, 1996.

Calcium sucrose phosphate
Dicalcium phosphate dihydrate
Disodium hydrogen phosphate\1\
---------------------------------------------------------------------------

    \1\ These ingredients are nonmonograph except when used to prepare 
acidulated phosphate fluoride treatment rinses identified in 
Sec. 355.10(a)(3) of this chapter.
---------------------------------------------------------------------------

Phosphoric acid1
Sodium dihydrogen phosphate
Sodium dihydrogen phosphate monohydrate
Sodium phosphate, dibasic anhydrous reagent1

    (3) Antidiarrheal drug products.

Aluminum hydroxide
Atropine sulfate
Calcium carbonate
Carboxymethylcellulose sodium
Glycine
Homatropine methylbromide
Hyoscyamine sulfate
Lactobacillus acidophilus
Lactobacillus bulgaricus
Opium, powdered
Opium tincture
Paregoric
Phenyl salicylate
Scopolamine hydrobromide
Zinc phenolsulfonate

    (4) Antiperspirant drug products.

Alum, potassium
Aluminum bromohydrate
Aluminum chloride (alcoholic solutions)
Aluminum chloride (aqueous solution) (aerosol only)
Aluminum sulfate
Aluminum sulfate, buffered (aerosol only)
Sodium aluminum chlorohydroxy lactate

    (5) [Reserved]
    (6) Cold, cough, allergy, bronchodilator, and antiasthmatic drug 
products--(i) Antihistamine drug products--(A) Ingredients.

Methapyrilene hydrochloride
Methapyrilene fumarate
Thenyldiamine hydrochloride

    (B) Ingredients.

Phenyltoloxamine dihydrogen citrate
Methapyrilene hydrochloride

[[Page 52]]

Methapyrilene fumarate
Thenyldiamine hydrochloride

    (ii) Nasal decongestant drug products--(A) Approved as of May 7, 
1991.

Allyl isothiocyanate
Camphor (lozenge)
Creosote, beechwood (oral)
Eucalyptol (lozenge)
Eucalyptol (mouthwash)
Eucalyptus oil (lozenge)
Eucalyptus oil (mouthwash)
Menthol (mouthwash)
Peppermint oil (mouthwash)
Thenyldiamine hydrochloride
Thymol
Thymol (lozenge)
Thymol (mouthwash)
Turpentine oil

    (B) Approved as of August 23, 1995.

Bornyl acetate (topical)
Cedar leaf oil (topical)
Creosote, beechwood (topical)
l-desoxyephedrine (topical)
Ephedrine (oral)
Ephedrine hydrochloride (oral)
Ephedrine sulfate (oral)
Racephedrine hydrochloride (oral/topical)

    (iii) Expectorant drug products.

Ammonium chloride
Antimony potassium tartrate
Beechwood creosote
Benzoin preparations (compound tincture of benzoin, tincture of benzoin)
Camphor
Chloroform
Eucalyptol/eucalyptus oil
Horehound
Iodides (calcium iodide anyhydrous, hydroidic acid syrup, iodized lime, 
potassium iodide)
Ipecac
Ipecac fluidextract
Ipecac syrup
Menthol/peppermint oil
Pine tar preparations (extract white pine compound, pine tar, syrup of 
pine tar, compound white pine syrup, white pine)
Potassium guaiacolsulfonate
Sodium citrate
Squill preparations (squill, squill extract)
Terpin hydrate preparations (terpin hydrate, terpin hydrate elixir)
Tolu preparations (tolu, tolu balsam, tolu balsam tincture)
Turpentine oil (spirits of turpentine)

    (iv) Bronchodilator drug products--(A) Approved as of October 2, 
1987.

Aminophylline
Belladonna alkaloids
Euphorbia pilulifera
Metaproterenol sulfate
Methoxyphenamine hydrochloride
Pseudoephedrine hydrochloride
Pseudoephedrine sulfate
Theophylline, anhydrous
Theophylline calcium salicylate
Theophylline sodium glycinate

    (B) Approved as of January 29, 1996. Any combination drug product 
containing theophylline (e.g., theophylline and ephedrine, or 
theophylline and ephedrine and phenobarbital).
     (C) Approved as of June 19, 1996. Any ingredient(s) in a 
pressurized metered-dose inhaler container.
    (7) Dandruff/seborrheic dermatitis/psoriasis drug products.

Alkyl isoquinolinium bromide
Allantoin
Benzalkonium chloride
Benzethonium chloride
Boric acid
Calcium undecylenate
Captan
Chloroxylenol
Colloidal oatmeal
Cresol, saponated
Ethohexadiol
Eucalyptol
Juniper tar
Lauryl isoquinolinium bromide
Menthol
Mercury oleate
Methylbenzethonium chloride
Methyl salicylate
Phenol
Phenolate sodium
Pine tar
Povidone-iodine
Resorcinol
Sodium borate
Sodium salicylate
Thymol
Undecylenic acid

    (8) Digestive aid drug products--(i) Approved as of May 7, 1991.

Bismuth sodium tartrate
Calcium carbonate
Cellulase
Dehydrocholic acid
Dihydroxyaluminum sodium carbonate
Duodenal substance
Garlic, dehydrated
Glutamic acid hydrochloride
Hemicellulase
Homatropine methylbromide
Magnesium hydroxide
Magnesium trisilicate
Ox bile extract
Pancreatin
Pancrelipase
Papain
Peppermint oil
Pepsin
Sodium bicarbonate

[[Page 53]]

Sodium citrate
Sorbitol

    (ii) Approved as of November 10, 1993.

Alcohol
Aluminum hydroxide
Amylase
Anise seed
Aromatic powder
Asafetida
Aspergillus oryza enzymes (except lactase enzyme derived from 
Aspergillus oryzae)
Bacillus acidophilus
Bean
Belladonna alkaloids
Belladonna leaves, powdered extract
Betaine hydrochloride
Bismuth subcarbonate
Bismuth subgallate
Black radish powder
Blessed thistle (cnicus benedictus)
Buckthorn
Calcium gluconate
Capsicum
Capsicum, fluid extract of
Carbon
Cascara sagrada extract
Catechu, tincture
Catnip
Chamomile flowers
Charcoal, wood
Chloroform
Cinnamon oil
Cinnamon tincture
Citrus pectin
Diastase
Diastase malt
Dog grass
Elecampane
Ether
Fennel acid
Galega
Ginger
Glycine
Hydrastis canadensis (golden seal)
Hectorite
Horsetail
Huckleberry
Hydrastis fluid extract
Hydrochloric acid
Iodine
Iron ox bile
Johnswort
Juniper
Kaolin, colloidal
Knotgrass
Lactic acid
Lactose
Lavender compound, tincture of
Linden
Lipase
Lysine hydrochloride
Mannitol
Mycozyme
Myrrh, fluid extract of
Nettle
Nickel-pectin
Nux vomica extract
Orthophosphoric acid
Papaya, natural
Pectin
Peppermint
Peppermint spirit
Phenacetin
Potassium bicarbonate
Potassium carbonate
Protease
Prolase
Rhubarb fluid extract
Senna
Sodium chloride
Sodium salicylate
Stem bromelain
Strawberry
Strychnine
Tannic acid
Trillium
Woodruff

    (iii) Charcoal, activated
    (9) [Reserved]
    (10) External analgesic drug products--(i) Analgesic and anesthetic 
drug products.

Aspirin
Chloral hydrate
Chlorobutanol
Cyclomethycaine sulfate
Eugenol
Hexylresorcinol
Methapyrilene hydrochloride
Salicylamide
Thymol

    (ii) Counterirritant drug products.

Chloral hydrate
Eucalyptus oil

    (iii) Male genital desensitizer drug products.

Benzyl alcohol
Camphorated metacresol
Ephedrine hydrochloride

    (iv) Diaper rash drug products.
    Any ingredient(s) labeled with claims or directions for use in the 
treatment and/or prevention of diaper rash.
    (v) Fever blister and cold sore treatment drug products.

Allyl isothiocyanate
Aspirin
Bismuth sodium tartrate
Camphor (exceeding 3 percent)
Capsaicin
Capsicum
Capsicum oleoresin
Chloral hydrate
Chlorobutanol
Cyclomethycaine sulfate
Eucalyptus oil
Eugenol

[[Page 54]]

Glycol salicylate
Hexylresorcinol
Histamine dihydrochloride
Menthol (exceeding 1 percent)
Methapyrilene hydrochloride
Methyl nicotinate
Methyl salicylate
Pectin
Salicylamide
Strong ammonia solution
Tannic acid
Thymol
Tripelennamine hydrochloride
Trolamine salicylate
Turpentine oil
Zinc sulfate

    (vi) Insect bite and sting drug products.

Alcohol
Alcohol, ethoxylated alkyl
Benzalkonium chloride
Calamine
Ergot fluidextract
Ferric chloride
Panthenol
Peppermint oil
Pyrilamine maleate
Sodium borate
Trolamine salicylate
Turpentine oil
Zinc oxide
Zirconium oxide

    (vii) Poison ivy, poison oak, and poison sumac drug products.

Alcohol
Aspirin
Benzethonium chloride
Benzocaine (0.5 to 1.25 percent)
Bithionol
Calamine
Cetalkonium chloride
Chloral hydrate
Chlorobutanol
Chlorpheniramine maleate
Creosote, beechwood
Cyclomethycaine sulfate
Dexpanthenol
Diperodon hydrochloride
Eucalyptus oil
Eugenol
Glycerin
Glycol salicylate
Hectorite
Hexylresorcinol
Hydrogen peroxide
Impatiens biflora tincture
Iron oxide
Isopropyl alcohol
Lanolin
Lead acetate
Merbromin
Mercuric chloride
Methapyrilene hydrochloride
Panthenol
Parethoxycaine hydrochloride
Phenyltoloxamine dihydrogen citrate
Povidone-vinylacetate copolymers
Pyrilamine maleate
Salicylamide
Salicylic acid
Simethicone
Sulfur
Tannic acid
Thymol
Trolamine salicylate
Turpentine oil
Zirconium oxide
Zyloxin

    (11) [Reserved]
    (12) Laxative drug products--(i) Bulk laxatives.

Agar
Carrageenan (degraded)
Carrageenan (native)
Guar gun

    (ii) Saline laxative.

Tartaric acid

    (iii) Stool softener.

Poloxamer 188

    (iv) Stimulant laxatives.

Aloin
Bile salts/acids
Calcium pantothenate
Calomel
Colocynth
Elaterin resin
Frangula
Gamboge
Ipomea
Jalap
Ox bile
Podophyllum resin
Prune concentrate dehydrate
Prune powder
Rhubarb, Chinese
Sodium Oleate

    (13) [Reserved]
    (14) Oral health care drug products (nonantimicrobial).

Antipyrine
Camphor
Cresol
Dibucaine
Dibucaine hydrochloride
Eucalyptol
Lidocaine
Lidocaine hydrochloride
Methly salicylate
Myrrh tincture
Pyrilamine maleate
Sorbitol
Sugars
Tetracaine
Tetracaine hydrochloride
Thymol


[[Page 55]]


    (15) Topical otic drug products for the prevention of swimmer's ear 
and for the drying of water-clogged ears--(i) Approved as of May 7, 
1991.

Acetic acid

    (ii) Approved as of August 15, 1995.

Glycerin and anhydrous glycerin
Isopropyl alcohol

    (16) Poison treatment drug products.

Ipecac fluidextract
Ipecac tincture
Zinc sulfate

    (17) Skin bleaching drug products.

Mercury, ammoniated

    (18) Skin protectant drug products. (i) Ingredients.

Allantoin (wound healing claims only)
Sulfur
Tannic acid
Zinc acetate (wound healing claims only)

    (ii) Astringent drug products.

Acetone
Alcohol
Alum, ammonium
Alum, potassium
Aluminum chlorhydroxy complex
Aromatics
Benzalkonium chloride
Benzethonium chloride
Benzocaine
Benzoic acid
Boric acid
Calcium acetate
Camphor gum
Clove oil
Colloidal oatmeal
Cresol
Cupric sulfate
Eucalyptus oil
Eugenol
Ferric subsulfate (Monsel's Solution)
Honey
Isopropyl alcohol
Menthol
Methyl salicylate
Oxyquinoline sulfate
P-t-butyl-m-cresol
Peppermint oil
Phenol
Polyoxeythylene laurate
Potassium ferrocyanide
Sage oil
Silver nitrate
Sodium borate
Sodium diacetate
Talc
Tannic acid glycerite
Thymol
Topical starch
Zinc chloride
Zinc oxide
Zinc phenolsulfonate
Zinc stearate
Zinc sulfate

    (iii) Diaper rash drug products.

Aluminum hydroxide
Cocoa butter
Cysteine hydrochloride
Glycerin
Protein hydrolysate
Racemethionine
Sulfur
Tannic acid
Zinc acetate
Zinc carbonate

    (iv) Fever blister and cold sore treatment drug products.

Bismuth subnitrate
Boric acid
Pyridoxine hydrochloride
Sulfur
Tannic acid
Topical starch
Trolamine
Zinc sulfate

    (v) Insect bite and sting drug products.

Alcohol
Alcohol, ethoxylated alkyl
Ammonia solution, strong
Ammonium hydroxide
Benzalkonium chloride
Camphor
Ergot fluidextract
Ferric chloride
Menthol
Peppermint oil
Phenol
Pyrilamine maleate
Sodium borate
Trolamine
Turpentine oil
Zirconium oxide

    (vi) Poison ivy, poison oak, and poison sumac drug products.

Alcohol
Anion and cation exchange resins buffered
Benzethonium chloride
Benzocaine
Benzyl alcohol
Bismuth subnitrate
Bithionol
Boric acid
Camphor
Cetalkonium chloride
Chloral hydrate
Chlorpheniramine maleate
Creosote
Diperodon hydrochloride
Diphenhydramine hydrochloride
Eucalyptus oil
Ferric chloride

[[Page 56]]

Glycerin
Hectorite
Hydrogen peroxide
Impatiens biflora tincture
Iron oxide
Isopropyl alcohol
Lanolin
Lead acetate
Lidocaine
Menthol
Merbromin
Mercuric chloride
Panthenol
Parethoxycaine hydrochloride
Phenol
Phenyltoloxamine dihydrogen citrate
Povidone-vinylacetate copolymers
Salicylic acid
Simethicone
Tannic acid
Topical starch
Trolamine
Turpentine oil
Zirconium oxide
Zyloxin

    (19) [Reserved]
    (20) Weight control drug products.

Alcohol
Alfalfa
Alginic acid
Anise oil
Arginine
Ascorbic acid
Bearberry
Biotin
Bone marrow, red
Buchu
Buchu, potassium extract
Caffeine
Caffeine citrate
Calcium
Calcium carbonate
Calcium caseinate
Calcium lactate
Calcium pantothenate
Carboxymethylcellulose sodium
Carrageenan
Cholecalcierol
Choline
Chondrus
Citric acid
Cnicus benedictus
Copper
Copper gluconate
Corn oil
Corn syrup
Corn silk, potassium extract
Cupric sulfate
Cyanocobalamin (vitamin B12)
Cystine
Dextrose
Docusate sodium
Ergocalciferol
Ferric ammonium citrate
Ferric pyrophosphate
Ferrous fumarate
Ferrous gluconate
Ferrous sulfate (iron)
Flax seed
Folic acid
Fructose
Guar gum
Histidine
Hydrastis canadensis
Inositol
Iodine
Isoleucine
Juniper, potassium extract
Karaya gum
Kelp
Lactose
Lecithin
Leucine
Liver concentrate
Lysine
Lysine hydrochloride
Magnesium
Magnesium oxide
Malt
Maltodextrin
Manganese citrate
Mannitol
Methionine
Methylcellulose
Mono- and di-glycerides
Niacinamide
Organic vegetables
Pancreatin
Pantothenic acid
Papain
Papaya enzymes
Pepsin
Phenacetin
Phenylalanine
Phosphorus
Phytolacca
Pineapple enzymes
Plantago seed
Potassium citrate
Pyridoxine hydrochloride (vitamin B6)
Riboflavin
Rice polishings
Saccharin
Sea minerals
Sesame seed
Sodium
Sodium bicarbonate
Sodium caseinate
Sodium chloride (salt)
Soybean protein
Soy meal
Sucrose
Thiamine hydrochloride (vitamin B1)
Thiamine mononitrate (vitamin B1 mononitrate)
Threonine
Tricalcium phosphate
Tryptophan
Tyrosine
Uva ursi, potassium extract
Valine
Vegetable

[[Page 57]]

Vitamin A
Vitamin A acetate
Vitamin A palmitate
Vitamin E
Wheat germ
Xanthan gum
Yeast

    (21) Ophthalmic drug products.
    (i) Ophthalmic anesthetic drug products.

Antipyrine
Piperocaine hydrochloride

    (ii) Ophthalmic anti-infective drug products.

Boric acid
Mild silver protein
Yellow mercuric oxide

    (iii) Ophthalmic astringent drug products.

Infusion of rose petals

    (iv) Ophthalmic demulcent drug products.

Polyethylene glycol 6000

    (v) Ophthalmic vasoconstrictor drug products.

Phenylephrine hydrochloride (less than 0.08 percent)

    (22) Topical antifungal drug products.
    (i) Diaper rash drug products. Any ingredient(s) labeled with claims 
or directions for use in the treatment and/or prevention of diaper rash.
    (ii) Ingredients.

Alcloxa
Alum, potassium
Aluminum sulfate
Amyltricresols, secondary
Basic fuchsin
Benzethonium chloride
Benzoic acid
Benzoxiquine
Boric acid
Camphor
Candicidin
Chlorothymol
Coal tar
Dichlorophen
Menthol
Methylparaben
Oxyquinoline
Oxyquinoline sulfate
Phenol
Phenolate sodium
Phenyl salicylate
Propionic acid
Propylparaben
Resorcinol
Salicylic acid
Sodium borate
Sodium caprylate
Sodium propionate
Sulfur
Tannic acid
Thymol
Tolindate
Triacetin
Zinc caprylate
Zinc propionate

    (iii) Any ingredient(s) labeled with claims or directions for use on 
the scalp or on the nails.
    (iv) Ingredients.

Camphorated metacresol
Chloroxylenol
m-cresol
Nystatin

    (23) Internal analgesic drug products.

Aminobenzoic acid
Antipyrine
Aspirin, aluminum
Calcium salicylate
Codeine
Codeine phosphate
Codeine sulfate
Iodoantipyrine
Lysine aspirin
Methapyrilene fumarate
Phenacetin
Pheniramine maleate
Pyrilamine maleate
Quinine
Salsalate
Sodium aminobenzoate

    (24) Orally administered menstrual drug products.

Alcohol
Alfalfa leaves
Aloes
Asclepias tuberosa
Asparagus
Barosma
Bearberry (extract of uva ursi)
Bearberry fluidextract (extract of bearberry)
Blessed thistle (cnicus benedictus)
Buchu powdered extract (extract of buchu)
Calcium lactate
Calcium pantothenate
Capsicum oleoresin
Cascara fluidextract, aromatic (extract of cascara)
Chlorprophenpyridamine maleate
Cimicifuga racemosa
Codeine
Collinsonia (extract stone root)
Corn silk
Couch grass
Dog grass extract
Ethyl nitrite
Ferric chloride
Ferrous sulfate
Gentiana lutea (gentian)

[[Page 58]]

Glycyrrhiza (licorice)
Homatropine methylbromide
Hydrangea, powdered extract (extract of hydrangea)
Hydrastis canadensis (golden seal)
Hyoscyamine sulfate
Juniper oil (oil of juniper)
Magnesium sulfate
Methapyrilene hydrochloride
Methenamine
Methylene blue
Natural estrogenic hormone
Niacinamide
Nutmeg oil (oil of nutmeg)
Oil of erigeron
Parsley
Peppermint spirit
Pepsin, essence
Phenacetin
Phenindamine tartrate
Phenyl salicylate
Piscidia erythrina
Pipsissewa
Potassium acetate
Potassium nitrate
Riboflavin
Saw palmetto
Senecio aureus
Sodium benzoate
Sodium nitrate
Sucrose
Sulferated oils of turpentine
Taraxacum officinale
Theobromine sodium salicylate
Theophylline
Thiamine hydrochloride
Triticum
Turpentine, venice (venice turpertine)
Urea

    (25) Pediculicide drug products--(i) Approved as of November 10, 
1993.

Benzocaine
Benzyl alcohol
Benzyl benzoate
Chlorophenothane (dichlorodiphenyl trichloroethane)
Coconut oil soap, aqueous
Copper oleate
Docusate sodium
Formic acid
Isobornyl thiocyanoacetate
Picrotoxin
Propylene glycol
Sabadilla alkaloids
Sulfur, sublimed
Thiocyanoacetate

    (ii) Approved as of June 14, 1994. The combination of pyrethrum 
extract (formerly named pyrethrins) and piperonyl butoxide in an aerosol 
dosage formulation.
    (26) Anorectal druq products--(i) Anticholinergic drug products.

Atropine
Belladonna extract

    (ii) Antiseptic drug products.

Boric acid
Boroglycerin
Hydrastis
Phenol
Resorcinol
Sodium salicylic acid phenolate

    (iii) Astringent drug products.

Tannic acid

    (iv) Counterirritant drug products.

Camphor (greater than 3 to 11 percent)
Hydrastis
Menthol (1.25 to 16 percent)
Turpentine oil (rectified) (6 to 50 percent)

    (v) Keratolytic drug products.

Precipitated sulfur
Sublimed sulfur

    (vi) Local anesthetic drug products.

Diperodon
Phenacaine hydrochloride

    (vii) Other druq products.

Collinsonia extract
Escherichia coli vaccines
Lappa extract
Leptandra extract
Live yeast cell derivative
Mullein

    (viii) Protectant druq products.

Bismuth oxide
Bismuth subcarbonate
Bismuth subgallate
Bismuth subnitrate
Lanolin alcohols

    (ix) Vasoconstrictor druq products.

Epinephrine undecylenate

    (x) Wound healinq druq products.

Cholecalciferol
Cod liver oil
Live yeast cell derivative
Peruvian balsam
Shark liver oil
Vitamin A

    (b) Any OTC drug product that is labeled, represented, or promoted 
for the uses specified and containing any active ingredient(s) as 
specified in paragraph (a) of this section is regarded as a new drug 
within the meaning of section 210(p) of the Federal Food, Drug, and 
Cosmetic Act (the Act), for which an approved new drug application under 
section 505 of the Act and part

[[Page 59]]

314 of this chapter is required for marketing. In the absence of an 
approved new drug application, such product is also misbranded under 
section 502 of the Act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for the OTC uses and 
containing any active ingredient(s) as specified in paragraph (a) of 
this section is safe and effective for the purpose intended must comply 
with the requirements and procedures governing the use of 
investigational new drugs set forth in part 312 of this chapter.
     (d) Any OTC drug product that is not in compliance with this 
section is subject to regulatory action if initially introduced or 
initially delivered for introduction into interstate commerce after the 
dates specified in paragraphs (d)(1) through (d)(25) of this section.
    (1) May 7, 1991, for products subject to paragraphs (a)(1) through 
(a)(2)(i), (a)(3) through (a)(4), (a)(6)(i)(A), (a)(6)(ii)(A), (a)(7) 
(except as covered by paragraph (d)(3) of this section), (a)(8)(i), 
(a)(9) through (a)(10)(iii), (a)(12)(i) through (a)(12)(iv), (a)(14) 
through (a)(15)(i), and (a)(16) through (a)(18)(i) of this section.
    (2) February 10, 1992, for products subject to paragraph (a)(20) of 
this section.
    (3) December 4, 1992, for products subject to paragraph (a)(7) of 
this section that contain menthol as an antipruritic in combination with 
the antidandruff ingredient coal tar identified in Sec. 358.710(a)(1) of 
this chapter.
    (4) February 28, 1990, for products subject to paragraph (a)(6)(iii) 
of this section, except those that contain ipecac.
    (5) September 14, 1993, for products subject to paragraph 
(a)(6)(iii) of this section that contain ipecac.
    (6) December 9, 1993, for products subject to paragraph (a)(6)(i)(B) 
of this section.
    (7) March 6, 1989, for products subject to paragraph (a)(21) of this 
section, except those that contain ophthalmic anti-infective ingredients 
listed in paragraph (a)(21)(ii).
    (8) June 18, 1993, for products subject to paragraph (a)(21) of this 
section that contain ophthalmic anti-infective ingredients.
    (9) June 18, 1993, for products subject to paragraph (a)(10)(iv) of 
this section.
    (10) June 18, 1993, for products subject to paragraph (a)(22)(i) of 
this section.
    (11) November 10, 1993, for products subject to paragraph 
(a)(18)(ii) of this section, except products that contain ferric 
subsulfate.
    (12) March 2, 1994, for products subject to paragraph (a)(22)(iii) 
of this section.
    (13) August 5, 1991, for products subject to paragraphs (a)(26) of 
this section, except for those that contain live yeast cell derivative.
    (14) September 2, 1994, for products subject to paragraph 
(a)(26)(vii) and (a)(26)(x) of this section that contain live yeast cell 
derivative.
    (15) September 23, 1994, for products subject to paragraph 
(a)(22)(iv) of this section.
    (16) June 14, 1994, for products subject to paragraph (a)(25)(ii) of 
this section.
    (17) [Reserved]
    (18) August 15, 1995, for products subject to paragraph (a)(15)(ii) 
of this section.
    (19) October 2, 1987, for products subject to paragraph 
(a)(6)(iv)(A) of this section.
    (20) January 29, 1996, for products subject to paragraph 
(a)(6)(iv)(B) of this section.
    (21) April 21, 1994, for products subject to paragraph (a)(8)(iii) 
of this section.
    (22) April 21, 1993, for products subject to paragraph (a)(18)(ii) 
of this section that contain ferric subsulfate.
    (23) August 23, 1995, for products subject to paragraph 
(a)(6)(ii)(B) of this section.
    (24) October 7, 1996, for products subject to paragraph (a)(2)(ii) 
of this section.
     (25) June 19, 1996, for products subject to paragraph (a)(6)(iv)(C) 
of this section.
[55 FR 46919, Nov. 7, 1990]

    Editorial Note: For Federal Register citations affecting 
Sec. 310.545, see the List of CFR Sections Affected in the Finding Aids 
section of this volume.

    Effective Date Notes:  1. At 60 FR 42436, Aug. 16, 1995, in 
Sec. 310.545, paragraph (a)(15)(ii)

[[Page 60]]

was stayed for topical otic drug products for the drying of water-
clogged ears.
    2. At 61 FR 9571, Mar. 8, 1996, in Sec. 310.545 in paragraph 
(a)(6)(ii)(B), the entry for ``l-desoxyephedrine (topical)'' was stayed 
until further notice.



Sec. 310.546  Drug products containing active ingredients offered over-the-counter (OTC) for the treatment and/or prevention of nocturnal leg muscle cramps.

    (a) Quinine sulfate alone or in combination with vitamin E has been 
present in over-the-counter (OTC) drug products for the treatment and/or 
prevention of nocturnal leg muscle cramps, i.e., a condition of 
localized pain in the lower extremities usually occurring in middle life 
and beyond with no regular pattern concerning time or severity. There is 
a lack of adequate data to establish general recognition of the safety 
and effectiveness of quinine sulfate, vitamin E, or any other 
ingredients for OTC use in the treatment and/or prevention of nocturnal 
leg muscle cramps. In the doses used to treat or prevent this condition, 
quinine sulfate has caused adverse events such as transient visual and 
auditory disturbances, dizziness, fever, nausea, vomiting, and diarrhea. 
Quinine sulfate may cause unpredictable serious and life-threatening 
hypersensitivity reactions requiring medical intervention and 
hospitalization; fatalities have been reported. The risk associated with 
use of quinine sulfate, in the absence of evidence of its effectiveness, 
outweighs any potential benefit in treating and/or preventing this 
benign, self-limiting condition. Based upon the adverse benefit-to-risk 
ratio, any drug product containing quinine or quinine sulfate cannot be 
considered generally recognized as safe for the treatment and/or 
prevention of nocturnal leg muscle cramps.
    (b) Any OTC drug product that is labeled, represented, or promoted 
for the treatment and/or prevention of nocturnal leg muscle cramps is 
regarded as a new drug within the meaning of section 201(p) of the 
Federal Food, Drug, and Cosmetic Act (the act), for which an approved 
application or abbreviated application under section 505 of the act and 
part 314 of this chapter is required for marketing. In the absence of an 
approved new drug application or abbreviated new drug application, such 
product is also misbranded under section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for OTC use for the 
treatment and/or prevention of nocturnal leg muscle cramps is safe and 
effective for the purpose intended must comply with the requirements and 
procedures governing the use of investigational new drugs set forth in 
part 312 of this chapter.
    (d) After February 22, 1995, any such OTC drug product initially 
introduced or initially delivered for introduction into interstate 
commerce that is not in compliance with this section is subject to 
regulatory action.
[59 FR 43252, Aug. 22, 1994]



Sec. 310.547  Drug products containing quinine offered over-the-counter (OTC) for the treatment and/or prevention of malaria.

    (a) Quinine and quinine salts have been used OTC for the treatment 
and/or prevention of malaria, a serious and potentially life-threatening 
disease. Quinine is no longer the drug of choice for the treatment and/
or prevention of most types of malaria. In addition, there are serious 
and complicating aspects of the disease itself and some potentially 
serious and life-threatening risks associated with the use of quinine at 
doses employed for the treatment of malaria. There is a lack of adequate 
data to establish general recognition of the safety of quinine drug 
products for OTC use in the treatment and/or prevention of malaria. 
Therefore, quinine or quinine salts cannot be safely and effectively 
used for the treatment and/or prevention of malaria except under the 
care and supervision of a doctor.
    (b) Any OTC drug product containing quinine or quinine salts that is 
labeled, represented, or promoted for the treatment and/or prevention of 
malaria is regarded as a new drug within the meaning of section 201(p) 
of the act, for which an approved application or abbreviated application 
under section 505 of the act and part 314 of this chapter

[[Page 61]]

is required for marketing. In the absence of an approved new drug 
application or abbreviated new drug application, such product is also 
misbranded under section 502 of the act.
    (c) Clinical investigations designed to obtain evidence that any 
drug product labeled, represented, or promoted for OTC use for the 
treatment and/or prevention of malaria is safe and effective for the 
purpose intended must comply with the requirements and procedures 
governing the use of investigational new drugs set forth in part 312 of 
this chapter.
    (d) After April 20, 1998, any such OTC drug product initially 
introduced or initially delivered for introduction into interstate 
commerce that is not in compliance with this section is subject to 
regulatory action.
[63 FR 13528, Mar. 20, 1998]

    Effective Date Note: At 63 FR 13528, Mar. 20, 1998, Sec. 310.547 was 
added to subpart E, effective Apr. 20, 1998.



PART 312--INVESTIGATIONAL NEW DRUG APPLICATION--Table of Contents




                      Subpart A--General Provisions

Sec.
312.1  Scope.
312.2  Applicability.
312.3  Definitions and interpretations.
312.6  Labeling of an investigational new drug.
312.7  Promotion and charging for investigational drugs.
312.10  Waivers.

          Subpart B--Investigational New Drug Application (IND)

312.20  Requirement for an IND.
312.21  Phases of an investigation.
312.22  General principles of the IND submission.
312.23  IND content and format.
312.30  Protocol amendments.
312.31  Information amendments.
312.32  IND safety reports.
312.33  Annual reports.
312.34  Treatment use of an investigational new drug.
312.35  Submissions for treatment use.
312.36  Emergency use of an investigational new drug.
312.38  Withdrawal of an IND.

                    Subpart C--Administrative Actions

312.40  General requirements for use of an investigational new drug in a 
          clinical investigation.
312.41  Comment and advice on an IND.
312.42  Clinical holds and requests for modification.
312.44  Termination.
312.45  Inactive status.
312.47  Meetings.
312.48  Dispute resolution.

        Subpart D--Responsibilities of Sponsors and Investigators

312.50  General responsibilities of sponsors.
312.52  Transfer of obligations to a contract research organization.
312.53  Selecting investigators and monitors.
312.54  Emergency research under Sec. 50.24 of this chapter.
312.55  Informing investigators.
312.56  Review of ongoing investigations.
312.57  Recordkeeping and record retention.
312.58  Inspection of sponsor's records and reports.
312.59  Disposition of unused supply of investigational drug.
312.60  General responsibilities of investigators.
312.61  Control of the investigational drug.
312.62  Investigator recordkeeping and record retention.
312.64  Investigator reports.
312.66  Assurance of IRB review.
312.68  Inspection of investigator's records and reports.
312.69  Handling of controlled substances.
312.70  Disqualification of a clinical investigator.

    Subpart E--Drugs Intended to Treat Life-threatening and Severely-
                         debilitating Illnesses

312.80  Purpose.
312.81  Scope.
312.82  Early consultation.
312.83  Treatment protocols.
312.84  Risk-benefit analysis in review of marketing applications for 
          drugs to treat life-threatening and severely-debilitating 
          illnesses.
312.85  Phase 4 studies.
312.86  Focused FDA regulatory research.
312.87  Active monitoring of conduct and evaluation of clinical trials.
312.88  Safeguards for patient safety.

                        Subpart F--Miscellaneous

312.110  Import and export requirements.
312.120  Foreign clinical studies not conducted under an IND.
312.130  Availability for public disclosure of data and information in 
          an IND.
312.140  Address for correspondence.
312.145  Guidelines.

[[Page 62]]

Subpart G--Drugs for Investigational Use in Laboratory Research Animals 
                            or in Vitro Tests

312.160  Drugs for investigational use in laboratory research animals or 
          in vitro tests.

    Authority:  21 U.S.C. 321, 331, 351, 352, 353, 355, 356, 357, 371; 
42 U.S.C. 262.

    Source:  52 FR 8831, Mar. 19, 1987, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 312.1  Scope.

    (a) This part contains procedures and requirements governing the use 
of investigational new drugs, including procedures and requirements for 
the submission to, and review by, the Food and Drug Administration of 
investigational new drug applications (IND's). An investigational new 
drug for which an IND is in effect in accordance with this part is 
exempt from the premarketing approval requirements that are otherwise 
applicable and may be shipped lawfully for the purpose of conducting 
clinical investigations of that drug.
    (b) References in this part to regulations in the Code of Federal 
Regulations are to chapter I of title 21, unless otherwise noted.



Sec. 312.2  Applicability.

    (a) Applicability. Except as provided in this section, this part 
applies to all clinical investigations of products that are subject to 
section 505 or 507 of the Federal Food, Drug, and Cosmetic Act or to the 
licensing provisions of the Public Health Service Act (58 Stat. 632, as 
amended (42 U.S.C. 201 et seq.)).
    (b) Exemptions. (1) The clinical investigation of a drug product 
that is lawfully marketed in the United States is exempt from the 
requirements of this part if all the following apply:
    (i) The investigation is not intended to be reported to FDA as a 
well-controlled study in support of a new indication for use nor 
intended to be used to support any other significant change in the 
labeling for the drug;
    (ii) If the drug that is undergoing investigation is lawfully 
marketed as a prescription drug product, the investigation is not 
intended to support a significant change in the advertising for the 
product;
    (iii) The investigation does not involve a route of administration 
or dosage level or use in a patient population or other factor that 
significantly increases the risks (or decreases the acceptability of the 
risks) associated with the use of the drug product;
    (iv) The investigation is conducted in compliance with the 
requirements for institutional review set forth in part 56 and with the 
requirements for informed consent set forth in part 50; and
    (v) The investigation is conducted in compliance with the 
requirements of Sec. 312.7.
    (2)(i) A clinical investigation involving an in vitro diagnostic 
biological product listed in paragraph (b)(2)(ii) of this section is 
exempt from the requirements of this part if (a) it is intended to be 
used in a diagnostic procedure that confirms the diagnosis made by 
another, medically established, diagnostic product or procedure and (b) 
it is shipped in compliance with Sec. 312.160.
    (ii) In accordance with paragraph (b)(2)(i) of this section, the 
following products are exempt from the requirements of this part: (a) 
blood grouping serum; (b) reagent red blood cells; and (c) anti-human 
globulin.
    (3) A drug intended solely for tests in vitro or in laboratory 
research animals is exempt from the requirements of this part if shipped 
in accordance with Sec. 312.160.
    (4) FDA will not accept an application for an investigation that is 
exempt under the provisions of paragraph (b)(1) of this section.
    (5) A clinical investigation involving use of a placebo is exempt 
from the requirements of this part if the investigation does not 
otherwise require submission of an IND.
    (6) A clinical investigation involving an exception from informed 
consent under Sec. 50.24 of this chapter is not exempt from the 
requirements of this part.
    (c) Bioavailability studies. The applicability of this part to in 
vivo bioavailability studies in humans is subject to the provisions of 
Sec. 320.31.

[[Page 63]]

    (d) Unlabeled indication. This part does not apply to the use in the 
practice of medicine for an unlabeled indication of a new drug or 
antibiotic drug product approved under part 314 or of a licensed 
biological product.
    (e) Guidance. FDA may, on its own initiative, issue guidance on the 
applicability of this part to particular investigational uses of drugs. 
On request, FDA will advise on the applicability of this part to a 
planned clinical investigation.
[52 FR 8831, Mar. 19, 1987, as amended at 61 FR 51529, Oct. 2, 1996]



Sec. 312.3  Definitions and interpretations.

    (a) The definitions and interpretations of terms contained in 
section 201 of the Act apply to those terms when used in this part:
    (b) The following definitions of terms also apply to this part:
    Act means the Federal Food, Drug, and Cosmetic Act (secs. 201-902, 
52 Stat. 1040 et seq., as amended (21 U.S.C. 301-392)).
    Clinical investigation means any experiment in which a drug is 
administered or dispensed to, or used involving, one or more human 
subjects. For the purposes of this part, an experiment is any use of a 
drug except for the use of a marketed drug in the course of medical 
practice.
    Contract research organization means a person that assumes, as an 
independent contractor with the sponsor, one or more of the obligations 
of a sponsor, e.g., design of a protocol, selection or monitoring of 
investigations, evaluation of reports, and preparation of materials to 
be submitted to the Food and Drug Administration.
    FDA means the Food and Drug Administration.
    IND means an investigational new drug application. For purposes of 
this part, ``IND'' is synonymous with ``Notice of Claimed 
Investigational Exemption for a New Drug.''
    Investigational new drug means a new drug, antibiotic drug, or 
biological drug that is used in a clinical investigation. The term also 
includes a biological product that is used in vitro for diagnostic 
purposes. The terms ``investigational drug'' and ``investigational new 
drug'' are deemed to be synonymous for purposes of this part.
    Investigator means an individual who actually conducts a clinical 
investigation (i.e., under whose immediate direction the drug is 
administered or dispensed to a subject). In the event an investigation 
is conducted by a team of individuals, the investigator is the 
responsible leader of the team. ``Subinvestigator'' includes any other 
individual member of that team.
    Marketing application means an application for a new drug submitted 
under section 505(b) of the Act, a request to provide for certification 
of an antibiotic submitted under section 507 of the Act, or a product 
license application for a biological product submitted under the Public 
Health Service Act.
    Sponsor means a person who takes responsibility for and initiates a 
clinical investigation. The sponsor may be an individual or 
pharmaceutical company, governmental agency, academic institution, 
private organization, or other organization. The sponsor does not 
actually conduct the investigation unless the sponsor is a sponsor-
investigator. A person other than an individual that uses one or more of 
its own employees to conduct an investigation that it has initiated is a 
sponsor, not a sponsor-investigator, and the employees are 
investigators.
    Sponsor-Investigator means an individual who both initiates and 
conducts an investigation, and under whose immediate direction the 
investigational drug is administered or dispensed. The term does not 
include any person other than an individual. The requirements applicable 
to a sponsor-investigator under this part include both those applicable 
to an investigator and a sponsor.
    Subject means a human who participates in an investigation, either 
as a recipient of the investigational new drug or as a control. A 
subject may be a healthy human or a patient with a disease.



Sec. 312.6  Labeling of an investigational new drug.

    (a) The immediate package of an investigational new drug intended 
for human use shall bear a label with the

[[Page 64]]

statement ``Caution: New Drug--Limited by Federal (or United States) law 
to investigational use.''
    (b) The label or labeling of an investigational new drug shall not 
bear any statement that is false or misleading in any particular and 
shall not represent that the investigational new drug is safe or 
effective for the purposes for which it is being investigated.



Sec. 312.7  Promotion and charging for investigational drugs.

    (a) Promotion of an investigational new drug. A sponsor or 
investigator, or any person acting on behalf of a sponsor or 
investigator, shall not represent in a promotional context that an 
investigational new drug is safe or effective for the purposes for which 
it is under investigation or otherwise promote the drug. This provision 
is not intended to restrict the full exchange of scientific information 
concerning the drug, including dissemination of scientific findings in 
scientific or lay media. Rather, its intent is to restrict promotional 
claims of safety or effectiveness of the drug for a use for which it is 
under investigation and to preclude commercialization of the drug before 
it is approved for commercial distribution.
    (b) Commercial distribution of an investigational new drug. A 
sponsor or investigator shall not commercially distribute or test market 
an investigational new drug.
    (c) Prolonging an investigation. A sponsor shall not unduly prolong 
an investigation after finding that the results of the investigation 
appear to establish sufficient data to support a marketing application.
    (d) Charging for and commercialization of investigational drugs--(1) 
Clinical trials under an IND. Charging for an investigational drug in a 
clinical trial under an IND is not permitted without the prior written 
approval of FDA. In requesting such approval, the sponsor shall provide 
a full written explanation of why charging is necessary in order for the 
sponsor to undertake or continue the clinical trial, e.g., why 
distribution of the drug to test subjects should not be considered part 
of the normal cost of doing business.
    (2) Treatment protocol or treatment IND. A sponsor or investigator 
may charge for an investigational drug for a treatment use under a 
treatment protocol or treatment IND provided: (i) There is adequate 
enrollment in the ongoing clinical investigations under the authorized 
IND; (ii) charging does not constitute commercial marketing of a new 
drug for which a marketing application has not been approved; (iii) the 
drug is not being commercially promoted or advertised; and (iv) the 
sponsor of the drug is actively pursuing marketing approval with due 
diligence. FDA must be notified in writing in advance of commencing any 
such charges, in an information amendment submitted under Sec. 312.31. 
Authorization for charging goes into effect automatically 30 days after 
receipt by FDA of the information amendment, unless the sponsor is 
notified to the contrary.
    (3) Noncommercialization of investigational drug. Under this 
section, the sponsor may not commercialize an investigational drug by 
charging a price larger than that necessary to recover costs of 
manufacture, research, development, and handling of the investigational 
drug.
    (4) Withdrawal of authorization. Authorization to charge for an 
investigational drug under this section may be withdrawn by FDA if the 
agency finds that the conditions underlying the authorization are no 
longer satisfied.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 19476, May 22, 1987]



Sec. 312.10  Waivers.

    (a) A sponsor may request FDA to waive applicable requirement under 
this part. A waiver request may be submitted either in an IND or in an 
information amendment to an IND. In an emergency, a request may be made 
by telephone or other rapid communication means. A waiver request is 
required to contain at least one of the following:
    (1) An explanation why the sponsor's compliance with the requirement 
is unnecessary or cannot be achieved;

[[Page 65]]

    (2) A description of an alternative submission or course of action 
that satisfies the purpose of the requirement; or
    (3) Other information justifying a waiver.
    (b) FDA may grant a waiver if it finds that the sponsor's 
noncompliance would not pose a significant and unreasonable risk to 
human subjects of the investigation and that one of the following is 
met:
    (1) The sponsor's compliance with the requirement is unnecessary for 
the agency to evaluate the application, or compliance cannot be 
achieved;
    (2) The sponsor's proposed alternative satisfies the requirement; or
    (3) The applicant's submission otherwise justifies a waiver.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987]



          Subpart B--Investigational New Drug Application (IND)



Sec. 312.20  Requirement for an IND.

    (a) A sponsor shall submit an IND to FDA if the sponsor intends to 
conduct a clinical investigation with an investigational new drug that 
is subject to Sec. 312.2(a).
    (b) A sponsor shall not begin a clinical investigation subject to 
Sec. 312.2(a) until the investigation is subject to an IND which is in 
effect in accordance with Sec. 312.40.
    (c) A sponsor shall submit a separate IND for any clinical 
investigation involving an exception from informed consent under 
Sec. 50.24 of this chapter. Such a clinical investigation is not 
permitted to proceed without the prior written authorization from FDA. 
FDA shall provide a written determination 30 days after FDA receives the 
IND or earlier.
[52 FR 8831, Mar. 19, 1987, as amended at 61 FR 51529, Oct. 2, 1996; 62 
FR 32479, June 16, 1997]



Sec. 312.21  Phases of an investigation.

    An IND may be submitted for one or more phases of an investigation. 
The clinical investigation of a previously untested drug is generally 
divided into three phases. Although in general the phases are conducted 
sequentially, they may overlap. These three phases of an investigation 
are a follows:
    (a) Phase 1. (1) Phase 1 includes the initial introduction of an 
investigational new drug into humans. Phase 1 studies are typically 
closely monitored and may be conducted in patients or normal volunteer 
subjects. These studies are designed to determine the metabolism and 
pharmacologic actions of the drug in humans, the side effects associated 
with increasing doses, and, if possible, to gain early evidence on 
effectiveness. During Phase 1, sufficient information about the drug's 
pharmacokinetics and pharmacological effects should be obtained to 
permit the design of well-controlled, scientifically valid, Phase 2 
studies. The total number of subjects and patients included in Phase 1 
studies varies with the drug, but is generally in the range of 20 to 80.
    (2) Phase 1 studies also include studies of drug metabolism, 
structure-activity relationships, and mechanism of action in humans, as 
well as studies in which investigational drugs are used as research 
tools to explore biological phenomena or disease processes.
    (b) Phase 2. Phase 2 includes the controlled clinical studies 
conducted to evaluate the effectiveness of the drug for a particular 
indication or indications in patients with the disease or condition 
under study and to determine the common short-term side effects and 
risks associated with the drug. Phase 2 studies are typically well 
controlled, closely monitored, and conducted in a relatively small 
number of patients, usually involving no more than several hundred 
subjects.
    (c) Phase 3. Phase 3 studies are expanded controlled and 
uncontrolled trials. They are performed after preliminary evidence 
suggesting effectiveness of the drug has been obtained, and are intended 
to gather the additional information about effectiveness and safety that 
is needed to evaluate the overall benefit-risk relationship of the drug 
and to provide an adequate basis for physician labeling. Phase 3 studies 
usually include from several hundred to several thousand subjects.

[[Page 66]]



Sec. 312.22  General principles of the IND submission.

    (a) FDA's primary objectives in reviewing an IND are, in all phases 
of the investigation, to assure the safety and rights of subjects, and, 
in Phase 2 and 3, to help assure that the quality of the scientific 
evaluation of drugs is adequate to permit an evaluation of the drug's 
effectiveness and safety. Therefore, although FDA's review of Phase 1 
submissions will focus on assessing the safety of Phase 1 
investigations, FDA's review of Phases 2 and 3 submissions will also 
include an assessment of the scientific quality of the clinical 
investigations and the likelihood that the investigations will yield 
data capable of meeting statutory standards for marketing approval.
    (b) The amount of information on a particular drug that must be 
submitted in an IND to assure the accomplishment of the objectives 
described in paragraph (a) of this section depends upon such factors as 
the novelty of the drug, the extent to which it has been studied 
previously, the known or suspected risks, and the developmental phase of 
the drug.
    (c) The central focus of the initial IND submission should be on the 
general investigational plan and the protocols for specific human 
studies. Subsequent amendments to the IND that contain new or revised 
protocols should build logically on previous submissions and should be 
supported by additional information, including the results of animal 
toxicology studies or other human studies as appropriate. Annual reports 
to the IND should serve as the focus for reporting the status of studies 
being conducted under the IND and should update the general 
investigational plan for the coming year.
    (d) The IND format set forth in Sec. 312.23 should be followed 
routinely by sponsors in the interest of fostering an efficient review 
of applications. Sponsors are expected to exercise considerable 
discretion, however, regarding the content of information submitted in 
each section, depending upon the kind of drug being studied and the 
nature of the available information. Section 312.23 outlines the 
information needed for a commercially sponsored IND for a new molecular 
entity. A sponsor-investigator who uses, as a research tool, an 
investigational new drug that is already subject to a manufacturer's IND 
or marketing application should follow the same general format, but 
ordinarily may, if authorized by the manufacturer, refer to the 
manufacturer's IND or marketing application in providing the technical 
information supporting the proposed clinical investigation. A sponsor-
investigator who uses an investigational drug not subject to a 
manufacturer's IND or marketing application is ordinarily required to 
submit all technical information supporting the IND, unless such 
information may be referenced from the scientific literature.



Sec. 312.23  IND content and format.

    (a) A sponsor who intends to conduct a clinical investigation 
subject to this part shall submit an ``Investigational New Drug 
Application'' (IND) including, in the following order:
    (1) Cover sheet (Form FDA-1571). A cover sheet for the application 
containing the following:
    (i) The name, address, and telephone number of the sponsor, the date 
of the application, and the name of the investigational new drug.
    (ii) Identification of the phase or phases of the clinical 
investigation to be conducted.
    (iii) A commitment not to begin clinical investigations until an IND 
covering the investigations is in effect.
    (iv) A commitment that an Institutional Review Board (IRB) that 
complies with the requirements set forth in part 56 will be responsible 
for the initial and continuing review and approval of each of the 
studies in the proposed clinical investigation and that the investigator 
will report to the IRB proposed changes in the research activity in 
accordance with the requirements of part 56.
    (v) A commitment to conduct the investigation in accordance with all 
other applicable regulatory requirements.
    (vi) The name and title of the person responsible for monitoring the 
conduct and progress of the clinical investigations.
    (vii) The name(s) and title(s) of the person(s) responsible under 
Sec. 312.32 for

[[Page 67]]

review and evaluation of information relevant to the safety of the drug.
    (viii) If a sponsor has transferred any obligations for the conduct 
of any clinical study to a contract research organization, a statement 
containing the name and address of the contract research organization, 
identification of the clinical study, and a listing of the obligations 
transferred. If all obligations governing the conduct of the study have 
been transferred, a general statement of this transfer--in lieu of a 
listing of the specific obligations transferred--may be submitted.
    (ix) The signature of the sponsor or the sponsor's authorized 
representative. If the person signing the application does not reside or 
have a place of business within the United States, the IND is required 
to contain the name and address of, and be countersigned by, an 
attorney, agent, or other authorized official who resides or maintains a 
place of business within the United States.
    (2) A table of contents.
    (3) Introductory statement and general investigational plan. (i) A 
brief introductory statement giving the name of the drug and all active 
ingredients, the drug's pharmacological class, the structural formula of 
the drug (if known), the formulation of the dosage form(s) to be used, 
the route of administration, and the broad objectives and planned 
duration of the proposed clinical investigation(s).
    (ii) A brief summary of previous human experience with the drug, 
with reference to other IND's if pertinent, and to investigational or 
marketing experience in other countries that may be relevant to the 
safety of the proposed clinical investigation(s).
    (iii) If the drug has been withdrawn from investigation or marketing 
in any country for any reason related to safety or effectiveness, 
identification of the country(ies) where the drug was withdrawn and the 
reasons for the withdrawal.
    (iv) A brief description of the overall plan for investigating the 
drug product for the following year. The plan should include the 
following: (a) The rationale for the drug or the research study; (b) the 
indication(s) to be studied; (c) the general approach to be followed in 
evaluating the drug; (d) the kinds of clinical trials to be conducted in 
the first year following the submission (if plans are not developed for 
the entire year, the sponsor should so indicate); (e) the estimated 
number of patients to be given the drug in those studies; and (f) any 
risks of particular severity or seriousness anticipated on the basis of 
the toxicological data in animals or prior studies in humans with the 
drug or related drugs.
    (4) [Reserved]
    (5) Investigator's brochure. If required under Sec. 312.55, a copy 
of the investigator's brochure, containing the following information:
    (i) A brief description of the drug substance and the formulation, 
including the structural formula, if known.
    (ii) A summary of the pharmacological and toxicological effects of 
the drug in animals and, to the extent known, in humans.
    (iii) A summary of the pharmacokinetics and biological disposition 
of the drug in animals and, if known, in humans.
    (iv) A summary of information relating to safety and effectiveness 
in humans obtained from prior clinical studies. (Reprints of published 
articles on such studies may be appended when useful.)
    (v) A description of possible risks and side effects to be 
anticipated on the basis of prior experience with the drug under 
investigation or with related drugs, and of precautions or special 
monitoring to be done as part of the investigational use of the drug.
    (6) Protocols. (i) A protocol for each planned study. (Protocols for 
studies not submitted initially in the IND should be submitted in 
accordance with Sec. 312.30(a).) In general, protocols for Phase 1 
studies may be less detailed and more flexible than protocols for Phase 
2 and 3 studies. Phase 1 protocols should be directed primarily at 
providing an outline of the investigation--an estimate of the number of 
patients to be involved, a description of safety exclusions, and a 
description of the dosing plan including duration, dose, or method to be 
used in determining dose--and should specify in detail only those 
elements of the study that are critical to safety, such as necessary

[[Page 68]]

monitoring of vital signs and blood chemistries. Modifications of the 
experimental design of Phase 1 studies that do not affect critical 
safety assessments are required to be reported to FDA only in the annual 
report.
    (ii) In Phases 2 and 3, detailed protocols describing all aspects of 
the study should be submitted. A protocol for a Phase 2 or 3 
investigation should be designed in such a way that, if the sponsor 
anticipates that some deviation from the study design may become 
necessary as the investigation progresses, alternatives or contingencies 
to provide for such deviation are built into the protocols at the 
outset. For example, a protocol for a controlled short-term study might 
include a plan for an early crossover of nonresponders to an alternative 
therapy.
    (iii) A protocol is required to contain the following, with the 
specific elements and detail of the protocol reflecting the above 
distinctions depending on the phase of study:
    (a) A statement of the objectives and purpose of the study.
    (b) The name and address and a statement of the qualifications 
(curriculum vitae or other statement of qualifications) of each 
investigator, and the name of each subinvestigator (e.g., research 
fellow, resident) working under the supervision of the investigator; the 
name and address of the research facilities to be used; and the name and 
address of each reviewing Institutional Review Board.
    (c) The criteria for patient selection and for exclusion of patients 
and an estimate of the number of patients to be studied.
    (d) A description of the design of the study, including the kind of 
control group to be used, if any, and a description of methods to be 
used to minimize bias on the part of subjects, investigators, and 
analysts.
    (e) The method for determining the dose(s) to be administered, the 
planned maximum dosage, and the duration of individual patient exposure 
to the drug.
    (f) A description of the observations and measurements to be made to 
fulfill the objectives of the study.
    (g) A description of clinical procedures, laboratory tests, or other 
measures to be taken to monitor the effects of the drug in human 
subjects and to minimize risk.
    (7) Chemistry, manufacturing, and control information. (i) As 
appropriate for the particular investigations covered by the IND, a 
section describing the composition, manufacture, and control of the drug 
substance and the drug product. Although in each phase of the 
investigation sufficient information is required to be submitted to 
assure the proper identification, quality, purity, and strength of the 
investigational drug, the amount of information needed to make that 
assurance will vary with the phase of the investigation, the proposed 
duration of the investigation, the dosage form, and the amount of 
information otherwise available. FDA recognizes that modifications to 
the method of preparation of the new drug substance and dosage form and 
changes in the dosage form itself are likely as the investigation 
progresses. Therefore, the emphasis in an initial Phase 1 submission 
should generally be placed on the identification and control of the raw 
materials and the new drug substance. Final specifications for the drug 
substance and drug product are not expected until the end of the 
investigational process.
    (ii) It should be emphasized that the amount of information to be 
submitted depends upon the scope of the proposed clinical investigation. 
For example, although stability data are required in all phases of the 
IND to demonstrate that the new drug substance and drug product are 
within acceptable chemical and physical limits for the planned duration 
of the proposed clinical investigation, if very short-term tests are 
proposed, the supporting stability data can be correspondingly limited.
    (iii) As drug development proceeds and as the scale or production is 
changed from the pilot-scale production appropriate for the limited 
initial clinical investigations to the larger-scale production needed 
for expanded clinical trials, the sponsor should submit information 
amendments to supplement the initial information submitted on the 
chemistry, manufacturing, and control processes with information 
appropriate to the expanded scope of the investigation.

[[Page 69]]

    (iv) Reflecting the distinctions described in this paragraph (a)(7), 
and based on the phase(s) to be studied, the submission is required to 
contain the following:
    (a) Drug substance. A description of the drug substance, including 
its physical, chemical, or biological characteristics; the name and 
address of its manufacturer; the general method of preparation of the 
drug substance; the acceptable limits and analytical methods used to 
assure the identity, strength, quality, and purity of the drug 
substance; and information sufficient to support stability of the drug 
substance during the toxicological studies and the planned clinical 
studies. Reference to the current edition of the United States 
Pharmacopeia--National Formulary may satisfy relevant requirements in 
this paragraph.
    (b) Drug product. A list of all components, which may include 
reasonable alternatives for inactive compounds, used in the manufacture 
of the investigational drug product, including both those components 
intended to appear in the drug product and those which may not appear 
but which are used in the manufacturing process, and, where applicable, 
the quantitative composition of the investigational drug product, 
including any reasonable variations that may be expected during the 
investigational stage; the name and address of the drug product 
manufacturer; a brief general description of the manufacturing and 
packaging procedure as appropriate for the product; the acceptable 
limits and analytical methods used to assure the identity, strength, 
quality, and purity of the drug product; and information sufficient to 
assure the product's stability during the planned clinical studies. 
Reference to the current edition of the United States Pharmacopeia--
National Formulary may satisfy certain requirements in this paragraph.
    (c) A brief general description of the composition, manufacture, and 
control of any placebo used in a controlled clinical trial.
    (d) Labeling. A copy of all labels and labeling to be provided to 
each investigator.
    (e) Environmental analysis requirements. A claim for categorical 
exclusion under Sec. 25.30 or 25.31 or an environmental assessment under 
Sec. 25.40.
    (8) Pharmacology and toxicology information. Adequate information 
about pharmacological and toxicological studies of the drug involving 
laboratory animals or in vitro, on the basis of which the sponsor has 
concluded that it is reasonably safe to conduct the proposed clinical 
investigations. The kind, duration, and scope of animal and other tests 
required varies with the duration and nature of the proposed clinical 
investigations. Guidelines are available from FDA that describe ways in 
which these requirements may be met. Such information is required to 
include the identification and qualifications of the individuals who 
evaluated the results of such studies and concluded that it is 
reasonably safe to begin the proposed investigations and a statement of 
where the investigations were conducted and where the records are 
available for inspection. As drug development proceeds, the sponsor is 
required to submit informational amendments, as appropriate, with 
additional information pertinent to safety.
    (i) Pharmacology and drug disposition. A section describing the 
pharmacological effects and mechanism(s) of action of the drug in 
animals, and information on the absorption, distribution, metabolism, 
and excretion of the drug, if known.
    (ii) Toxicology. (a) An integrated summary of the toxicological 
effects of the drug in animals and in vitro. Depending on the nature of 
the drug and the phase of the investigation, the description is to 
include the results of acute, subacute, and chronic toxicity tests; 
tests of the drug's effects on reproduction and the developing fetus; 
any special toxicity test related to the drug's particular mode of 
administration or conditions of use (e.g., inhalation, dermal, or ocular 
toxicology); and any in vitro studies intended to evaluate drug 
toxicity.
    (b) For each toxicology study that is intended primarily to support 
the safety of the proposed clinical investigation, a full tabulation of 
data suitable for detailed review.
    (iii) For each nonclinical laboratory study subject to the good 
laboratory practice regulations under part 58, a

[[Page 70]]

statement that the study was conducted in compliance with the good 
laboratory practice regulations in part 58, or, if the study was not 
conducted in compliance with those regulations, a brief statement of the 
reason for the noncompliance.
    (9) Previous human experience with the investigational drug. A 
summary of previous human experience known to the applicant, if any, 
with the investigational drug. The information is required to include 
the following:
    (i) If the investigational drug has been investigated or marketed 
previously, either in the United States or other countries, detailed 
information about such experience that is relevant to the safety of the 
proposed investigation or to the investigation's rationale. If the durg 
has been the subject of controlled trials, detailed information on such 
trials that is relevant to an assessment of the drug's effectiveness for 
the proposed investigational use(s) should also be provided. Any 
published material that is relevant to the safety of the proposed 
investigation or to an assessment of the drug's effectiveness for its 
proposed investigational use should be provided in full. Published 
material that is less directly relevant may be supplied by a 
bibliography.
    (ii) If the drug is a combination of drugs previously investigated 
or marketed, the information required under paragraph (a)(9)(i) of this 
section should be provided for each active drug component. However, if 
any component in such combination is subject to an approved marketing 
application or is otherwise lawfully marketed in the United States, the 
sponsor is not required to submit published material concerning that 
active drug component unless such material relates directly to the 
proposed investigational use (including publications relevant to 
component-component interaction).
    (iii) If the drug has been marketed outside the United States, a 
list of the countries in which the drug has been marketed and a list of 
the countries in which the drug has been withdrawn from marketing for 
reasons potentially related to safety or effectiveness.
    (10) Additional information. In certain applications, as described 
below, information on special topics may be needed. Such information 
shall be submitted in this section as follows:
    (i) Drug dependence and abuse potential. If the drug is a 
psychotropic substance or otherwise has abuse potential, a section 
describing relevant clinical studies and experience and studies in test 
animals.
    (ii) Radioactive drugs. If the drug is a radioactive drug, 
sufficient data from animal or human studies to allow a reasonable 
calculation of radiation-absorbed dose to the whole body and critical 
organs upon administration to a human subject. Phase 1 studies of 
radioactive drugs must include studies which will obtain sufficient data 
for dosimetry calculations.
    (iii) Other information. A brief statement of any other information 
that would aid evaluation of the proposed clinical investigations with 
respect to their safety or their design and potential as controlled 
clinical trials to support marketing of the drug.
    (11) Relevant information. If requested by FDA, any other relevant 
information needed for review of the application.
    (b) Information previously submitted. The sponsor ordinarily is not 
required to resubmit information previously submitted, but may 
incorporate the information by reference. A reference to information 
submitted previously must identify the file by name, reference number, 
volume, and page number where the information can be found. A reference 
to information submitted to the agency by a person other than the 
sponsor is required to contain a written statement that authorizes the 
reference and that is signed by the person who submitted the 
information.
    (c) Material in a foreign language. The sponsor shall submit an 
accurate and complete English translation of each part of the IND that 
is not in English. The sponsor shall also submit a copy of each original 
literature publication for which an English translation is submitted.
    (d) Number of copies. The sponsor shall submit an original and two 
copies of all submissions to the IND file, including the original 
submission and all amendments and reports.
    (e) Numbering of IND submissions. Each submission relating to an IND 
is

[[Page 71]]

required to be numbered serially using a single, three-digit serial 
number. The initial IND is required to be numbered 000; each subsequent 
submission (e.g., amendment, report, or correspondence) is required to 
be numbered chronologically in sequence.
    (f) Identification of exception from informed consent. If the 
investigation involves an exception from informed consent under 
Sec. 50.24 of this chapter, the sponsor shall prominently identify on 
the cover sheet that the investigation is subject to the requirements in 
Sec. 50.24 of this chapter.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 53 
FR 1918, Jan. 25, 1988; 61 FR 51529, Oct. 2, 1996; 62 FR 40599, July 29, 
1997]



Sec. 312.30  Protocol amendments.

    Once an IND is in effect, a sponsor shall amend it as needed to 
ensure that the clinical investigations are conducted according to 
protocols included in the application. This section sets forth the 
provisions under which new protocols may be submitted and changes in 
previously submitted protocols may be made. Whenever a sponsor intends 
to conduct a clinical investigation with an exception from informed 
consent for emergency research as set forth in Sec. 50.24 of this 
chapter, the sponsor shall submit a separate IND for such investigation.
    (a) New protocol. Whenever a sponsor intends to conduct a study that 
is not covered by a protocol already contained in the IND, the sponsor 
shall submit to FDA a protocol amendment containing the protocol for the 
study. Such study may begin provided two conditions are met: (1) The 
sponsor has submitted the protocol to FDA for its review; and (2) the 
protocol has been approved by the Institutional Review Board (IRB) with 
responsibility for review and approval of the study in accordance with 
the requirements of part 56. The sponsor may comply with these two 
conditions in either order.
    (b) Changes in a protocol. (1) A sponsor shall submit a protocol 
amendment describing any change in a Phase 1 protocol that significantly 
affects the safety of subjects or any change in a Phase 2 or 3 protocol 
that significantly affects the safety of subjects, the scope of the 
investigation, or the scientific quality of the study. Examples of 
changes requiring an amendment under this paragraph include:
    (i) Any increase in drug dosage or duration of exposure of 
individual subjects to the drug beyond that in the current protocol, or 
any significant increase in the number of subjects under study.
    (ii) Any significant change in the design of a protocol (such as the 
addition or dropping of a control group).
    (iii) The addition of a new test or procedure that is intended to 
improve monitoring for, or reduce the risk of, a side effect or adverse 
event; or the dropping of a test intended to monitor safety.
    (2)(i) A protocol change under paragraph (b)(1) of this section may 
be made provided two conditions are met:
    (a) The sponsor has submitted the change to FDA for its review; and
    (b) The change has been approved by the IRB with responsibility for 
review and approval of the study. The sponsor may comply with these two 
conditions in either order.
    (ii) Notwithstanding paragraph (b)(2)(i) of this section, a protocol 
change intended to eliminate an apparent immediate hazard to subjects 
may be implemented immediately provided FDA is subsequently notified by 
protocol amendment and the reviewing IRB is notified in accordance with 
Sec. 56.104(c).
    (c) New investigator. A sponsor shall submit a protocol amendment 
when a new investigator is added to carry out a previously submitted 
protocol, except that a protocol amendment is not required when a 
licensed practitioner is added in the case of a treatment protocol under 
Sec. 312.34. Once the investigator is added to the study, the 
investigational drug may be shipped to the investigator and the 
investigator may begin participating in the study. The sponsor shall 
notify FDA of the new investigator within 30 days of the investigator 
being added.

[[Page 72]]

    (d) Content and format. A protocol amendment is required to be 
prominently identified as such (i.e., ``Protocol Amendment: New 
Protocol'', ``Protocol Amendment: Change in Protocol'', or ``Protocol 
Amendment: New Investigator''), and to contain the following:
    (1)(i) In the case of a new protocol, a copy of the new protocol and 
a brief description of the most clinically significant differences 
between it and previous protocols.
    (ii) In the case of a change in protocol, a brief description of the 
change and reference (date and number) to the submission that contained 
the protocol.
    (iii) In the case of a new investigator, the investigator's name, 
the qualifications to conduct the investigation, reference to the 
previously submitted protocol, and all additional information about the 
investigator's study as is required under Sec. 312.23(a)(6)(iii)(b).
    (2) Reference, if necessary, to specific technical information in 
the IND or in a concurrently submitted information amendment to the IND 
that the sponsor relies on to support any clinically significant change 
in the new or amended protocol. If the reference is made to supporting 
information already in the IND, the sponsor shall identify by name, 
reference number, volume, and page number the location of the 
information.
    (3) If the sponsor desires FDA to comment on the submission, a 
request for such comment and the specific questions FDA's response 
should address.
    (e) When submitted. A sponsor shall submit a protocol amendment for 
a new protocol or a change in protocol before its implementation. 
Protocol amendments to add a new investigator or to provide additional 
information about investigators may be grouped and submitted at 30-day 
intervals. When several submissions of new protocols or protocol changes 
are anticipated during a short period, the sponsor is encouraged, to the 
extent feasible, to include these all in a single submission.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 53 
FR 1918, Jan. 25, 1988; 61 FR 51530, Oct. 2, 1996]



Sec. 312.31  Information amendments.

    (a) Requirement for information amendment. A sponsor shall report in 
an information amendment essential information on the IND that is not 
within the scope of a protocol amendment, IND safety reports, or annual 
report. Examples of information requiring an information amendment 
include:
    (1) New toxicology, chemistry, or other technical information; or
    (2) A report regarding the discontinuance of a clinical 
investigation.
    (b) Content and format of an information amendment. An information 
amendment is required to bear prominent identification of its contents 
(e.g., ``Information Amendment: Chemistry, Manufacturing, and Control'', 
``Information Amendment: Pharmacology-Toxicology'', ``Information 
Amendment: Clinical''), and to contain the following:
    (1) A statement of the nature and purpose of the amendment.
    (2) An organized submission of the data in a format appropriate for 
scientific review.
    (3) If the sponsor desires FDA to comment on an information 
amendment, a request for such comment.
    (c) When submitted. Information amendments to the IND should be 
submitted as necessary but, to the extent feasible, not more than every 
30 days.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 53 
FR 1918, Jan. 25, 1988]



Sec. 312.32  IND safety reports.

    (a) Definitions. The following definitions of terms apply to this 
section:-

[[Page 73]]

    Associated with the use of the drug. There is a reasonable 
possibility that the experience may have been caused by the drug.
    Disability. A substantial disruption of a person's ability to 
conduct normal life functions.
    Life-threatening adverse drug experience. Any adverse drug 
experience that places the patient or subject, in the view of the 
investigator, at immediate risk of death from the reaction as it 
occurred, i.e., it does not include a reaction that, had it occurred in 
a more severe form, might have caused death.
    Serious adverse drug experience: Any adverse drug experience 
occurring at any dose that results in any of the following outcomes: 
Death, a life-threatening adverse drug experience, inpatient 
hospitalization or prolongation of existing hospitalization, a 
persistent or significant disability/incapacity, or a congenital 
anomaly/birth defect. Important medical events that may not result in 
death, be life-threatening, or require hospitalization may be considered 
a serious adverse drug experience when, based upon appropriate medical 
judgment, they may jeopardize the patient or subject and may require 
medical or surgical intervention to prevent one of the outcomes listed 
in this definition. Examples of such medical events include allergic 
bronchospasm requiring intensive treatment in an emergency room or at 
home, blood dyscrasias or convulsions that do not result in inpatient 
hospitalization, or the development of drug dependency or drug abuse.
    Unexpected adverse drug experience: Any adverse drug experience, the 
specificity or severity of which is not consistent with the current 
investigator brochure; or, if an investigator brochure is not required 
or available, the specificity or severity of which is not consistent 
with the risk information described in the general investigational plan 
or elsewhere in the current application, as amended. For example, under 
this definition, hepatic necrosis would be unexpected (by virtue of 
greater severity) if the investigator brochure only referred to elevated 
hepatic enzymes or hepatitis. Similarly, cerebral thromboembolism and 
cerebral vasculitis would be unexpected (by virtue of greater 
specificity) if the investigator brochure only listed cerebral vascular 
accidents. ``Unexpected,'' as used in this definition, refers to an 
adverse drug experience that has not been previously observed (e.g., 
included in the investigator brochure) rather than from the perspective 
of such experience not being anticipated from the pharmacological 
properties of the pharmaceutical product.
    (b) Review of safety information. The sponsor shall promptly review 
all information relevant to the safety of the drug obtained or otherwise 
received by the sponsor from any source, foreign or domestic, including 
information derived from any clinical or epidemiological investigations, 
animal investigations, commercial marketing experience, reports in the 
scientific literature, and unpublished scientific papers, as well as 
reports from foreign regulatory authorities that have not already been 
previously reported to the agency by the sponsor.
    (c) IND safety reports. (1) Written reports--(i) The sponsor shall 
notify FDA and all participating investigators in a written IND safety 
report of:
    (A) Any adverse experience associated with the use of the drug that 
is both serious and unexpected; or
    (B) Any finding from tests in laboratory animals that suggests a 
significant risk for human subjects including reports of mutagenicity, 
teratogenicity, or carcinogenicity. Each notification shall be made as 
soon as possible and in no event later than 15 calendar days after the 
sponsor's initial receipt of the information. Each written notification 
may be submitted on FDA Form 3500A or in a narrative format (foreign 
events may be submitted either on an FDA Form 3500A or, if preferred, on 
a CIOMS I form; reports from animal or epidemiological studies shall be 
submitted in a narrative format) and shall bear prominent identification 
of its contents, i.e., ``IND Safety Report.'' Each written notification 
to FDA shall be transmitted to the FDA new drug review division in the 
Center for Drug Evaluation and Research or the product review division 
in the Center for Biologics Evaluation and Research that has 
responsibility

[[Page 74]]

for review of the IND. If FDA determines that additional data are 
needed, the agency may require further data to be submitted.
    (ii) In each written IND safety report, the sponsor shall identify 
all safety reports previously filed with the IND concerning a similar 
adverse experience, and shall analyze the significance of the adverse 
experience in light of the previouos, similar reports.
    (2) Telephone and facsimile transmission safety reports. The sponsor 
shall also notify FDA by telephone or by facsimile transmission of any 
unexpected fatal or life-threatening experience associated with the use 
of the drug as soon as possible but in no event later than 7 calendar 
days after the sponsor's initial receipt of the information. Each 
telephone call or facsimile transmission to FDA shall be transmitted to 
the FDA new drug review division in the Center for Drug Evaluation and 
Research or the product review division in the Center for Biologics 
Evaluation and Research that has responsibility for review of the IND.
    (3) Reporting format or frequency. FDA may request a sponsor to 
submit IND safety reports in a format or at a frequency different than 
that required under this paragraph. The sponsor may also propose and 
adopt a different reporting format or frequency if the change is agreed 
to in advance by the director of the new drug review division in the 
Center for Drug Evaluation and Research or the director of the products 
review division in the Center for Biologics Evaluation and Research 
which is responsible for review of the IND.
    (4) A sponsor of a clinical study of a marketed drug is not required 
to make a safety report for any adverse experience associated with use 
of the drug that is not from the clinical study itself.
    (d) Followup. (1) The sponsor shall promptly investigate all safety 
information received by it.
    (2) Followup information to a safety report shall be submitted as 
soon as the relevant information is available.
    (3) If the results of a sponsor's investigation show that an adverse 
drug experience not initially determined to be reportable under 
paragraph (c) of this section is so reportable, the sponsor shall report 
such experience in a written safety report as soon as possible, but in 
no event later than 15 calendar days after the determination is made.
    (4) Results of a sponsor's investigation of other safety information 
shall be submitted, as appropriate, in an information amendment or 
annual report.
    (e) Disclaimer. A safety report or other information submitted by a 
sponsor under this part (and any release by FDA of that report or 
information) does not necessarily reflect a conclusion by the sponsor or 
FDA that the report or information constitutes an admission that the 
drug caused or contributed to an adverse experience. A sponsor need not 
admit, and may deny, that the report or information submitted by the 
sponsor constitutes an admission that the drug caused or contributed to 
an adverse experience.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 55 
FR 11579, Mar. 29, 1990; 62 FR 52250, Oct. 7, 1997]

    Effective Date Note: At 62 FR 52250, Oct. 7, 1997, Sec. 312.32 was 
amended by revising paragraphs (a), (b), (c)(1)(i), (c)(2), and (d)(3); 
by adding in the second sentence of paragraph (c)(3) the words ``new 
drug review'' before the phrase ``division in the Center for Drug 
Evaluation and Research'' and the words ``the director of the product 
review division in'' before the phrase ``the Center for Biologics 
Evaluation and Research''; and by removing in paragraph (e) the word 
``section'' and replacing it with the word ``part'', effective Apr. 6, 
1998. For the convenience of the user, the superseded text is set forth 
as follows:

Sec. 312.32  IND safety reports.

    (a) Definitions. The following definitions of terms apply to this 
section:
    Associated with the use of the drug means that there is a reasonable 
possibility that the experience may have been caused by the drug.
    Serious adverse experience means any experience that suggests a 
significant hazard, contraindication, side effect, or precaution. With 
respect to human clinical experience, a serious adverse drug experience 
includes any experience that is fatal or life-threatening, is 
permanently disabling, requires inpatient hospitalization, or is a 
congenital anomaly, cancer, or overdose. With respect to results 
obtained from tests in laboratory animals, a

[[Page 75]]

serious adverse drug experience includes any experience suggesting a 
significant risk for human subjects, including any finding of 
mutagenicity, teratogenicity, or carcinogenicity.
    Unexpected adverse experience means any adverse experience that is 
not identified in nature, severity, or frequency in the current 
investigator brochure; or, if an investigator brochure is not required, 
that is not identified in nature, severity, or freuquency in the risk 
information described in the general investigational plan or elsewhere 
in the current application, as amended.
    (b) Review of safety information. The sponsor shall promptly review 
all information relevant to the safety of the drug obtained or otherwise 
received by the sponsor from any source, foreign or domestic, including 
information derived from clinical investigations, animal investigations, 
commercial marketing experience, reports in the scientific literature, 
and unpublished scientific papers.
    (c) IND safety reports. (1) Written reports. (i) The sponsor shall 
notify FDA and all participating investigators in a written IND safety 
report of any adverse experience associated with use of the drug that is 
both serious and unexpected. Such notification shall be made as soon as 
possible and in no event later than 10 working days after the sponsor's 
initial receipt of the information. Each written notification shall bear 
prominent identification of its contents, i.e., ``IND Safety Report.'' 
Each written notification to FDA shall be transmitted to the FDA 
division of the Center for Drug Evaluation and Research or the Center 
for Biologics Evaluation and Research which has responsibility for 
review of the IND.
    (ii)* * *
    (2) Telephone report. The sponsor shall also notify FDA by telephone 
of any unexpected fatal or life-threatening experience associated with 
use of the drug in the clinical studies conducted under the IND no later 
than 3 working days after receipt of the information. Each telephone 
call to FDA shall be transmitted to the FDA division of the Center for 
Drug Evaluation and Research or the Center for Biologics Evaluation and 
Research which has responsibility for review of the IND. For purposes of 
this section, life-threatening means that the patient was, in the view 
of the investigator, at immediate (emphasis added) risk of death from 
the reaction as it occurred, i.e., it does not include a reaction that, 
had it occurred in a more serious form, might have caused death. For 
example, drug-induced hepatitis that resolved without evidence of 
hepatic failure would not be considered life-threatening even though 
drug-induced hepatitis can be fatal.

                                * * * * *

    (d)* * *
    (3) If the results of a sponsor's investigation show that an adverse 
experience not initially determined to be reportable under paragraph (c) 
of this section is so reportable, the sponsor shall report such 
experience in a safety report as soon as possible after the 
determination is made, but in no event longer than 10-working days.

                                * * * * *



Sec. 312.33  Annual reports.

    A sponsor shall within 60 days of the anniversary date that the IND 
went into effect, submit a brief report of the progress of the 
investigation that includes:
    (a) Individual study information. A brief summary of the status of 
each study in progress and each study completed during the previous 
year. The summary is required to include the following information for 
each study:
    (1) The title of the study (with any appropriate study identifiers 
such as protocol number), its purpose, a brief statement identifying the 
patient population, and a statement as to whether the study is 
completed.
    (2) The total number of subjects initially planned for inclusion in 
the study; the number entered into the study to date, tabulated by age 
group, gender, and race; the number whose participation in the study was 
completed as planned; and the number who dropped out of the study for 
any reason.
    (3) If the study has been completed, or if interim results are 
known, a brief description of any available study results.
    (b) Summary information. Information obtained during the previous 
year's clinical and nonclinical investigations, including:
    (1) A narrative or tabular summary showing the most frequent and 
most serious adverse experiences by body system.
    (2) A summary of all IND safety reports submitted during the past 
year.
    (3) A list of subjects who died during participation in the 
investigation, with the cause of death for each subject.
    (4) A list of subjects who dropped out during the course of the 
investigation in association with any adverse experience, whether or not 
thought to be drug related.

[[Page 76]]

    (5) A brief description of what, if anything, was obtained that is 
pertinent to an understanding of the drug's actions, including, for 
example, information about dose response, information from controlled 
trails, and information about bioavailability.
    (6) A list of the preclinical studies (including animal studies) 
completed or in progress during the past year and a summary of the major 
preclinical findings.
    (7) A summary of any significant manufacturing or microbiological 
changes made during the past year.
    (c) A description of the general investigational plan for the coming 
year to replace that submitted 1 year earlier. The general 
investigational plan shall contain the information required under 
Sec. 312.23(a)(3)(iv).
    (d) If the investigator brochure has been revised, a description of 
the revision and a copy of the new brochure.
    (e) A description of any significant Phase 1 protocol modifications 
made during the previous year and not previously reported to the IND in 
a protocol amendment.
    (f) A brief summary of significant foreign marketing developments 
with the drug during the past year, such as approval of marketing in any 
country or withdrawal or suspension from marketing in any country.
    (g) If desired by the sponsor, a log of any outstanding business 
with respect to the IND for which the sponsor requests or expects a 
reply, comment, or meeting.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 63 
FR 6862, Feb. 11, 1998]

    Effective Date Note: At 63 FR 6862, Feb. 11, 1998, Sec. 312.33 was 
amended by revising paragraph (a)(2), effective Aug. 10, 1998. For the 
convenience of the user, the superseded text is set forth as follows:

Sec. 312.33  Annual reports.

                                * * * * *

    (a) * * *
    (2) The total number of subjects initially planned for inclusion in 
the study, the number entered into the study to date, the number whose 
participation in the study was completed as planned, and the number who 
dropped out of the study for any reason.

                                * * * * *



Sec. 312.34  Treatment use of an investigational new drug.

    (a) General. A drug that is not approved for marketing may be under 
clinical investigation for a serious or immediately life-threatening 
disease condition in patients for whom no comparable or satisfactory 
alternative drug or other therapy is available. During the clinical 
investigation of the drug, it may be appropriate to use the drug in the 
treatment of patients not in the clinical trials, in accordance with a 
treatment protocol or treatment IND. The purpose of this section is to 
facilitate the availability of promising new drugs to desperately ill 
patients as early in the drug development process as possible, before 
general marketing begins, and to obtain additional data on the drug's 
safety and effectiveness. In the case of a serious disease, a drug 
ordinarily may be made available for treatment use under this section 
during Phase 3 investigations or after all clinical trials have been 
completed; however, in appropriate circumstances, a drug may be made 
available for treatment use during Phase 2. In the case of an 
immediately life-threatening disease, a drug may be made available for 
treatment use under this section earlier than Phase 3, but ordinarily 
not earlier than Phase 2. For purposes of this section, the ``treatment 
use'' of a drug includes the use of a drug for diagnostic purposes. If a 
protocol for an investigational drug meets the criteria of this section, 
the protocol is to be submitted as a treatment protocol under the 
provisions of this section.
    (b) Criteria. (1) FDA shall permit an investigational drug to be 
used for a treatment use under a treatment protocol or treatment IND if:
    (i) The drug is intended to treat a serious or immediately life-
threatening disease;
    (ii) There is no comparable or satisfactory alternative drug or 
other therapy available to treat that stage of the

[[Page 77]]

disease in the intended patient population;
    (iii) The drug is under investigation in a controlled clinical trial 
under an IND in effect for the trial, or all clinical trials have been 
completed; and
    (iv) The sponsor of the controlled clinical trial is actively 
pursuing marketing approval of the investigational drug with due 
diligence.
    (2) Serious disease. For a drug intended to treat a serious disease, 
the Commissioner may deny a request for treatment use under a treatment 
protocol or treatment IND if there is insufficient evidence of safety 
and effectiveness to support such use.
    (3) Immediately life-threatening disease. (i) For a drug intended to 
treat an immediately life-threatening disease, the Commissioner may deny 
a request for treatment use of an investigational drug under a treatment 
protocol or treatment IND if the available scientific evidence, taken as 
a whole, fails to provide a reasonable basis for concluding that the 
drug:
    (A) May be effective for its intended use in its intended patient 
population; or
    (B) Would not expose the patients to whom the drug is to be 
administered to an unreasonable and significant additional risk of 
illness or injury.
    (ii) For the purpose of this section, an ``immediately life-
threatening'' disease means a stage of a disease in which there is a 
reasonable likelihood that death will occur within a matter of months or 
in which premature death is likely without early treatment.
    (c) Safeguards. Treatment use of an investigational drug is 
conditioned on the sponsor and investigators complying with the 
safeguards of the IND process, including the regulations governing 
informed consent (21 CFR part 50) and institutional review boards (21 
CFR part 56) and the applicable provisions of part 312, including 
distribution of the drug through qualified experts, maintenance of 
adequate manufacturing facilities, and submission of IND safety reports.
    (d) Clinical hold. FDA may place on clinical hold a proposed or 
ongoing treatment protocol or treatment IND in accordance with 
Sec. 312.42.
[52 FR 19476, May 22, 1987, as amended at 57 FR 13248, Apr. 15, 1992]



Sec. 312.35  Submissions for treatment use.

    (a) Treatment protocol submitted by IND sponsor. Any sponsor of a 
clinical investigation of a drug who intends to sponsor a treatment use 
for the drug shall submit to FDA a treatment protocol under Sec. 312.34 
if the sponsor believes the criteria of Sec. 312.34 are satisfied. If a 
protocol is not submitted under Sec. 312.34, but FDA believes that the 
protocol should have been submitted under this section, FDA may deem the 
protocol to be submitted under Sec. 312.34. A treatment use under a 
treatment protocol may begin 30 days after FDA receives the protocol or 
on earlier notification by FDA that the treatment use described in the 
protocol may begin.
    (1) A treatment protocol is required to contain the following:
    (i) The intended use of the drug.
    (ii) An explanation of the rationale for use of the drug, including, 
as appropriate, either a list of what available regimens ordinarily 
should be tried before using the investigational drug or an explanation 
of why the use of the investigational drug is preferable to the use of 
available marketed treatments.
    (iii) A brief description of the criteria for patient selection.
    (iv) The method of administration of the drug and the dosages.
    (v) A description of clinical procedures, laboratory tests, or other 
measures to monitor the effects of the drug and to minimize risk.
    (2) A treatment protocol is to be supported by the following:
    (i) Informational brochure for supplying to each treating physician.
    (ii) The technical information that is relevant to safety and 
effectiveness of the drug for the intended treatment purpose. 
Information contained in the sponsor's IND may be incorporated by 
reference.
    (iii) A commitment by the sponsor to assure compliance of all 
participating investigators with the informed consent requirements of 21 
CFR part 50.

[[Page 78]]

    (3) A licensed practioner who receives an investigational drug for 
treatment use under a treatment protocol is an ``investigator'' under 
the protocol and is responsible for meeting all applicable investigator 
responsibilities under this part and 21 CFR parts 50 and 56.
    (b) Treatment IND submitted by licensed practitioner. (1) If a 
licensed medical practitioner wants to obtain an investigational drug 
subject to a controlled clinical trial for a treatment use, the 
practitioner should first attempt to obtain the drug from the sponsor of 
the controlled trial under a treatment protocol. If the sponsor of the 
controlled clinical investigation of the drug will not establish a 
treatment protocol for the drug under paragraph (a) of this section, the 
licensed medical practitioner may seek to obtain the drug from the 
sponsor and submit a treatment IND to FDA requesting authorization to 
use the investigational drug for treatment use. A treatment use under a 
treatment IND may begin 30 days after FDA receives the IND or on earlier 
notification by FDA that the treatment use under the IND may begin. A 
treatment IND is required to contain the following:
    (i) A cover sheet (Form FDA 1571) meeting Sec. 312.23(g)(1).
    (ii) Information (when not provided by the sponsor) on the drug's 
chemistry, manufacturing, and controls, and prior clinical and 
nonclinical experience with the drug submitted in accordance with 
Sec. 312.23. A sponsor of a clinical investigation subject to an IND who 
supplies an investigational drug to a licensed medical practitioner for 
purposes of a separate treatment clinical investigation shall be deemed 
to authorize the incorporation-by-reference of the technical information 
contained in the sponsor's IND into the medical practitioner's treatment 
IND.
    (iii) A statement of the steps taken by the practitioner to obtain 
the drug under a treatment protocol from the drug sponsor.
    (iv) A treatment protocol containing the same information listed in 
paragraph (a)(1) of this section.
    (v) A statement of the practitioner's qualifications to use the 
investigational drug for the intended treatment use.
    (vi) The practitioner's statement of familiarity with information on 
the drug's safety and effectiveness derived from previous clinical and 
nonclinical experience with the drug.
    (vii) Agreement to report to FDA safety information in accordance 
with Sec. 312.32.
    (2) A licensed practitioner who submits a treatment IND under this 
section is the sponsor-investigator for such IND and is responsible for 
meeting all applicable sponsor and investigator responsibilities under 
this part and 21 CFR parts 50 and 56.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 19477, May 22, 1987, as amended at 57 FR 13249, Apr. 15, 1992]



Sec. 312.36  Emergency use of an investigational new drug.

    Need for an investigational drug may arise in an emergency situation 
that does not allow time for submission of an IND in accordance with 
Sec. 312.23 or Sec. 312.34. In such a case, FDA may authorize shipment 
of the drug for a specified use in advance of submission of an IND. A 
request for such authorization may be transmitted to FDA by telephone or 
other rapid communication means. For investigational biological drugs, 
the request should be directed to the Division of Biological 
Investigational New Drugs (HFB-230), Center for Biologics Evaluation and 
Research, 8800 Rockville Pike, Bethesda, MD 20892, 301-443-4864. For all 
other investigational drugs, the request for authorization should be 
directed to the Document Management and Reporting Branch (HFD-53), 
Center for Drug Evaluation and Research, 5600 Fishers Lane, Rockville, 
MD 20857, 301-443-4320. After normal working hours, eastern standard 
time, the request should be directed to the FDA Division of Emergency 
and Epidemiological Operations, 202-857-8400. Except in extraordinary 
circumstances, such authorization will be conditioned on the sponsor 
making an appropriate IND submission as soon

[[Page 79]]

as practicable after receiving the authorization.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 55 
FR 11579, Mar. 29, 1990]



Sec. 312.38  Withdrawal of an IND.

    (a) At any time a sponsor may withdraw an effective IND without 
prejudice.
    (b) If an IND is withdrawn, FDA shall be so notified, all clinical 
investigations conducted under the IND shall be ended, all current 
investigators notified, and all stocks of the drug returned to the 
sponsor or otherwise disposed of at the request of the sponsor in 
accordance with Sec. 312.59.
    (c) If an IND is withdrawn because of a safety reason, the sponsor 
shall promptly so inform FDA, all participating investigators, and all 
reviewing Institutional Review Boards, together with the reasons for 
such withdrawal.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987]



                    Subpart C--Administrative Actions



Sec. 312.40  General requirements for use of an investigational new drug in a clinical investigation.

    (a) An investigational new drug may be used in a clinical 
investigation if the following conditions are met:
    (1) The sponsor of the investigation submits an IND for the drug to 
FDA; the IND is in effect under paragraph (b) of this section; and the 
sponsor complies with all applicable requirements in this part and parts 
50 and 56 with respect to the conduct of the clinical investigations; 
and
    (2) Each participating investigator conducts his or her 
investigation in compliance with the requirements of this part and parts 
50 and 56.
    (b) An IND goes into effect:
    (1) Thirty days after FDA receives the IND, unless FDA notifies the 
sponsor that the investigations described in the IND are subject to a 
clinical hold under Sec. 312.42; or
    (2) On earlier notification by FDA that the clinical investigations 
in the IND may begin. FDA will notify the sponsor in writing of the date 
it receives the IND.
    (c) A sponsor may ship an investigational new drug to investigators 
named in the IND:
    (1) Thirty days after FDA receives the IND; or
    (2) On earlier FDA authorization to ship the drug.
    (d) An investigator may not administer an investigational new drug 
to human subjects until the IND goes into effect under paragraph (b) of 
this section.



Sec. 312.41  Comment and advice on an IND.

    (a) FDA may at any time during the course of the investigation 
communicate with the sponsor orally or in writing about deficiencies in 
the IND or about FDA's need for more data or information.
    (b) On the sponsor's request, FDA will provide advice on specific 
matters relating to an IND. Examples of such advice may include advice 
on the adequacy of technical data to support an investigational plan, on 
the design of a clinical trial, and on whether proposed investigations 
are likely to produce the data and information that is needed to meet 
requirements for a marketing application.
    (c) Unless the communication is accompanied by a clinical hold order 
under Sec. 312.42, FDA communications with a sponsor under this section 
are solely advisory and do not require any modification in the planned 
or ongoing clinical investigations or response to the agency.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987]



Sec. 312.42  Clinical holds and requests for modification.

    (a) General. A clinical hold is an order issued by FDA to the 
sponsor to delay a proposed clinical investigation or to

[[Page 80]]

suspend an ongoing investigation. The clinical hold order may apply to 
one or more of the investigations covered by an IND. When a proposed 
study is placed on clinical hold, subjects may not be given the 
investigational drug. When an ongoing study is placed on clinical hold, 
no new subjects may be recruited to the study and placed on the 
investigational drug; patients already in the study should be taken off 
therapy involving the investigational drug unless specifically permitted 
by FDA in the interest of patient safety.
    (b) Grounds for imposition of clinical hold--(1) Clinical hold of a 
Phase 1 study under an IND. FDA may place a proposed or ongoing Phase 1 
investigation on clinical hold if it finds that:
    (i) Human subjects are or would be exposed to an unreasonable and 
significant risk of illness or injury;
    (ii) The clinical investigators named in the IND are not qualified 
by reason of their scientific training and experience to conduct the 
investigation described in the IND;
    (iii) The investigator brochure is misleading, erroneous, or 
materially incomplete; or
    (iv) The IND does not contain sufficient information required under 
Sec. 312.23 to assess the risks to subjects of the proposed studies.
    (2) Clinical hold of a Phase 2 or 3 study under an IND. FDA may 
place a proposed or ongoing Phase 2 or 3 investigation on clinical hold 
if it finds that:
    (i) Any of the conditions in paragraph (b)(1)(i) through (iv) of 
this section apply; or
    (ii) The plan or protocol for the investigation is clearly deficient 
in design to meet its stated objectives.
    (3) Clinical hold of a treatment IND or treatment protocol.
    (i) Proposed use. FDA may place a proposed treatment IND or 
treatment protocol on clinical hold if it is determined that:
    (A) The pertinent criteria in Sec. 312.34(b) for permitting the 
treatment use to begin are not satisfied; or
    (B) The treatment protocol or treatment IND does not contain the 
information required under Sec. 312.35 (a) or (b) to make the specified 
determination under Sec. 312.34(b).
    (ii) Ongoing use. FDA may place an ongoing treatment protocol or 
treatment IND on clinical hold if it is determined that:
    (A) There becomes available a comparable or satisfactory alternative 
drug or other therapy to treat that stage of the disease in the intended 
patient population for which the investigational drug is being used;
    (B) The investigational drug is not under investigation in a 
controlled clinical trial under an IND in effect for the trial and not 
all controlled clinical trials necessary to support a marketing 
application have been completed, or a clinical study under the IND has 
been placed on clinical hold:
    (C) The sponsor of the controlled clinical trial is not pursuing 
marketing approval with due diligence;
    (D) If the treatment IND or treatment protocol is intended for a 
serious disease, there is insufficient evidence of safety and 
effectiveness to support such use; or
    (E) If the treatment protocol or treatment IND was based on an 
immediately life-threatening disease, the available scientific evidence, 
taken as a whole, fails to provide a reasonable basis for concluding 
that the drug:
    (1) May be effective for its intended use in its intended 
population; or
    (2) Would not expose the patients to whom the drug is to be 
administered to an unreasonable and significant additional risk of 
illness or injury.
    (iii) FDA may place a proposed or ongoing treatment IND or treatment 
protocol on clinical hold if it finds that any of the conditions in 
paragraph (b)(4)(i) through (b)(4)(viii) of this section apply.
    (4) Clinical hold of any study that is not designed to be adequate 
and well-controlled. FDA may place a proposed or ongoing investigation 
that is not designed to be adequate and well-controlled on clinical hold 
if it finds that:
    (i) Any of the conditions in paragraph (b)(1) or (b)(2) of this 
section apply; or
    (ii) There is reasonable evidence the investigation that is not 
designed to be adequate and well-controlled is impeding enrollment in, 
or otherwise interfering with the conduct or completion of, a study that 
is designed to be an

[[Page 81]]

adequate and well-controlled investigation of the same or another 
investigational drug; or
    (iii) Insufficient quantities of the investigational drug exist to 
adequately conduct both the investigation that is not designed to be 
adequate and well-controlled and the investigations that are designed to 
be adequate and well-controlled; or
    (iv) The drug has been studied in one or more adequate and well-
controlled investigations that strongly suggest lack of effectiveness; 
or
    (v) Another drug under investigation or approved for the same 
indication and available to the same patient population has demonstrated 
a better potential benefit/risk balance; or
    (vi) The drug has received marketing approval for the same 
indication in the same patient population; or
    (vii) The sponsor of the study that is designed to be an adequate 
and well-controlled investigation is not actively pursuing marketing 
approval of the investigational drug with due diligence; or
    (viii) The Commissioner determines that it would not be in the 
public interest for the study to be conducted or continued. FDA 
ordinarily intends that clinical holds under paragraphs (b)(4)(ii), 
(b)(4)(iii) and (b)(4)(v) of this section would only apply to additional 
enrollment in nonconcurrently controlled trials rather than eliminating 
continued access to individuals already receiving the investigational 
drug.
    (5) Clinical hold of any investigation involving an exception from 
informed consent under Sec. 50.24 of this chapter. FDA may place a 
proposed or ongoing investigation involving an exception from informed 
consent under Sec. 50.24 of this chapter on clinical hold if it is 
determined that:
    (i) Any of the conditions in paragraphs (b)(1) or (b)(2) of this 
section apply; or
    (ii) The pertinent criteria in Sec. 50.24 of this chapter for such 
an investigation to begin or continue are not submitted or not 
satisfied.
    (c) Discussion of deficiency. Whenever FDA concludes that a 
deficiency exists in a clinical investigation that may be grounds for 
the imposition of clinical hold FDA will, unless patients are exposed to 
immediate and serious risk, attempt to discuss and satisfactorily 
resolve the matter with the sponsor before issuing the clinical hold 
order.
    (d) Imposition of clinical hold. The clinical hold order may be made 
by telephone or other means of rapid communication or in writing. The 
clinical hold order will identify the studies under the IND to which the 
hold applies, and will briefly explain the basis for the action. The 
clinical hold order will be made by or on behalf of the Division 
Director with responsibility for review of the IND. As soon as possible, 
and no more than 30 days after imposition of the clinical hold, the 
Division Director will provide the sponsor a written explanation of the 
basis for the hold.
    (e) Resumption of clinical investigations. If, by the terms of the 
clinical hold order, resumption of the affected investigation is 
permitted without prior notification by FDA once a stated correction or 
modification is made, the investigation may proceed as soon as the 
correction or modification is made. In all other cases, an investigation 
may only resume after the Division Director (or the Director's designee) 
with responsibility for review of the IND has notified the sponsor that 
the investigation may proceed. In these cases resumption of the affected 
investigation(s) will be authorized when the sponsor corrects the 
deficiency(ies) previously cited or otherwise satisfied the agency that 
the investigation(s) can proceed. Resumption of a study may be 
authorized by telephone or other means of rapid communication.
    (f) Appeal. If the sponsor disagrees with the reasons cited for the 
clinical hold, the sponsor may request reconsideration of the decision 
in accordance with Sec. 312.48.
    (g) Conversion of IND on clinical hold to inactive status. If all 
investigations covered by an IND remain on clinical hold for 1 year or 
more, the IND may be placed on inactive status by FDA under Sec. 312.45.
[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 19477, May 22, 1987; 57 
FR 13249, Apr. 15, 1992; 61 FR 51530, Oct. 2, 1996]

[[Page 82]]



Sec. 312.44  Termination.

    (a) General. This section describes the procedures under which FDA 
may terminate an IND. If an IND is terminated, the sponsor shall end all 
clinical investigations conducted under the IND and recall or otherwise 
provide for the disposition of all unused supplies of the drug. A 
termination action may be based on deficiencies in the IND or in the 
conduct of an investigation under an IND. Except as provided in 
paragraph (d) of this section, a termination shall be preceded by a 
proposal to terminate by FDA and an opportunity for the sponsor to 
respond. FDA will, in general, only initiate an action under this 
section after first attempting to resolve differences informally or, 
when appropriate, through the clinical hold procedures described in 
Sec. 312.42.
    (b) Grounds for termination--(1) Phase 1. FDA may propose to 
terminate an IND during Phase 1 if it finds that:
    (i) Human subjects would be exposed to an unreasonable and 
significant risk of illness or unjury.
    (ii) The IND does not contain sufficient information required under 
Sec. 312.23 to assess the safety to subjects of the clinical 
investigations.
    (iii) The methods, facilities, and controls used for the 
manufacturing, processing, and packing of the investigational drug are 
inadequate to establish and maintain appropriate standards of identity, 
strength, quality, and purity as needed for subject safety.
    (iv) The clinical investigations are being conducted in a manner 
substantially different than that described in the protocols submitted 
in the IND.
    (v) The drug is being promoted or distributed for commercial 
purposes not justified by the requirements of the investigation or 
permitted by Sec. 312.7.
    (vi) The IND, or any amendment or report to the IND, contains an 
untrue statement of a material fact or omits material information 
required by this part.
    (vii) The sponsor fails promptly to investigate and inform the Food 
and Drug Administration and all investigators of serious and unexpected 
adverse experiences in accordance with Sec. 312.32 or fails to make any 
other report required under this part.
    (viii) The sponsor fails to submit an accurate annual report of the 
investigations in accordance with Sec. 312.33.
    (ix) The sponsor fails to comply with any other applicable 
requirement of this part, part 50, or part 56.
    (x) The IND has remained on inactive status for 5 years or more.
    (xi) The sponsor fails to delay a proposed investigation under the 
IND or to suspend an ongoing investigation that has been placed on 
clinical hold under Sec. 312.42(b)(4).
    (2) Phase 2 or 3. FDA may propose to terminate an IND during Phase 2 
or Phase 3 if FDA finds that:
    (i) Any of the conditions in paragraphs (b)(1)(i) through (b)(1)(xi) 
of this section apply; or
    (ii) The investigational plan or protocol(s) is not reasonable as a 
bona fide scientific plan to determine whether or not the drug is safe 
and effective for use; or
    (iii) There is convincing evidence that the drug is not effective 
for the purpose for which it is being investigated.
    (3) FDA may propose to terminate a treatment IND if it finds that:
    (i) Any of the conditions in paragraphs (b)(1)(i) through (x) of 
this section apply; or
    (ii) Any of the conditions in Sec. 312.42(b)(3) apply.
    (c) Opportunity for sponsor response. (1) If FDA proposes to 
terminate an IND, FDA will notify the sponsor in writing, and invite 
correction or explanation within a period of 30 days.
    (2) On such notification, the sponsor may provide a written 
explanation or correction or may request a conference with FDA to 
provide the requested explanation or correction. If the sponsor does not 
respond to the notification within the allocated time, the IND shall be 
terminated.
    (3) If the sponsor responds but FDA does not accept the explanation 
or correction submitted, FDA shall inform the sponsor in writing of the 
reason for the nonacceptance and provide the sponsor with an opportunity 
for a regulatory hearing before FDA under part 16 on the question of 
whether the IND should be terminated. The sponsor's request for a 
regulatory hearing must be made within 10 days of the sponsor's

[[Page 83]]

receipt of FDA's notification of nonacceptance.
    (d) Immediate termination of IND. Notwithstanding paragraphs (a) 
through (c) of this section, if at any time FDA concludes that 
continuation of the investigation presents an immediate and substantial 
danger to the health of individuals, the agency shall immediately, by 
written notice to the sponsor from the Director of the Center for Drug 
Evaluation and Research or the Director of the Center for Biologics 
Evaluation and Research, terminate the IND. An IND so terminated is 
subject to reinstatement by the Director on the basis of additional 
submissions that eliminate such danger. If an IND is terminated under 
this paragraph, the agency will afford the sponsor an opportunity for a 
regulatory hearing under part 16 on the question of whether the IND 
should be reinstated.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 55 
FR 11579, Mar. 29, 1990; 57 FR 13249, Apr. 15, 1992]



Sec. 312.45  Inactive status.

    (a) If no subjects are entered into clinical studies for a period of 
2 years or more under an IND, or if all investigations under an IND 
remain on clinical hold for 1 year or more, the IND may be placed by FDA 
on inactive status. This action may be taken by FDA either on request of 
the sponsor or on FDA's own initiative. If FDA seeks to act on its own 
initiative under this section, it shall first notify the sponsor in 
writing of the proposed inactive status. Upon receipt of such 
notification, the sponsor shall have 30 days to respond as to why the 
IND should continue to remain active.
    (b) If an IND is placed on inactive status, all investigators shall 
be so notified and all stocks of the drug shall be returned or otherwise 
disposed of in accordance with Sec. 312.59.
    (c) A sponsor is not required to submit annual reports to an IND on 
inactive status. An inactive IND is, however, still in effect for 
purposes of the public disclosure of data and information under 
Sec. 312.130.
    (d) A sponsor who intends to resume clinical investigation under an 
IND placed on inactive status shall submit a protocol amendment under 
Sec. 312.30 containing the proposed general investigational plan for the 
coming year and appropriate protocols. If the protocol amendment relies 
on information previously submitted, the plan shall reference such 
information. Additional information supporting the proposed 
investigation, if any, shall be submitted in an information amendment. 
Notwithstanding the provisions of Sec. 312.30, clinical investigations 
under an IND on inactive status may only resume (1) 30 days after FDA 
receives the protocol amendment, unless FDA notifies the sponsor that 
the investigations described in the amendment are subject to a clinical 
hold under Sec. 312.42, or (2) on earlier notification by FDA that the 
clinical investigations described in the protocol amendment may begin.
    (e) An IND that remains on inactive status for 5 years or more may 
be terminated under Sec. 312.44.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987]



Sec. 312.47  Meetings.

    (a) General. Meetings between a sponsor and the agency are 
frequently useful in resolving questions and issues raised during the 
course of a clinical investigation. FDA encourages such meetings to the 
extent that they aid in the evaluation of the drug and in the solution 
of scientific problems concerning the drug, to the extent that FDA's 
resources permit. The general principle underlying the conduct of such 
meetings is that there should be free, full, and open communication 
about any scientific or medical question that may arise during the 
clinical investigation. These meetings shall be conducted and documented 
in accordance with part 10.
    (b) ``End-of-Phase 2'' meetings and meetings held before submission 
of a marketing application. At specific times during the drug 
investigation process, meetings between FDA and a sponsor can be 
especially helpful in minimizing wasteful expenditures of time and

[[Page 84]]

money and thus in speeding the drug development and evaluation process. 
In particular, FDA has found that meetings at the end of Phase 2 of an 
investigation (end-of-Phase 2 meetings) are of considerable assistance 
in planning later studies and that meetings held near completion of 
Phase 3 and before submission of a marketing application (``pre-NDA'' 
meetings) are helpful in developing methods of presentation and 
submission of data in the marketing application that facilitate review 
and allow timely FDA response.
    (1) End-of-Phase 2 meetings--(i) Purpose. The purpose of an end-of-
Phase 2 meeting is to determine the safety of proceeding to Phase 3, to 
evaluate the Phase 3 plan and protocols, and to identify any additional 
information necessary to support a marketing application for the uses 
under investigation.
    (ii) Eligibility for meeting. While the end-of-Phase 2 meeting is 
designed primarily for IND's involving new molecular entities or major 
new uses of marketed drugs, a sponsor of any IND may request and obtain 
an end-of-Phase 2 meeting.
    (iii) Timing. To be most useful to the sponsor, end-of-Phase 2 
meetings should be held before major commitments of effort and resources 
to specific Phase 3 tests are made. The scheduling of an end-of-Phase 2 
meeting is not, however, intended to delay the transition of an 
investigation from Phase 2 to Phase 3.
    (iv) Advance information. At least 1 month in advance of an end-of-
Phase 2 meeting, the sponsor should submit background information on the 
sponsor's plan for Phase 3, including summaries of the Phase 1 and 2 
investigations, the specific protocols for Phase 3 clinical studies, 
plans for any additional nonclinical studies, and, if available, 
tentative labeling for the drug. The recommended contents of such a 
submission are described more fully in FDA Staff Manual Guide 4850.7 
that is publicly available under FDA's public information regulations in 
part 20.
    (v) Conduct of meeting. Arrangements for an end-of-Phase 2 meeting 
are to be made with the division in FDA's Center for Drug Evaluation and 
Research or the Center for Biologics Evaluation and Research which is 
responsible for review of the IND. The meeting will be scheduled by FDA 
at a time convenient to both FDA and the sponsor. Both the sponsor and 
FDA may bring consultants to the meeting. The meeting should be directed 
primarily at establishing agreement between FDA and the sponsor of the 
overall plan for Phase 3 and the objectives and design of particular 
studies. The adequacy of technical information to support Phase 3 
studies and/or a marketing application may also be discussed. Agreements 
reached at the meeting on these matters will be recorded in minutes of 
the conference that will be taken by FDA in accordance with Sec. 10.65 
and provided to the sponsor. The minutes along with any other written 
material provided to the sponsor will serve as a permanent record of any 
agreements reached. Barring a significant scientific development that 
requires otherwise, studies conducted in accordance with the agreement 
shall be presumed to be sufficient in objective and design for the 
purpose of obtaining marketing approval for the drug.
    (2) ``Pre-NDA'' meetings. FDA has found that delays associated with 
the initial review of a marketing application may be reduced by 
exchanges of information about a proposed marketing application. The 
primary purpose of this kind of exchange is to uncover any major 
unresolved problems, to identify those studies that the sponsor is 
relying on as adequate and well-controlled to establish the drug's 
effectiveness, to acquaint FDA reviewers with the general information to 
be submitted in the marketing application (including technical 
information), to discuss appropriate methods for statistical analysis of 
the data, and to discuss the best approach to the presentation and 
formatting of data in the marketing application. Arrangements for such a 
meeting are to be initiated by the sponsor with the division responsible 
for review of the IND. To permit FDA to provide the sponsor with the 
most useful advice on preparing a marketing application, the sponsor 
should submit to FDA's reviewing division at least 1 month in advance of 
the meeting the following information:

[[Page 85]]

    (i) A brief summary of the clinical studies to be submitted in the 
application.
    (ii) A proposed format for organizing the submission, including 
methods for presenting the data.
    (iii) Any other information for discussion at the meeting.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 55 
FR 11580, Mar. 29, 1990]



Sec. 312.48  Dispute resolution.

    (a) General. The Food and Drug Administration is committed to 
resolving differences between sponsors and FDA reviewing divisions with 
respect to requirements for IND's as quickly and amicably as possible 
through the cooperative exchange of information and views.
    (b) Administrative and procedural issues. When administrative or 
procedural disputes arise, the sponsor should first attempt to resolve 
the matter with the division in FDA's Center for Drug Evaluation and 
Research or Center for Biologics Evaluation and Research which is 
responsible for review of the IND, beginning with the consumer safety 
officer assigned to the application. If the dispute is not resolved, the 
sponsor may raise the matter with the person designated as ombudsman, 
whose function shall be to investigate what has happened and to 
facilitate a timely and equitable resolution. Appropriate issues to 
raise with the ombudsman include resolving difficulties in scheduling 
meetings and obtaining timely replies to inquiries. Further details on 
this procedure are contained in FDA Staff Manual Guide 4820.7 that is 
publicly available under FDA's public information regulations in part 
20.
    (c) Scientific and medical disputes. (1) When scientific or medical 
disputes arise during the drug investigation process, sponsors should 
discuss the matter directly with the responsible reviewing officials. If 
necessary, sponsors may request a meeting with the appropriate reviewing 
officials and management representatives in order to seek a resolution. 
Requests for such meetings shall be directed to the director of the 
division in FDA's Center for Drug Evaluation and Research or Center for 
Biologics Evaluation and Research which is responsible for review of the 
IND. FDA will make every attempt to grant requests for meetings that 
involve important issues and that can be scheduled at mutually 
convenient times.
    (2) The ``end-of-Phase 2'' and ``pre-NDA'' meetings described in 
Sec. 312.47(b) will also provide a timely forum for discussing and 
resolving scientific and medical issues on which the sponsor disagrees 
with the agency.
    (3) In requesting a meeting designed to resolve a scientific or 
medical dispute, applicants may suggest that FDA seek the advice of 
outside experts, in which case FDA may, in its discretion, invite to the 
meeting one or more of its advisory committee members or other 
consultants, as designated by the agency. Applicants may rely on, and 
may bring to any meeting, their own consultants. For major scientific 
and medical policy issues not resolved by informal meetings, FDA may 
refer the matter to one of its standing advisory committees for its 
consideration and recommendations.
[52 FR 8831, Mar. 19, 1987, as amended at 55 FR 11580, Mar. 29, 1990]



        Subpart D--Responsibilities of Sponsors and Investigators



Sec. 312.50  General responsibilities of sponsors.

    Sponsors are responsibile for selecting qualified investigators, 
providing them with the information they need to conduct an 
investigation properly, ensuring proper monitoring of the 
investigation(s), ensuring that the investigation(s) is conducted in 
accordance with the general investigational plan and protocols contained 
in the IND, maintaining an effective IND with respect to the 
investigations, and ensuring that FDA and all participating 
investigators are promptly informed of significant new adverse effects 
or risks with respect to the drug. Additional specific responsibilities 
of sponsors are described elsewhere in this part.

[[Page 86]]



Sec. 312.52  Transfer of obligations to a contract research organization.

    (a) A sponsor may transfer responsibility for any or all of the 
obligations set forth in this part to a contract research organization. 
Any such transfer shall be described in writing. If not all obligations 
are transferred, the writing is required to describe each of the 
obligations being assumed by the contract research organization. If all 
obligations are transferred, a general statement that all obligations 
have been transferred is acceptable. Any obligation not covered by the 
written description shall be deemed not to have been transferred.
    (b) A contract research organization that assumes any obligation of 
a sponsor shall comply with the specific regulations in this chapter 
applicable to this obligation and shall be subject to the same 
regulatory action as a sponsor for failure to comply with any obligation 
assumed under these regulations. Thus, all references to ``sponsor'' in 
this part apply to a contract research organization to the extent that 
it assumes one or more obligations of the sponsor.



Sec. 312.53  Selecting investigators and monitors.

    (a) Selecting investigators. A sponsor shall select only 
investigators qualified by training and experience as appropriate 
experts to investigate the drug.
    (b) Control of drug. A sponsor shall ship investigational new drugs 
only to investigators participating in the investigation.
    (c) Obtaining information from the investigator. Before permitting 
an investigator to begin participation in an investigation, the sponsor 
shall obtain the following:
    (1) A signed investigator statement (Form FDA-1572) containing:
    (i) The name and address of the investigator;
    (ii) The name and code number, if any, of the protocol(s) in the IND 
identifying the study(ies) to be conducted by the investigator;
    (iii) The name and address of any medical school, hospital, or other 
research facility where the clinical investigation(s) will be conducted;
    (iv) The name and address of any clinical laboratory facilities to 
be used in the study;
    (v) The name and address of the IRB that is responsible for review 
and approval of the study(ies);
    (vi) A commitment by the investigator that he or she:
    (a) Will conduct the study(ies) in accordance with the relevant, 
current protocol(s) and will only make changes in a protocol after 
notifying the sponsor, except when necessary to protect the safety, the 
rights, or welfare of subjects;
    (b) Will comply with all requirements regarding the obligations of 
clinical investigators and all other pertinent requirements in this 
part;
    (c) Will personally conduct or supervise the described 
investigation(s);
    (d) Will inform any potential subjects that the drugs are being used 
for investigational purposes and will ensure that the requirements 
relating to obtaining informed consent (21 CFR part 50) and 
institutional review board review and approval (21 CFR part 56) are met;
    (e) Will report to the sponsor adverse experiences that occur in the 
course of the investigation(s) in accordance with Sec. 312.64;
    (f) Has read and understands the information in the investigator's 
brochure, including the potential risks and side effects of the drug; 
and
    (g) Will ensure that all associates, colleagues, and employees 
assisting in the conduct of the study(ies) are informed about their 
obligations in meeting the above commitments.
    (vii) A commitment by the investigator that, for an investigation 
subject to an institutional review requirement under part 56, an IRB 
that complies with the requirements of that part will be responsible for 
the initial and continuing review and approval of the clinical 
investigation and that the investigator will promptly report to the IRB 
all changes in the research activity and all unanticipated problems 
involving risks to human subjects or others, and will not make any 
changes in the research without IRB approval, except where necessary to 
eliminate apparent immediate hazards to the human subjects.

[[Page 87]]

    (viii) A list of the names of the subinvestigators (e.g., research 
fellows, residents) who will be assisting the investigator in the 
conduct of the investigation(s).
    (2) Curriculum vitae. A curriculum vitae or other statement of 
qualifications of the investigator showing the education, training, and 
experience that qualifies the investigator as an expert in the clinical 
investigation of the drug for the use under investigation.
    (3) Clinical protocol. (i) For Phase 1 investigations, a general 
outline of the planned investigation including the estimated duration of 
the study and the maximum number of subjects that will be involved.
    (ii) For Phase 2 or 3 investigations, an outline of the study 
protocol including an approximation of the number of subjects to be 
treated with the drug and the number to be employed as controls, if any; 
the clinical uses to be investigated; characteristics of subjects by 
age, sex, and condition; the kind of clinical observations and 
laboratory tests to be conducted; the estimated duration of the study; 
and copies or a description of case report forms to be used.
    (4) Financial disclosure information. Sufficient accurate financial 
information to allow the sponsor to submit complete and accurate 
certification or disclosure statements required under part 54 of this 
chapter. The sponsor shall obtain a commitment from the clinical 
investigator to promptly update this information if any relevant changes 
occur during the course of the investigation and for 1 year following 
the completion of the study.
    (d) Selecting monitors. A sponsor shall select a monitor qualified 
by training and experience to monitor the progress of the investigation.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 61 
FR 57280, Nov. 5, 1996; 63 FR 5252, Feb. 2, 1998]

    Effective Date Note: At 63 FR 5252, Feb. 2, 1998, Sec. 312.53 was 
amended by adding new paragraph (c)(4), effective Feb. 2, 1999.



Sec. 312.54  Emergency research under Sec. 50.24 of this chapter.

    (a) The sponsor shall monitor the progress of all investigations 
involving an exception from informed consent under Sec. 50.24 of this 
chapter. When the sponsor receives from the IRB information concerning 
the public disclosures required by Sec. 50.24(a)(7)(ii) and (a)(7)(iii) 
of this chapter, the sponsor promptly shall submit to the IND file and 
to Docket Number 95S-0158 in the Dockets Management Branch (HFA-305), 
Food and Drug Administration, 12420 Parklawn Dr., rm. 1-23, Rockville, 
MD 20857, copies of the information that was disclosed, identified by 
the IND number.
    (b) The sponsor also shall monitor such investigations to identify 
when an IRB determines that it cannot approve the research because it 
does not meet the criteria in the exception in Sec. 50.24(a) of this 
chapter or because of other relevant ethical concerns. The sponsor 
promptly shall provide this information in writing to FDA, investigators 
who are asked to participate in this or a substantially equivalent 
clinical investigation, and other IRB's that are asked to review this or 
a substantially equivalent investigation.
[61 FR 51530, Oct. 2, 1996]



Sec. 312.55  Informing investigators.

    (a) Before the investigation begins, a sponsor (other than a 
sponsor-investigator) shall give each participating clinical 
investigator an investigator brochure containing the information 
described in Sec. 312.23(a)(5).
    (b) The sponsor shall, as the overall investigation proceeds, keep 
each participating investigator informed of new observations discovered 
by or reported to the sponsor on the drug, particularly with respect to 
adverse effects and safe use. Such information may be distributed to 
investigators by means of periodically revised investigator brochures, 
reprints or published studies, reports or letters to clinical 
investigators, or other appropriate means.

[[Page 88]]

Important safety information is required to be relayed to investigators 
in accordance with Sec. 312.32.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987]



Sec. 312.56  Review of ongoing investigations.

    (a) The sponsor shall monitor the progress of all clinical 
investigations being conducted under its IND.
    (b) A sponsor who discovers that an investigator is not complying 
with the signed agreement (Form FDA-1572), the general investigational 
plan, or the requirements of this part or other applicable parts shall 
promptly either secure compliance or discontinue shipments of the 
investigational new drug to the investigator and end the investigator's 
participation in the investigation. If the investigator's participation 
in the investigation is ended, the sponsor shall require that the 
investigator dispose of or return the investigational drug in accordance 
with the requirements of Sec. 312.59 and shall notify FDA.
    (c) The sponsor shall review and evaluate the evidence relating to 
the safety and effectiveness of the drug as it is obtained from the 
investigator. The sponsors shall make such reports to FDA regarding 
information relevant to the safety of the drug as are required under 
Sec. 312.32. The sponsor shall make annual reports on the progress of 
the investigation in accordance with Sec. 312.33.
    (d) A sponsor who determines that its investigational drug presents 
an unreasonable and significant risk to subjects shall discontinue those 
investigations that present the risk, notify FDA, all institutional 
review boards, and all investigators who have at any time participated 
in the investigation of the discontinuance, assure the disposition of 
all stocks of the drug outstanding as required by Sec. 312.59, and 
furnish FDA with a full report of the sponsor's actions. The sponsor 
shall discontinue the investigation as soon as possible, and in no event 
later than 5 working days after making the determination that the 
investigation should be discontinued. Upon request, FDA will confer with 
a sponsor on the need to discontinue an investigation.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987]



Sec. 312.57  Recordkeeping and record retention.

    (a) A sponsor shall maintain adequate records showing the receipt, 
shipment, or other disposition of the investigational drug. These 
records are required to include, as appropriate, the name of the 
investigator to whom the drug is shipped, and the date, quantity, and 
batch or code mark of each such shipment.
    (b) A sponsor shall maintain complete and accurate records showing 
any financial interest in Sec. 54.4(a)(3)(i), (a)(3)(ii), (a)(3)(iii), 
and (a)(3)(iv) of this chapter paid to clinical investigators by the 
sponsor of the covered study. A sponsor shall also maintain complete and 
accurate records concerning all other financial interests of 
investigators subject to part 54 of this chapter.
    (c) A sponsor shall retain the records and reports required by this 
part for 2 years after a marketing application is approved for the drug; 
or, if an application is not approved for the drug, until 2 years after 
shipment and delivery of the drug for investigational use is 
discontinued and FDA has been so notified.
    (d) A sponsor shall retain reserve samples of any test article and 
reference standard identified in, and used in any of the bioequivalence 
or bioavailability studies described in, Sec. 320.38 or Sec. 320.63 of 
this chapter, and release the reserve samples to FDA upon request, in 
accordance with, and for the period specified in Sec. 320.38.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 58 
FR 25926, Apr. 28, 1993; 63 FR 5252, Feb. 2, 1998]

    Effective Date Note: At 63 FR 5252, Feb. 2, 1998, Sec. 312.57 was 
amended by redesignating paragraphs (b) and (c) as paragraphs (c) and

[[Page 89]]

(d) and by adding a new paragraph (b), effective Feb. 2, 1999.



Sec. 312.58  Inspection of sponsor's records and reports.

    (a) FDA inspection. A sponsor shall upon request from any properly 
authorized officer or employee of the Food and Drug Administration, at 
reasonable times, permit such officer or employee to have access to and 
copy and verify any records and reports relating to a clinical 
investigation conducted under this part. Upon written request by FDA, 
the sponsor shall submit the records or reports (or copies of them) to 
FDA. The sponsor shall discontinue shipments of the drug to any 
investigator who has failed to maintain or make available records or 
reports of the investigation as required by this part.
    (b) Controlled substances. If an investigational new drug is a 
substance listed in any schedule of the Controlled Substances Act (21 
U.S.C. 801; 21 CFR part 1308), records concerning shipment, delivery, 
receipt, and disposition of the drug, which are required to be kept 
under this part or other applicable parts of this chapter shall, upon 
the request of a properly authorized employee of the Drug Enforcement 
Administration of the U.S. Department of Justice, be made available by 
the investigator or sponsor to whom the request is made, for inspection 
and copying. In addition, the sponsor shall assure that adequate 
precautions are taken, including storage of the investigational drug in 
a securely locked, substantially constructed cabinet, or other securely 
locked, substantially constructed enclosure, access to which is limited, 
to prevent theft or diversion of the substance into illegal channels of 
distribution.



Sec. 312.59  Disposition of unused supply of investigational drug.

    The sponsor shall assure the return of all unused supplies of the 
investigational drug from each individual investigator whose 
participation in the investigation is discontinued or terminated. The 
sponsor may authorize alternative disposition of unused supplies of the 
investigational drug provided this alternative disposition does not 
expose humans to risks from the drug. The sponsor shall maintain written 
records of any disposition of the drug in accordance with Sec. 312.57.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987]



Sec. 312.60  General responsibilities of investigators.

    An investigator is responsible for ensuring that an investigation is 
conducted according to the signed investigator statement, the 
investigational plan, and applicable regulations; for protecting the 
rights, safety, and welfare of subjects under the investigator's care; 
and for the control of drugs under investigation. An investigator shall, 
in accordance with the provisions of part 50 of this chapter, obtain the 
informed consent of each human subject to whom the drug is administered, 
except as provided in Secs. 50.23 or 50.24 of this chapter. Additional 
specific responsibilities of clinical investigators are set forth in 
this part and in parts 50 and 56 of this chapter.
[52 FR 8831, Mar. 19, 1987, as amended at 61 FR 51530, Oct. 2, 1996]



Sec. 312.61  Control of the investigational drug.

    An investigator shall administer the drug only to subjects under the 
investigator's personal supervision or under the supervision of a 
subinvestigator responsible to the investigator. The investigator shall 
not supply the investigational drug to any person not authorized under 
this part to receive it.



Sec. 312.62  Investigator recordkeeping and record retention.

    (a) Disposition of drug. An investigator is required to maintain 
adequate records of the disposition of the drug, including dates, 
quantity, and use by subjects. If the investigation is terminated, 
suspended, discontinued, or completed, the investigator shall return the 
unused supplies of the drug to the sponsor, or otherwise provide for 
disposition of the unused supplies of the drug under Sec. 312.59.

[[Page 90]]

    (b) Case histories. An investigator is required to prepare and 
maintain adequate and accurate case histories that record all 
observations and other data pertinent to the investigation on each 
individual administered the investigational drug or employed as a 
control in the investigation. Case histories include the case report 
forms and supporting data including, for example, signed and dated 
consent forms and medical records including, for example, progress notes 
of the physician, the individual's hospital chart(s), and the nurses' 
notes. The case history for each individual shall document that informed 
consent was obtained prior to participation in the study.
    (c) Record retention. An investigator shall retain records required 
to be maintained under this part for a period of 2 years following the 
date a marketing application is approved for the drug for the indication 
for which it is being investigated; or, if no application is to be filed 
or if the application is not approved for such indication, until 2 years 
after the investigation is discontinued and FDA is notified.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 61 
FR 57280, Nov. 5, 1996]



Sec. 312.64  Investigator reports.

    (a) Progress reports. The investigator shall furnish all reports to 
the sponsor of the drug who is responsible for collecting and evaluating 
the results obtained. The sponsor is required under Sec. 312.33 to 
submit annual reports to FDA on the progress of the clinical 
investigations.
    (b) Safety reports. An investigator shall promptly report to the 
sponsor any adverse effect that may reasonably be regarded as caused by, 
or probably caused by, the drug. If the adverse effect is alarming, the 
investigator shall report the adverse effect immediately.
    (c) Final report. An investigator shall provide the sponsor with an 
adequate report shortly after completion of the investigator's 
participation in the investigation.
    (d) Financial disclosure reports. The clinical investigator shall 
provide the sponsor with sufficient accurate financial information to 
allow an applicant to submit complete and accurate certification or 
disclosure statements as required under part 54 of this chapter. The 
clinical investigator shall promptly update this information if any 
relevant changes occur during the course of the investigation and for 1 
year following the completion of the study.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 63 
FR 5252, Feb. 2, 1998]

    Effective Date Note: At 63 FR 5252, Feb. 2, 1998, Sec. 312.64 was 
amended by adding new paragraph (d), effective Feb. 2, 1999.



Sec. 312.66  Assurance of IRB review.

    An investigator shall assure that an IRB that complies with the 
requirements set forth in part 56 will be responsible for the initial 
and continuing review and approval of the proposed clinical study. The 
investigator shall also assure that he or she will promptly report to 
the IRB all changes in the research activity and all unanticipated 
problems involving risk to human subjects or others, and that he or she 
will not make any changes in the research without IRB approval, except 
where necessary to eliminate apparent immediate hazards to human 
subjects.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987]



Sec. 312.68  Inspection of investigator's records and reports.

    An investigator shall upon request from any properly authorized 
officer or employee of FDA, at reasonable times, permit such officer or 
employee to have access to, and copy and verify any records or reports 
made by the investigator pursuant to Sec. 312.62. The investigator is 
not required to divulge subject names unless the records of particular 
individuals require a more detailed study of the cases, or unless

[[Page 91]]

there is reason to believe that the records do not represent actual case 
studies, or do not represent actual results obtained.



Sec. 312.69  Handling of controlled substances.

    If the investigational drug is subject to the Controlled Substances 
Act, the investigator shall take adequate precautions, including storage 
of the investigational drug in a securely locked, substantially 
constructed cabinet, or other securely locked, substantially constructed 
enclosure, access to which is limited, to prevent theft or diversion of 
the substance into illegal channels of distribution.



Sec. 312.70  Disqualification of a clinical investigator.

    (a) If FDA has information indicating that an investigator 
(including a sponsor-investigator) has repeatedly or deliberately failed 
to comply with the requirements of this part, part 50, or part 56 of 
this chapter, or has submitted to FDA or to the sponsor false 
information in any required report, the Center for Drug Evaluation and 
Research or the Center for Biologics Evaluation and Research will 
furnish the investigator written notice of the matter complained of and 
offer the investigator an opportunity to explain the matter in writing, 
or, at the option of the investigator, in an informal conference. If an 
explanation is offered but not accepted by the Center for Drug 
Evaluation and Research or the Center for Biologics Evaluation and 
Research, the investigator will be given an opportunity for a regulatory 
hearing under part 16 on the question of whether the investigator is 
entitled to receive investigational new drugs.
    (b) After evaluating all available information, including any 
explanation presented by the investigator, if the Commissioner 
determines that the investigator has repeatedly or deliberately failed 
to comply with the requirements of this part, part 50, or part 56 of 
this chapter, or has deliberately or repeatedly submitted false 
information to FDA or to the sponsor in any required report, the 
Commissioner will notify the investigator and the sponsor of any 
investigation in which the investigator has been named as a participant 
that the investigator is not entitled to receive investigational drugs. 
The notification will provide a statement of basis for such 
determination.
    (c) Each IND and each approved application submitted under part 314 
containing data reported by an investigator who has been determined to 
be ineligible to receive investigational drugs will be examined to 
determine whether the investigator has submitted unreliable data that 
are essential to the continuation of the investigation or essential to 
the approval of any marketing application.
    (d) If the Commissioner determines, after the unreliable data 
submitted by the investigator are eliminated from consideration, that 
the data remaining are inadequate to support a conclusion that it is 
reasonably safe to continue the investigation, the Commissioner will 
notify the sponsor who shall have an opportunity for a regulatory 
hearing under part 16. If a danger to the public health exists, however, 
the Commissioner shall terminate the IND immediately and notify the 
sponsor of the determination. In such case, the sponsor shall have an 
opportunity for a regulatory hearing before FDA under part 16 on the 
question of whether the IND should be reinstated.
    (e) If the Commissioner determines, after the unreliable data 
submitted by the investigator are eliminated from consideration, that 
the continued approval of the drug product for which the data were 
submitted cannot be justified, the Commissioner will proceed to withdraw 
approval of the drug product in accordance with the applicable 
provisions of the act.
    (f) An investigator who has been determined to be ineligible to 
receive investigational drugs may be reinstated as eligible when the 
Commissioner determines that the investigator has presented adequate 
assurances that the investigator will employ investigatioal drugs solely 
in compliance with the

[[Page 92]]

provisions of this part and of parts 50 and 56.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 55 
FR 11580, Mar. 29, 1990; 62 FR 46876, Sept. 5, 1997]



    Subpart E--Drugs Intended to Treat Life-threatening and Severely-
                         debilitating Illnesses

    Authority:  Secs. 501, 502, 503, 505, 506, 507, 701, 52 Stat. 1049-
1053 as amended, 1055-1056 as amended, 55 Stat. 851, 59 Stat. 463 as 
amended (21 U.S.C. 351, 352, 353, 355, 356, 357, 371); sec. 351, 58 
Stat. 702 as amended (42 U.S.C. 262); 21 CFR 5.10, 5.11.

    Source:  53 FR 41523, Oct. 21, 1988, unless otherwise noted.



Sec. 312.80  Purpose.

    The purpose of this section is to establish procedures designed to 
expedite the development, evaluation, and marketing of new therapies 
intended to treat persons with life-threatening and severely-
debilitating illnesses, especially where no satisfactory alternative 
therapy exists. As stated Sec. 314.105(c) of this chapter, while the 
statutory standards of safety and effectiveness apply to all drugs, the 
many kinds of drugs that are subject to them, and the wide range of uses 
for those drugs, demand flexibility in applying the standards. The Food 
and Drug Administration (FDA) has determined that it is appropriate to 
exercise the broadest flexibility in applying the statutory standards, 
while preserving appropriate guarantees for safety and effectiveness. 
These procedures reflect the recognition that physicians and patients 
are generally willing to accept greater risks or side effects from 
products that treat life-threatening and severely-debilitating 
illnesses, than they would accept from products that treat less serious 
illnesses. These procedures also reflect the recognition that the 
benefits of the drug need to be evaluated in light of the severity of 
the disease being treated. The procedure outlined in this section should 
be interpreted consistent with that purpose.



Sec. 312.81  Scope.

    This section applies to new drug, antibiotic, and biological 
products that are being studied for their safety and effectiveness in 
treating life-threatening or severely-debilitating diseases.
    (a) For purposes of this section, the term ``life-threatening'' 
means:
    (1) Diseases or conditions where the likelihood of death is high 
unless the course of the disease is interrupted; and
    (2) Diseases or conditions with potentially fatal outcomes, where 
the end point of clinical trial analysis is survival.
    (b) For purposes of this section, the term ``severely debilitating'' 
means diseases or conditions that cause major irreversible morbidity.
    (c) Sponsors are encouraged to consult with FDA on the applicability 
of these procedures to specific products.



Sec. 312.82  Early consultation.

    For products intended to treat life-threatening or severely-
debilitating illnesses, sponsors may request to meet with FDA-reviewing 
officials early in the drug development process to review and reach 
agreement on the design of necessary preclinical and clinical studies. 
Where appropriate, FDA will invite to such meetings one or more outside 
expert scientific consultants or advisory committee members. To the 
extent FDA resources permit, agency reviewing officials will honor 
requests for such meetings
    (a) Pre-investigational new drug (IND) meetings. Prior to the 
submission of the initial IND, the sponsor may request a meeting with 
FDA-reviewing officials. The primary purpose of this meeting is to 
review and reach agreement on the design of animal studies needed to 
initiate human testing. The meeting may also provide an opportunity for 
discussing the scope and design of phase 1 testing, and the best 
approach for presentation and formatting of data in the IND.
    (b) End-of-phase 1 meetings. When data from phase 1 clinical testing 
are available, the sponsor may again request a meeting with FDA-
reviewing officials. The primary purpose of this meeting is to review 
and reach agreement on the design of phase 2 controlled clinical

[[Page 93]]

trials, with the goal that such testing will be adequate to provide 
sufficient data on the drug's safety and effectiveness to support a 
decision on its approvability for marketing. The procedures outlined in 
Sec. 312.47(b)(1) with respect to end-of-phase 2 conferences, including 
documentation of agreements reached, would also be used for end-of-phase 
1 meetings.



Sec. 312.83  Treatment protocols.

    If the preliminary analysis of phase 2 test results appears 
promising, FDA may ask the sponsor to submit a treatment protocol to be 
reviewed under the procedures and criteria listed in Secs. 312.34 and 
312.35. Such a treatment protocol, if requested and granted, would 
normally remain in effect while the complete data necessary for a 
marketing application are being assembled by the sponsor and reviewed by 
FDA (unless grounds exist for clinical hold of ongoing protocols, as 
provided in Sec. 312.42(b)(3)(ii)).



Sec. 312.84  Risk-benefit analysis in review of marketing applications for drugs to treat life-threatening and severely-debilitating illnesses.

    (a) FDA's application of the statutory standards for marketing 
approval shall recognize the need for a medical risk-benefit judgment in 
making the final decision on approvability. As part of this evaluation, 
consistent with the statement of purpose in Sec. 312.80, FDA will 
consider whether the benefits of the drug outweigh the known and 
potential risks of the drug and the need to answer remaining questions 
about risks and benefits of the drug, taking into consideration the 
severity of the disease and the absence of satisfactory alternative 
therapy.
    (b) In making decisions on whether to grant marketing approval for 
products that have been the subject of an end-of-phase 1 meeting under 
Sec. 312.82, FDA will usually seek the advice of outside expert 
scientific consultants or advisory committees. Upon the filing of such a 
marketing application under Sec. 314.101 or part 601 of this chapter, 
FDA will notify the members of the relevant standing advisory committee 
of the application's filing and its availability for review.
    (c) If FDA concludes that the data presented are not sufficient for 
marketing approval, FDA will issue (for a drug) a not approvable letter 
pursuant to Sec. 314.120 of this chapter, or (for a biologic) a 
deficiencies letter consistent with the biological product licensing 
procedures. Such letter, in describing the deficiencies in the 
application, will address why the results of the research design agreed 
to under Sec. 312.82, or in subsequent meetings, have not provided 
sufficient evidence for marketing approval. Such letter will also 
describe any recommendations made by the advisory committee regarding 
the application.
    (d) Marketing applications submitted under the procedures contained 
in this section will be subject to the requirements and procedures 
contained in part 314 or part 600 of this chapter, as well as those in 
this subpart.



Sec. 312.85  Phase 4 studies.

    Concurrent with marketing approval, FDA may seek agreement from the 
sponsor to conduct certain postmarketing (phase 4) studies to delineate 
additional information about the drug's risks, benefits, and optimal 
use. These studies could include, but would not be limited to, studying 
different doses or schedules of administration than were used in phase 2 
studies, use of the drug in other patient populations or other stages of 
the disease, or use of the drug over a longer period of time.



Sec. 312.86  Focused FDA regulatory research.

    At the discretion of the agency, FDA may undertake focused 
regulatory research on critical rate-limiting aspects of the 
preclinical, chemical/manufacturing, and clinical phases of drug 
development and evaluation. When initiated, FDA will undertake such 
research efforts as a means for meeting a public health need in 
facilitating the development of therapies to treat life-threatening or 
severely debilitating illnesses.

[[Page 94]]



Sec. 312.87  Active monitoring of conduct and evaluation of clinical trials.

    For drugs covered under this section, the Commissioner and other 
agency officials will monitor the progress of the conduct and evaluation 
of clinical trials and be involved in facilitating their appropriate 
progress.



Sec. 312.88  Safeguards for patient safety.

    All of the safeguards incorporated within parts 50, 56, 312, 314, 
and 600 of this chapter designed to ensure the safety of clinical 
testing and the safety of products following marketing approval apply to 
drugs covered by this section. This includes the requirements for 
informed consent (part 50 of this chapter) and institutional review 
boards (part 56 of this chapter). These safeguards further include the 
review of animal studies prior to initial human testing (Sec. 312.23), 
and the monitoring of adverse drug experiences through the requirements 
of IND safety reports (Sec. 312.32), safety update reports during agency 
review of a marketing application (Sec. 314.50 of this chapter), and 
postmarketing adverse reaction reporting (Sec. 314.80 of this chapter).



                        Subpart F--Miscellaneous



Sec. 312.110  Import and export requirements.

    (a) Imports. An investigational new drug offered for import into the 
United States complies with the requirements of this part if it is 
subject to an IND that is in effect for it under Sec. 312.40 and: (1) 
The consignee in the United States is the sponsor of the IND; (2) the 
consignee is a qualified investigator named in the IND; or (3) the 
consignee is the domestic agent of a foreign sponsor, is responsible for 
the control and distribution of the investigational drug, and the IND 
identifies the consignee and describes what, if any, actions the 
consignee will take with respect to the investigational drug.
    (b) Exports. An investigational new drug intended for export from 
the United States complies with the requirements of this part as 
follows:
    (1) If an IND is in effect for the drug under Sec. 312.40 and each 
person who receives the drug is an investigator named in the 
application; or
    (2) If FDA authorizes shipment of the drug for use in a clinical 
investigation. Authorization may be obtained as follows:
    (i) Through submission to the International Affairs Staff (HFY-50), 
Associate Commissioner for Health Affairs, Food and Drug Administration, 
5600 Fishers Lane, Rockville, MD 20857, of a written request from the 
person that seeks to export the drug. A request must provide adequate 
information about the drug to satisfy FDA that the drug is appropriate 
for the proposed investigational use in humans, that the drug will be 
used for investigational purposes only, and that the drug may be legally 
used by that consignee in the importing country for the proposed 
investigational use. The request shall specify the quantity of the drug 
to be shipped per shipment and the frequency of expected shipments. If 
FDA authorizes exportation under this paragraph, the agency shall 
concurrently notify the government of the importing country of such 
authorization.
    (ii) Through submission to the International Affairs Staff (HFY-50), 
Associate Commissioner for Health Affairs, Food and Drug Administration, 
5600 Fishers Lane, Rockville, MD 20857, of a formal request from an 
authorized official of the government of the country to which the drug 
is proposed to be shipped. A request must specify that the foreign 
government has adequate information about the drug and the proposed 
investigational use, that the drug will be used for investigational 
purposes only, and that the foreign government is satisfied that the 
drug may legally be used by the intended consignee in that country. Such 
a request shall specify the quantity of drug to be shipped per shipment 
and the frequency of expected shipments.
    (iii) Authorization to export an investigational drug under 
paragraph (b)(2)(i) or (ii) of this section may be revoked by FDA if the 
agency finds that the conditions underlying its authorization are not 
longer met.
    (3) This paragraph applies only where the drug is to be used for the 
purpose of clinical investigation.

[[Page 95]]

    (4) This paragraph does not apply to the export of an antibiotic 
drug product shipped in accordance with the provisions of section 801(d) 
of the act.
    (5) This paragraph does not apply to the export of new drugs 
(including biological products) approved for export under section 802 of 
the act or section 351(h)(1)(A) of the Public Health Service Act.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987]



Sec. 312.120  Foreign clinical studies not conducted under an IND.

    (a) Introduction. This section describes the criteria for acceptance 
by FDA of foreign clinical studies not conducted under an IND. In 
general, FDA accepts such studies provided they are well designed, well 
conducted, performed by qualified investigators, and conducted in 
accordance with ethical principles acceptable to the world community. 
Studies meeting these criteria may be utilized to support clinical 
investigations in the United States and/or marketing approval. Marketing 
approval of a new drug or antibiotic drug based solely on foreign 
clinical data is governed by Sec. 314.106.
    (b) Data submissions. A sponsor who wishes to rely on a foreign 
clinical study to support an IND or to support an application for 
marketing approval shall submit to FDA the following information:
    (1) A description of the investigator's qualifications;
    (2) A description of the research facilities;
    (3) A detailed summary of the protocol and results of the study, 
and, should FDA request, case records maintained by the investigator or 
additional background data such as hospital or other institutional 
records;
    (4) A description of the drug substance and drug product used in the 
study, including a description of components, formulation, 
specifications, and bioavailability of the specific drug product used in 
the clinical study, if available; and
    (5) If the study is intended to support the effectiveness of a drug 
product, information showing that the study is adequate and well 
controlled under Sec. 314.126.
    (c) Conformance with ethical principles. (1) Foreign clinical 
research is required to have been conducted in accordance with the 
ethical principles stated in the ``Declaration of Helsinki'' (see 
paragraph (c)(4) of this section) or the laws and regulations of the 
country in which the research was conducted, whichever represents the 
greater protection of the individual.
    (2) For each foreign clinical study submitted under this section, 
the sponsor shall explain how the research conformed to the ethical 
principles contained in the ``Declaration of Helsinki'' or the foreign 
country's standards, whichever were used. If the foreign country's 
standards were used, the sponsor shall explain in detail how those 
standards differ from the ``Declaration of Helsinki'' and how they offer 
greater protection.
    (3) When the research has been approved by an independent review 
committee, the sponsor shall submit to FDA documentation of such review 
and approval, including the names and qualifications of the members of 
the committee. In this regard, a ``review committee'' means a committee 
composed of scientists and, where practicable, individuals who are 
otherwise qualified (e.g., other health professionals or laymen). The 
investigator may not vote on any aspect of the review of his or her 
protocol by a review committee.
    (4) The ``Declaration of Helsinki'' states as follows:

  Recommendations Guiding Physicians in Biomedical Research Involving 
                             Human Subjects

                              Introduction

    It is the mission of the physician to safeguard the health of the 
people. His or her knowledge and conscience are dedicated to the 
fulfillment of this mission.
    The Declaration of Geneva of the World Medical Association binds the 
physician with the words, ``The health of my patient will be my first 
consideration,'' and the International Code of Medical Ethics declares 
that, ``A physician shall act only in the patient's interest when 
providing medical care

[[Page 96]]

which might have the effect of weakening the physical and mental 
condition of the patient.''
    The purpose of biomedical research involving human subjects must be 
to improve diagnostic, therapeutic and prophylactic procedures and the 
understanding of the aetiology and pathogenesis of disease.
    In current medical practice most diagnostic, therapeutic or 
prophylactic procedures involve hazards. This applies especially to 
biomedical research.
    Medical progress is based on research which ultimately must rest in 
part on experimentation involving human subjects.
    In the field of biomedical research a fundamental distinction must 
be recognized between medical research in which the aim is essentially 
diagnostic or therapeutic for a patient, and medical research, the 
essential object of which is purely scientific and without implying 
direct diagnostic or therapeutic value to the person subjected to the 
research.
    Special caution must be exercised in the conduct of research which 
may affect the environment, and the welfare of animals used for research 
must be respected.
    Because it is essential that the results of laboratory experiments 
be applied to human beings to further scientific knowledge and to help 
suffering humanity, the World Medical Association has prepared the 
following recommendations as a guide to every physician in biomedical 
research involving human subjects. They should be kept under review in 
the future. It must be stressed that the standards as drafted are only a 
guide to physicians all over the world. Physicians are not relieved from 
criminal, civil and ethical responsibilities under the laws of their own 
countries.

                           I. Basic Principles

    1. Biomedical research involving human subjects must conform to 
generally accepted scientific principles and should be based on 
adequately performed laboratory and animal experimentation and on a 
thorough knowledge of the scientific literature.
    2. The design and performance of each experimental procedure 
involving human subjects should be clearly formulated in an experimental 
protocol which should be transmitted for consideration, comment and 
guidance to a specially appointed committee independent of the 
investigator and the sponsor provided that this independent committee is 
in conformity with the laws and regulations of the country in which the 
research experiment is performed.
    3. Biomedical research involving human subjects should be conducted 
only by scientifically qualified persons and under the supervision of a 
clinically competent medical person. The responsibility for the human 
subject must always rest with a medically qualified person and never 
rest on the subject of the research, even though the subject has given 
his or her consent.
    4. Biomedical research involving human subjects cannot legitimately 
be carried out unless the importance of the objective is in proportion 
to the inherent risk to the subject.
    5. Every biomedical research project involving human subjects should 
be preceded by careful assessment of predictable risks in comparison 
with foreseeable benefits to the subject or to others. Concern for the 
interests of the subject must always prevail over the interests of 
science and society.
    6. The right of the research subject to safeguard his or her 
integrity must always be respected. Every precaution should be taken to 
respect the privacy of the subject and to minimize the impact of the 
study on the subject's physical and mental integrity and on the 
personality of the subject.
    7. Physicians should abstain from engaging in research projects 
involving human subjects unless they are satisfied that the hazards 
involved are believed to be predictable. Physicians should cease any 
investigation if the hazards are found to outweigh the potential 
benefits.
    8. In publication of the results of his or her research, the 
physician is obliged to preserve the accuracy of the results. Reports of 
experimentation not in accordance with the principles laid down in this 
Declaration should not be accepted for publication.
    9. In any research on human beings, each potential subject must be 
adequately informed of the aims, methods, anticipated benefits and 
potential hazards of the study and the discomfort it may entail. He or 
she should be informed that he or she is at liberty to abstain from 
participation in the study and that he or she is free to withdraw his or 
her consent to participation at any time. The physician should then 
obtain the subject's freely-given informed consent, preferably in 
writing.
    10. When obtaining informed consent for the research project the 
physician should be particularly cautious if the subject is in a 
dependent relationship to him or her or may consent under duress. In 
that case the informed consent should be obtained by a physician who is 
not engaged in the investigation and who is completely independent of 
this official relationship.
    11. In case of legal incompetence, informed consent should be 
obtained from the legal guardian in accordance with national 
legislation. Where physical or mental incapacity makes it impossible to 
obtain informed consent, or when the subject is a minor, permission from 
the responsible relative replaces that of the subject in accordance with 
national legislation.
    Whenever the minor child is in fact able to give a consent, the 
minor's consent must be

[[Page 97]]

obtained in addition to the consent of the minor's legal guardian.
    12. The research protocol should always contain a statement of the 
ethical considerations involved and should indicate that the principles 
enunciated in the present Declaration are complied with.

II. Medical Research Combined with Professional Care (Clinical Research)

    1. In the treatment of the sick person, the physician must be free 
to use a new diagnostic and therapeutic measure, if in his or her 
judgment it offers hope of saving life, reestablishing health or 
alleviating suffering.
    2. The potential benefits, hazards and discomfort of a new method 
should be weighed against the advantages of the best current diagnostic 
and therapeutic methods.
    3. In any medical study, every patient--including those of a control 
group, if any--should be assured of the best proven diagnostic and 
therapeutic method.
    4. The refusal of the patient to participate in a study must never 
interfere with the physician-patient relationship.
    5. If the physician considers it essential not to obtain informed 
consent, the specific reasons for this proposal should be stated in the 
experimental protocol for transmission to the independent committee (I, 
2).
    6. The physician can combine medical research with professional 
care, the objective being the acquisition of new medical knowledge, only 
to the extent that medical research is justified by its potential 
diagnostic or therapeutic value for the patient.

 III. Non-Therapeutic Biomedical Research Involving Human Subjects (Non-
                      Clinical Biomedical Research)

    1. In the purely scientific application of medical research carried 
out on a human being, it is the duty of the physician to remain the 
protector of the life and health of that person on whom biomedical 
research is being carried out.
    2. The subjects should be volunteers--either healthy persons or 
patients for whom the experimental design is not related to the 
patient's illness.
    3. The investigator or the investigating team should discontinue the 
research if in his/her or their judgment it may, if continued, be 
harmful to the individual.
    4. In research on man, the interest of science and society should 
never take precedence over considerations related to the well-being of 
the subject.
(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)
[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 56 
FR 22113, May 14, 1991]



Sec. 312.130  Availability for public disclosure of data and information in an IND.

    (a) The existence of an investigational new drug application will 
not be disclosed by FDA unless it has previously been publicly disclosed 
or acknowledged.
    (b) The availability for public disclosure of all data and 
information in an investigational new drug application for a new drug or 
antibiotic drug will be handled in accordance with the provisions 
established in Sec. 314.430 for the confidentiality of data and 
information in applications submitted in part 314. The availability for 
public disclosure of all data and information in an investigational new 
drug application for a biological product will be governed by the 
provisions of Secs. 601.50 and 601.51.
    (c) Notwithstanding the provisions of Sec. 314.430, FDA shall 
disclose upon request to an individual to whom an investigational new 
drug has been given a copy of any IND safety report relating to the use 
in the individual.
    (d) The availability of information required to be publicly 
disclosed for investigations involving an exception from informed 
consent under Sec. 50.24 of this chapter will be handled as follows: 
Persons wishing to request the publicly disclosable information in the 
IND that was required to be filed in Docket Number 95S-0158 in the 
Dockets Management Branch (HFA-305), Food and Drug Administration, 12420 
Parklawn Dr., rm. 1-23, Rockville, MD 20857, shall submit a request 
under the Freedom of Information Act.
[52 FR 8831, Mar. 19, 1987. Redesignated at 53 FR 41523, Oct. 21, 1988, 
as amended at 61 FR 51530, Oct. 2, 1996]



Sec. 312.140  Address for correspondence.

    (a) Except as provided in paragraph (b) of this section, a sponsor 
shall send an initial IND submission to the Central Document Room, 
Center for Drug Evaluation and Research, Food and Drug Administration, 
Park Bldg., Rm. 214, 12420 Parklawn Dr., Rockville, MD 20852. On 
receiving the IND, FDA will inform the sponsor which one of the 
divisions in the Center for Drug Evaluation and Research or the Center 
for Biologics Evaluation and Research is responsible for the IND. 
Amendments,

[[Page 98]]

reports, and other correspondence relating to matters covered by the IND 
should be directed to the appropriate division. The outside wrapper of 
each submission shall state what is contained in the submission, for 
example, ``IND Application'', ``Protocol Amendment'', etc.
    (b) Applications for the products listed below should be submitted 
to the Division of Biological Investigational New Drugs (HFB-230), 
Center for Biologics Evaluation and Research, Food and Drug 
Administration, 8800 Rockville Pike, Bethesda, MD 20892. (1) Products 
subject to the licensing provisions of the Public Health Service Act of 
July 1, 1944 (58 Stat. 682, as amended (42 U.S.C. 201 et seq.)) or 
subject to part 600; (2) ingredients packaged together with containers 
intended for the collection, processing, or storage of blood or blood 
components; (3) urokinase products; (4) plasma volume expanders and 
hydroxyethyl starch for leukapheresis; and (5) coupled antibodies, i.e., 
products that consist of an antibody component coupled with a drug or 
radionuclide component in which both components provide a 
pharmacological effect but the biological component determines the site 
of action.
    (c) All correspondence relating to biological products for human use 
which are also radioactive drugs shall be submitted to the Division of 
Oncology and Radiopharmaceutical Drug Products (HFD-150), Center for 
Drug Evaluation and Research, Food and Drug Administration, 5600 Fishers 
Lane, Rockville, MD 20857, except that applications for coupled 
antibodies shall be submitted in accordance with paragraph (b) of this 
section.
    (d) All correspondence relating to export of an investigational drug 
under Sec. 312.110(b)(2) shall be submitted to the International Affairs 
Staff (HFY-50), Office of Health Affairs, Food and Drug Administration, 
5600 Fishers Lane, Rockville, MD 20857.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987; 55 
FR 11580, Mar. 29, 1990]



Sec. 312.145  Guidelines.

    (a) FDA has made available guidelines under Sec. 10.90(b) to help 
persons to comply with certain requirements of this part.
    (b) The Center for Drug Evaluation and Research and the Center for 
Biologics Evaluation and Research maintain lists of guidelines that 
apply to the Centers' regulations. The lists state how a person can 
obtain a copy of each guideline. A request for a copy of the lists 
should be directed to the CDER Executive Secretariat Staff (HFD-8), 
Center for Drug Evaluation and Research, Food and Drug Administration, 
5600 Fishers Lane, Rockville, MD 20857, for drug products, and the 
Congressional, Consumer, and International Affairs Staff (HFB-142), 
Center for Biologics Evaluation and Research, Food and Drug 
Administration, 8800 Rockville Pike, Bethesda, MD 20892, for biological 
products.
[52 FR 8831, Mar. 19, 1987, as amended at 55 FR 11580, Mar. 29, 1990; 56 
FR 3776, Jan. 31, 1991; 57 FR 10814, Mar. 31, 1992]



Subpart G--Drugs for Investigational Use in Laboratory Research Animals 
                            or In Vitro Tests



Sec. 312.160  Drugs for investigational use in laboratory research animals or in vitro tests.

    (a) Authorization to ship. (1)(i) A person may ship a drug intended 
solely for tests in vitro or in animals used only for laboratory 
research purposes if it is labeled as follows:

    CAUTION: Contains a new drug for investigational use only in 
laboratory research animals, or for tests in vitro. Not for use in 
humans.

    (ii) A person may ship a biological product for investigational in 
vitro diagnostic use that is listed in Sec. 312.2(b)(2)(ii) if it is 
labeled as follows:

    CAUTION: Contains a biological product for investigational in vitro 
diagnostic tests only.

    (2) A person shipping a drug under paragraph (a) of this section 
shall use due diligence to assure that the consignee is regularly 
engaged in conducting such tests and that the shipment of

[[Page 99]]

the new drug will actually be used for tests in vitro or in animals used 
only for laboratory research.
    (3) A person who ships a drug under paragraph (a) of this section 
shall maintain adequate records showing the name and post office address 
of the expert to whom the drug is shipped and the date, quantity, and 
batch or code mark of each shipment and delivery. Records of shipments 
under paragraph (a)(1)(i) of this section are to be maintained for a 
period of 2 years after the shipment. Records and reports of data and 
shipments under paragraph (a)(1)(ii) of this section are to be 
maintained in accordance with Sec. 312.57(b). The person who ships the 
drug shall upon request from any properly authorized officer or employee 
of the Food and Drug Administration, at reasonable times, permit such 
officer or employee to have access to and copy and verify records 
required to be maintained under this section.
    (b) Termination of authorization to ship. FDA may terminate 
authorization to ship a drug under this section if it finds that:
    (1) The sponsor of the investigation has failed to comply with any 
of the conditions for shipment established under this section; or
    (2) The continuance of the investigation is unsafe or otherwise 
contrary to the public interest or the drug is used for purposes other 
than bona fide scientific investigation. FDA will notify the person 
shipping the drug of its finding and invite immediate correction. If 
correction is not immediately made, the person shall have an opportunity 
for a regulatory hearing before FDA pursuant to part 16.
    (c) Disposition of unused drug. The person who ships the drug under 
paragraph (a) of this section shall assure the return of all unused 
supplies of the drug from individual investigators whenever the 
investigation discontinues or the investigation is terminated. The 
person who ships the drug may authorize in writing alternative 
disposition of unused supplies of the drug provided this alternative 
disposition does not expose humans to risks from the drug, either 
directly or indirectly (e.g., through food-producing animals). The 
shipper shall maintain records of any alternative disposition.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0014)

[52 FR 8831, Mar. 19, 1987, as amended at 52 FR 23031, June 17, 1987. 
Redesignated at 53 FR 41523, Oct. 21, 1988]



PART 314--APPLICATIONS FOR FDA APPROVAL TO MARKET A NEW DRUG OR AN ANTIBIOTIC DRUG--Table of Contents




                      Subpart A--General Provisions

Sec.
314.1  Scope of this part.
314.2  Purpose.
314.3  Definitions.

                         Subpart B--Applications

314.50  Content and format of an application.
314.52  Notice of certification of invalidity or noninfringement of a 
          patent.
314.53  Submission of patent information.
314.54  Procedure for submission of an application requiring 
          investigations for approval of a new indication for, or other 
          change from, a listed drug.
314.60  Amendments to an unapproved application.
314.65  Withdrawal by the applicant of an unapproved application.
314.70  Supplements and other changes to an approved application.
314.71  Procedures for submission of a supplement to an approved 
          application.
314.72  Change in ownership of an application.
314.80  Postmarketing reporting of adverse drug experiences.
314.81  Other postmarketing reports.
314.90  Waivers.

                   Subpart C--Abbreviated Applications

314.92  Drug products for which abbreviated applications may be 
          submitted.
314.93  Petition to request a change from a listed drug.
314.94  Content and format of an abbreviated application.
314.95  Notice of certification of invalidity or noninfringement of a 
          patent.
314.96  Amendments to an unapproved abbreviated application.
314.97  Supplements and other changes to an approved abbreviated 
          application.
314.98  Postmarketing reports.
314.99  Other responsibilities of an applicant of an abbreviated 
          application.

[[Page 100]]

   Subpart D--FDA Action on Applications and Abbreviated Applications

314.100  Timeframes for reviewing applications and abbreviated 
          applications.
314.101  Filing an application and an abbreviated antibiotic application 
          and receiving an abbreviated new drug application.
314.102  Communications between FDA and applicants.
314.103  Dispute resolution.
314.104  Drugs with potential for abuse.
314.105  Approval of an application and an abbreviated application.
314.106  Foreign data.
314.107  Effective date of approval of a 505(b)(2) application or 
          abbreviated new drug application under section 505(j) of the 
          act.
314.108  New drug product exclusivity.
314.110  Approvable letter to the applicant.
314.120  Not approvable letter to the applicant.
314.122  Submitting an abbreviated application for, or a 505(j)(2)(C) 
          petition that relies on, a listed drug that is no longer 
          marketed.
314.125  Refusal to approve an application or abbreviated antibiotic 
          application.
314.126  Adequate and well-controlled studies.
314.127  Refusal to approve an abbreviated new drug application.
314.150  Withdrawal of approval of an application or abbreviated 
          application.
314.151  Withdrawal of approval of an abbreviated new drug application 
          under section 505(j)(5) of the act.
314.152  Notice of withdrawal of approval of an application or 
          abbreviated application for a new drug.
314.153  Suspension of approval of an abbreviated new drug application.
314.160  Approval of an application or abbreviated application for which 
          approval was previously refused, suspended, or withdrawn.
314.161  Determination of reasons for voluntary withdrawal of a listed 
          drug.
314.162  Removal of a drug product from the list.
314.170  Adulteration and misbranding of an approved drug.

               Subpart E--Hearing Procedures for New Drugs

314.200  Notice of opportunity for hearing; notice of participation and 
          request for hearing; grant or denial of hearing.
314.201  Procedure for hearings.
314.235  Judicial review.

          Subpart F--Administrative Procedures for Antibiotics

314.300  Procedure for the issuance, amendment, or repeal of 
          regulations.

                   Subpart G--Miscellaneous Provisions

314.410  Imports and exports of new drugs and antibiotics.
314.420  Drug master files.
314.430  Availability for public disclosure of data and information in 
          an application or abbreviated application.
314.440  Addresses for applications and abbreviated applications.
314.445  Guidelines.

    Subpart H--Accelerated Approval of New Drugs for Serious or Life-
                          Threatening Illnesses

314.500  Scope.
314.510  Approval based on a surrogate endpoint or on an effect on a 
          clinical endpoint other than survival or irreversible 
          morbidity.
314.520  Approval with restrictions to assure safe use.
314.530  Withdrawal procedures.
314.540  Postmarketing safety reporting.
314.550  Promotional materials.
314.560  Termination of requirements.

    Authority:  21 U.S.C. 321, 331, 351, 352, 353, 355, 356, 357, 371, 
374, 379e.

    Source:  50 FR 7493, Feb. 22, 1985, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 314.1  Scope of this part.

    (a) This part sets forth procedures and requirements for the 
submission to, and the review by, the Food and Drug Administration of 
applications and abbreviated applications, as well as amendments, 
supplements, and postmarketing reports to them, by persons seeking or 
holding approval from FDA of the following:
    (1) An application or abbreviated application under section 505 of 
the Federal Food, Drug, and Cosmetic Act to market a new drug.
    (2) An application or abbreviated application under section 507 of 
the Federal Food, Drug, and Cosmetic Act to market an antibiotic drug.
    (b) This part does not apply to drug products subject to licensing 
by FDA under the Public Health Service Act (58 Stat. 632 as amended (42 
U.S.C. 201 et seq.)) and subchapter F of chapter I of title 21 of the 
Code of Federal Regulations.

[[Page 101]]

    (c) References in this part to regulations in the Code of Federal 
Regulations are to chapter I of title 21, unless otherwise noted.
[50 FR 7493, Feb. 22, 1985, as amended at 57 FR 17981, Apr. 28, 1992]



Sec. 314.2  Purpose.

    The purpose of this part is to establish an efficient and thorough 
drug review process in order to: (a) Facilitate the approval of drugs 
shown to be safe and effective; and (b) ensure the disapproval of drugs 
not shown to be safe and effective. These regulations are also intended 
to establish an effective system for FDA's surveillance of marketed 
drugs. These regulations shall be construed in light of these 
objectives.



Sec. 314.3  Definitions.

    (a) The definitions and interpretations contained in section 201 of 
the act apply to those terms when used in this part.
    (b) The following definitions of terms apply to this part:
    Abbreviated application means the application described under 
Sec. 314.94, including all amendments and supplements to the 
application. ``Abbreviated application'' applies to both an abbreviated 
new drug application and an abbreviated antibiotic application.
    Act means the Federal Food, Drug, and Cosmetic Act (sections 201-901 
(21 U.S.C. 301-392)).
    Applicant means any person who submits an application or abbreviated 
application or an amendment or supplement to them under this part to 
obtain FDA approval of a new drug or an antibiotic drug and any person 
who owns an approved application or abbreviated application.
    Application means the application described under Sec. 314.50, 
including all amendements and supplements to the application.
    505(b)(2) Application means an application submitted under section 
505(b)(1) of the act for a drug for which the investigations described 
in section 505(b)(1)(A) of the act and relied upon by the applicant for 
approval of the application were not conducted by or for the applicant 
and for which the applicant has not obtained a right of reference or use 
from the person by or for whom the investigations were conducted.
    Approvable letter means a written communication to an applicant from 
FDA stating that the agency will approve the application or abbreviated 
application if specific additional information or material is submitted 
or specific conditions are met. An approvable letter does not constitute 
approval of any part of an application or abbreviated application and 
does not permit marketing of the drug that is the subject of the 
application or abbreviated application.
    Approval letter means a written communication to an applicant from 
FDA approving an application or an abbreviated application.
    Drug product means a finished dosage form, for example, tablet, 
capsule, or solution, that contains a drug substance, generally, but not 
necessarily, in association with one or more other ingredients.
    Drug substance means an active ingredient that is intended to 
furnish pharmacological activity or other direct effect in the 
diagnosis, cure, mitigation, treatment, or prevention of disease or to 
affect the structure or any function of the human body, but does not 
include intermediates use in the synthesis of such ingredient.
    FDA means the Food and Drug Administration.
    Listed drug means a new drug product that has an effective approval 
under section 505(c) of the act for safety and effectiveness or under 
section 505(j) of the act, which has not been withdrawn or suspended 
under section 505(e)(1) through (e)(5) or (j)(5) of the act, and which 
has not been withdrawn from sale for what FDA has determined are reasons 
of safety or effectiveness. Listed drug status is evidenced by the drug 
product's identification as a drug with an effective approval in the 
current edition of FDA's ``Approved Drug Products with Therapeutic 
Equivalence Evaluations'' (the list) or any current supplement thereto, 
as a drug with an effective approval. A drug product is deemed to be a 
listed drug on the date of effective approval of the application or 
abbreviated application for that drug product.

[[Page 102]]

    Not approvable letter means a written communication to an applicant 
from FDA stating that the agency does not consider the application or 
abbreviated application approvable because one or more deficiencies in 
the application or abbreviated application preclude the agency from 
approving it.
    Reference listed drug means the listed drug identified by FDA as the 
drug product upon which an applicant relies in seeking approval of its 
abbreviated application.
    Right of reference or use means the authority to rely upon, and 
otherwise use, an investigation for the purpose of obtaining approval of 
an application, including the ability to make available the underlying 
raw data from the investigation for FDA audit, if necessary.
    The list means the list of drug products with effective approvals 
published in the current edition of FDA's publication ``Approved Drug 
Products with Therapeutic Equivalence Evaluations'' and any current 
supplement to the publication.
[50 FR 7493, Feb. 22, 1985, as amended at 57 FR 17981, Apr. 28, 1992]



                         Subpart B--Applications



Sec. 314.50  Content and format of an application.

    Applications and supplements to approved applications are required 
to be submitted in the form and contain the information, as appropriate 
for the particular submission, required under this section. Three copies 
of the application are required: An archival copy, a review copy, and a 
field copy. An application for a new chemical entity will generally 
contain an application form, an index, a summary, five or six technical 
sections, case report tabulations of patient data, case report forms, 
drug samples, and labeling. Other applications will generally contain 
only some of those items, and information will be limited to that needed 
to support the particular submission. These include an application of 
the type described in section 505(b)(2) of the act, an amendment, and a 
supplement. The application is required to contain reports of all 
investigations of the drug product sponsored by the applicant, and all 
other information about the drug pertinent to an evaluation of the 
application that is received or otherwise obtained by the applicant from 
any source. FDA will maintain guidelines on the format and content of 
applications to assist applicants in their preparation.
    (a) Application form. The applicant shall submit a completed and 
signed application form that contains the following:
    (1) The name and address of the applicant; the date of the 
application; the application number if previously issued (for example, 
if the application is a resubmission, an amendment, or a supplement); 
the name of the drug product, including its established, proprietary, 
code, and chemical names; the dosage form and strength; the route of 
administration; the identification numbers of all investigational new 
drug applications that are referenced in the application; the 
identification numbers of all drug master files and other applications 
under this part that are referenced in the application; and the drug 
product's proposed indications for use.
    (2) A statement whether the submission is an original submission, a 
505(b)(2) application, a resubmission, or a supplement to an application 
under Sec. 314.70.
    (3) A statement whether the applicant proposes to market the drug 
product as a prescription or an over-the-counter product.
    (4) A check-list identifying what enclosures required under this 
section the applicant is submitting.
    (5) The applicant, or the applicant's attorney, agent, or other 
authorized official shall sign the application. If the person signing 
the application does not reside or have a place of business within the 
United States, the application is required to contain the name and 
address of, and be countersigned by, an attorney, agent, or other 
authorized official who resides or maintains a place of business within 
the United States.
    (b) Index. The archival copy of the application is required to 
contain a comprehensive index by volume number and page number to the 
summary under paragraph (c) of this section, the technical sections 
under paragraph (d)

[[Page 103]]

of this section, and the supporting information under paragraph (f) of 
this section.
    (c) Summary. (1) An application is required to contain a summary of 
the application in enough detail that the reader may gain a good general 
understanding of the data and information in the application, including 
an understanding of the quantitative aspects of the data. The summary is 
not required for supplements under Sec. 314.70. Resubmissions of an 
application should contain an updated summary, as appropriate. The 
summary should discuss all aspects of the application, and synthesize 
the information into a well-structured and unified document. The summary 
should be written at approximately the level of detail required for 
publication in, and meet the editorial standards generally applied by, 
refereed scientific and medical journals. In addition to the agency 
personnel reviewing the summary in the context of their review of the 
application, FDA may furnish the summary to FDA advisory committee 
members and agency officials whose duties require an understanding of 
the application. To the extent possible, data in the summary should be 
presented in tabular and graphic forms. FDA has prepared a guideline 
under Sec. 10.90(b) that provides information about how to prepare a 
summary. The summary required under this paragraph may be used by FDA or 
the applicant to prepare the Summary Basis of Approval document for 
public disclosure (under Sec. 314.430(e)(2)(ii)) when the application is 
approved.
    (2) The summary is required to contain the following information:
    (i) The proposed text of the labeling for the drug, with annotations 
to the information in the summary and technical sections of the 
application that support the inclusion of each statement in the 
labeling, and, if the application is for a prescription drug, statements 
describing the reasons for omitting a section or subsection of the 
labeling format in Sec. 201.57.
    (ii) A statement identifying the pharmacologic class of the drug and 
a discussion of the scientific rationale for the drug, its intended use, 
and the potential clinical benefits of the drug product.
    (iii) A brief description of the marketing history, if any, of the 
drug outside the United States, including a list of the countries in 
which the drug has been marketed, a list of any countries in which the 
drug has been withdrawn from marketing for any reason related to safety 
or effectiveness, and a list of countries in which applications for 
marketing are pending. The description is required to describe both 
marketing by the applicant and, if known, the marketing history of other 
persons.
    (iv) A summary of the chemistry, manufacturing, and controls section 
of the application.
    (v) A summary of the nonclinical pharmacology and toxicology section 
of the application.
    (vi) A summary of the human pharmacokinetics and bioavailability 
section of the application.
    (vii) A summary of the microbiology section of the application (for 
anti-infective drugs only).
    (viii) A summary of the clinical data section of the application, 
including the results of statistical analyses of the clinical trials.
    (ix) A concluding discussion that presents the benefit and risk 
considerations related to the drug, including a discussion of any 
proposed additional studies or surveillance the applicant intends to 
conduct postmarketing.
    (d) Technical sections. The application is required to contain the 
technical sections described below. Each technical section is required 
to contain data and information in sufficient detail to permit the 
agency to make a knowledgeable judgment about whether to approve the 
application or whether grounds exist under section 505(d) or 507 of the 
act to refuse to approve the application. The required technical 
sections are as follows:
    (1) Chemistry, manufacturing, and controls section. A section 
describing the composition, manufacture, and specification of the drug 
substance and the drug product, including the following:
    (i) Drug substance. A full description of the drug substance 
including its physical and chemical characteristics and stability; the 
name and address of

[[Page 104]]

its manufacturer; the method of synthesis (or isolation) and 
purification of the drug substance; the process controls used during 
manufacture and packaging; and such specifications and analytical 
methods as are necessary to assure the identity, strength, quality, and 
purity of the drug substance and the bioavailability of the drug 
products made from the substance, including, for example, specifications 
relating to stability, sterility, particle size, and crystalline form. 
The application may provide additionally for the use of alternatives to 
meet any of these requirements, including alternative sources, process 
controls, methods, and specifications. Reference to the current edition 
of the U.S. Pharmacopeia and the National Formulary may satisfy relevant 
requirements in this paragraph.
    (ii)(a) Drug product. A list of all components used in the 
manufacture of the drug product (regardless of whether they appear in 
the drug product); and a statement of the composition of the drug 
product; a statement of the specifications and analytical methods for 
each component; the name and address of each manufacturer the drug 
product; a description of the manufacturing and packaging procedures and 
in-process controls for the drug product; such specifications and 
analytical methods as are necessary to assure the identity, strength, 
quality, purity, and bioavailability of the drug product, including, for 
example, specifications relating to sterility, dissolution rate, 
containers and closure systems; and stability data with proposed 
expiration dating. The application may provide additionally for the use 
of alternatives to meet any of these requirements, including alternative 
components, manufacturing and packaging procedures, in-process controls, 
methods, and specifications. Reference to the current edition of the 
U.S. Pharmacopeia and the National Formulary may satisfy relevant 
requirements in this paragraph.
    (b) Unless provided by paragraph (d)(1)(ii)(a) of this section, for 
each batch of the drug product used to conduct a bioavailability or 
bioequivalence study described in Sec. 320.38 or Sec. 320.63 of this 
chapter or used to conduct a primary stability study: The batch 
production record; the specifications and test procedures for each 
component and for the drug product; the names and addresses of the 
sources of the active and noncompendial inactive components and of the 
container and closure system for the drug product; the name and address 
of each contract facility involved in the manufacture, processing, 
packaging, or testing of the drug product and identification of the 
operation performed by each contract facility; and the results of any 
test performed on the components used in the manufacture of the drug 
product as required by Sec. 211.84(d) of this chapter and on the drug 
product as required by Sec. 211.165 of this chapter.
    (c) The proposed or actual master production record, including a 
description of the equipment, to be used for the manufacture of a 
commercial lot of the drug product or a comparably detailed description 
of the production process for a representative batch of the drug 
product.
    (iii) Environmental impact. The application is required to contain 
either a claim for categorical exclusion under Sec. 25.30 or 25.31 of 
this chapter or an environmental assessment under Sec. 25.40 of this 
chapter.
    (iv) The applicant may, at its option, submit a complete chemistry, 
manufacturing, and controls section 90 to 120 days before the 
anticipated submission of the remainder of the application. FDA will 
review such early submissions as resources permit.
    (v) Except for a foreign applicant, the applicant shall include a 
statement certifying that the field copy of the application has been 
provided to the applicant's home FDA district office.
    (2) Nonclinical pharmacology and toxicology section. A section 
describing, with the aid of graphs and tables, animal and in vitro 
studies with drug, including the following:
    (i) Studies of the pharmacological actions of the drug in relation 
to its proposed therapeutic indication and studies that otherwise define 
the pharmacologic properties of the drug or are pertinent to possible 
adverse effects.
    (ii) Studies of the toxicological effects of the drug as they relate 
to the

[[Page 105]]

drug's intended clinical uses, including, as appropriate, studies 
assessing the drug's acute, subacute, and chronic toxicity; 
carcinogenicity; and studies of toxicities related to the drug's 
particular mode of administration or conditions of use.
    (iii) Studies, as appropriate, of the effects of the drug on 
reproduction and on the developing fetus.
    (iv) Any studies of the absorption, distribution, metabolism, and 
excretion of the drug in animals.
    (v) For each nonclinical laboratory study subject to the good 
laboratory practice regulations under part 58 a statement that it was 
conducted in compliance with the good laboratory practice regulations in 
part 58, or, if the study was not conducted in compliance with those 
regulations, a brief statement of the reason for the noncompliance.
    (3) Human pharmacokinetics and bioavailability section. A section 
describing the human pharmacokinetic data and human bioavailability 
data, or information supporting a waiver of the submission of in vivo 
bioavailability data under subpart B of part 320, including the 
following:
    (i) A description of each of the bioavailability and pharmacokinetic 
studies of the drug in humans performed by or on behalf of the applicant 
that includes a description of the analytical and statistical methods 
used in each study and a statement with respect to each study that it 
either was conducted in compliance with the institutional review board 
regulations in part 56, or was not subject to the regulations under 
Sec. 56.104 or Sec. 56.105, and that it was conducted in compliance with 
the informed consent regulations in part 50.
    (ii) If the application describes in the chemistry, manufacturing, 
and controls section specifications or analytical methods needed to 
assure the bioavailability of the drug product or drug substance, or 
both, a statement in this section of the rationale for establishing the 
specification or analytical methods, including data and information 
supporting the rationale.
    (iii) A summarizing discussion and analysis of the pharmacokinetics 
and metabolism of the active ingredients and the bioavailability or 
bioequivalence, or both, of the drug product.
    (4) Microbiology section. If the drug is an anti-infective drug, a 
section describing the microbiology data, including the following:
    (i) A description of the biochemical basis of the drug's action on 
microbial physiology.
    (ii) A description of the antimicrobial spectra of the drug, 
including results of in vitro preclinical studies to demonstrate 
concentrations of the drug required for effective use.
    (iii) A description of any known mechanisms of resistance to the 
drug, including results of any known epidemiologic studies to 
demonstrate prevalence of resistance factors.
    (iv) A description of clinical microbiology laboratory methods (for 
example, in vitro sensitivity discs) needed for effective use of the 
drug.
    (5) Clinical data section. A section describing the clinical 
investigations of the drug, including the following:
    (i) A description and analysis of each clinical pharmacology study 
of the drug, including a brief comparison of the results of the human 
studies with the animal pharmacology and toxicology data.
    (ii) A description and analysis of each controlled clinical study 
pertinent to a proposed use of the drug, including the protocol and a 
description of the statistical analyses used to evaluate the study. If 
the study report is an interim analysis, this is to be noted and a 
projected completion date provided. Controlled clinical studies that 
have not been analyzed in detail for any reason (e.g., because they have 
been discontinued or are incomplete) are to be included in this section, 
including a copy of the protocol and a brief description of the results 
and status of the study.
    (iii) A description of each uncontrolled clinical study, a summary 
of the results, and a brief statement explaining why the study is 
classified as uncontrolled.
    (iv) A description and analysis of any other data or information 
relevant to an evaluation of the safety and effectiveness of the drug 
product obtained or otherwise received by the applicant from any source, 
foreign or domestic, including information derived from

[[Page 106]]

clinical investigations, including controlled and uncontrolled studies 
of uses of the drug other than those proposed in the application, 
commercial marketing experience, reports in the scientific literature, 
and unpublished scientific papers.
    (v) An integrated summary of the data demonstrating substantial 
evidence of effectiveness for the claimed indications. Evidence is also 
required to support the dosage and administration section of the 
labeling, including support for the dosage and dose interval 
recommended. The effectiveness data shall be presented by gender, age, 
and racial subgroups and shall identify any modifications of dose or 
dose interval needed for specific subgroups. Effectiveness data from 
other subgroups of the population of patients treated, when appropriate, 
such as patients with renal failure or patients with different levels of 
severity of the disease, also shall be presented.
    (vi) A summary and updates of safety information, as follows:
    (a) The applicant shall submit an integrated summary of all 
available information about the safety of the drug product, including 
pertinent animal data, demonstrated or potential adverse effects of the 
drug, clinically significant drug/drug interactions, and other safety 
considerations, such as data from epidemiological studies of related 
drugs. The safety data shall be presented by gender, age, and racial 
subgroups. When appropriate, safety data from other subgroups of the 
population of patients treated also shall be presented, such as for 
patients with renal failure or patients with different levels of 
severity of the disease. A description of any statistical analyses 
performed in analyzing safety data should also be included, unless 
already included under paragraph (d)(5)(ii) of this section.
    (b) The applicant shall, under section 505(i) of the act, update 
periodically its pending application with new safety information learned 
about the drug that may reasonably affect the statement of 
contraindications, warnings, precautions, and adverse reactions in the 
draft labeling. These ``safety update reports'' are required to include 
the same kinds of information (from clinical studies, animal studies, 
and other sources) and are required to be submitted in the same format 
as the integrated summary in paragraph (d)(5)(vi)(a) of this section. In 
addition, the reports are required to include the case report forms for 
each patient who died during a clinical study or who did not complete 
the study because of an adverse event (unless this requirement is 
waived). The applicant shall submit these reports (1) 4 months after the 
initial submission; (2) following receipt of an approvable letter; and 
(3) at other times as requested by FDA. Prior to the submission of the 
first such report, applicants are encouraged to consult with FDA 
regarding further details on its form and content.
    (vii) If the drug has a potential for abuse, a description and 
analysis of studies or information related to abuse of the drug, 
including a proposal for scheduling under the Controlled Substances Act. 
A description of any studies related to overdosage is also required, 
including information on dialysis, antidotes, or other treatments, if 
known.
    (viii) An integrated summary of the benefits and risks of the drug, 
including a discussion of why the benefits exceed the risks under the 
conditions stated in the labeling.
    (ix) A statement with respect to each clinical study involving human 
subjects that it either was conducted in compliance with the 
institutional review board regulations in part 56, or was not subject to 
the regulations under Sec. 56.104 or Sec. 56.105, and that it was 
conducted in compliance with the informed consent regulations in part 
50.
    (x) If a sponsor has transferred any obligations for the conduct of 
any clinical study to a contract research organization, a statement 
containing the name and address of the contract research organization, 
identification of the clinical study, and a listing of the obligations 
transferred. If all obligations governing the conduct of the study have 
been transferred, a general statement of this transfer--in lieu of a 
listing of the specific obligations transferred--may be submitted.
    (xi) If original subject records were audited or reviewed by the 
sponsor in the course of monitoring any clinical

[[Page 107]]

study to verify the accuracy of the case reports submitted to the 
sponsor, a list identifying each clinical study so audited or reviewed.
    (6) Statistical section. A section describing the statistical 
evaluation of clinical data, including the following:
    (i) A copy of the information submitted under paragraph (d)(5)(ii) 
of this section concerning the description and analysis of each 
controlled clinical study, and the documentation and supporting 
statistical analyses used in evaluating the controlled clinical studies.
    (ii) A copy of the information submitted under paragraph 
(d)(5)(vi)(a) of this section concerning a summary of information about 
the safety of the drug product, and the documentation and supporting 
statistical analyses used in evaluating the safety information.
    (e) Samples and labeling. (1) Upon request from FDA, the applicant 
shall submit the samples described below to the places identified in the 
agency's request. FDA will generally ask applicants to submit samples 
directly to two or more agency laboratories that will perform all 
necessary tests on the samples and validate the applicant's analytical 
methods.
    (i) Four representative samples of the following, each sample in 
sufficient quantity to permit FDA to perform three times each test 
described in the application to determine whether the drug substance and 
the drug product meet the specifications given in the application:
    (a) The drug product proposed for marketing;
    (b) The drug substance used in the drug product from which the 
samples of the drug product were taken; and
    (c) Reference standards and blanks (except that reference standards 
recognized in an official compendium need not be submitted).
    (ii) Samples of the finished market package, if requested by FDA.
    (2) The applicant shall submit the following in the archival copy of 
the application:
    (i) Three copies of the analytical methods and related descriptive 
information contained in the chemistry, manufacturing, and controls 
section under paragraph (d)(1) of this section for the drug substance 
and the drug product that are necessary for FDA's laboratories to 
perform all necessary tests on the samples and to validate the 
applicant's analytical methods. The related descriptive information 
includes a description of each sample; the proposed regulatory 
specifications for the drug; a detailed description of the methods of 
analysis; supporting data for accuracy, specificity, precision and 
ruggedness; and complete results of the applicant's tests on each 
sample.
    (ii) Copies of the label and all labeling for the drug product (4 
copies of draft labeling or 12 copies of final printed labeling).
    (f) Case report forms and tabulations. The archival copy of the 
application is required to contain the following case report tabulations 
and case report forms:
    (1) Case report tabulations. The application is required to contain 
tabulations of the data from each adequate and well-controlled study 
under Sec. 314.126 (Phase 2 and Phase 3 studies as described in 
Secs. 312.21 (b) and (c) of this chapter), tabulations of the data from 
the earliest clinical pharmacology studies (Phase 1 studies as described 
in Sec. 312.21(a) of this chapter), and tabulations of the safety data 
from other clinical studies. Routine submission of other patient data 
from uncontrolled studies is not required. The tabulations are required 
to include the data on each patient in each study, except that the 
applicant may delete those tabulations which the agency agrees, in 
advance, are not pertinent to a review of the drug's safety or 
effectiveness. Upon request, FDA will discuss with the applicant in a 
``pre-NDA'' conference those tabulations that may be appropriate for 
such deletion. Barring unforeseen circumstances, tabulations agreed to 
be deleted at such a conference will not be requested during the conduct 
of FDA's review of the application. If such unforeseen circumstances do 
occur, any request for deleted tabulations will be made by the director 
of the FDA division responsible for reviewing the application, in 
accordance with paragraph (f)(3) of this section.

[[Page 108]]

    (2) Case report forms. The application is required to contain copies 
of individual case report forms for each patient who died during a 
clinical study or who did not complete the study because of an adverse 
event, whether believed to be drug related or not, including patients 
receiving reference drugs or placebo. This requirement may be waived by 
FDA for specific studies if the case report forms are unnecessary for a 
proper review of the study.
    (3) Additional data. The applicant shall submit to FDA additional 
case report forms and tabulations needed to conduct a proper review of 
the application, as requested by the director of the FDA division 
responsible for reviewing the application. The applicant's failure to 
submit information requested by FDA within 30 days after receipt of the 
request may result in the agency viewing any eventual submission as a 
major amendment under Sec. 314.60 and extending the review period as 
necessary. If desired by the applicant, the FDA division director will 
verify in writing any request for additional data that was made orally.
    (4) Applicants are invited to meet with FDA before submitting an 
application to discuss the presentation and format of supporting 
information. If the applicant and FDA agree, the applicant may submit 
tabulations of patient data and case report forms in a form other than 
hard copy, for example, on microfiche or computer tapes.
    (g) Other. The following general requirements apply to the 
submission of information within the summary under paragraph (c) of this 
section and within the technical sections under paragraph (d) of this 
section.
    (1) The applicant ordinarily is not required to resubmit information 
previously submitted, but may incorporate the information by reference. 
A reference to information submitted previously is required to identify 
the file by name, reference number, volume, and page number in the 
agency's records where the information can be found. A reference to 
information submitted to the agency by a person other than the applicant 
is required to contain a written statement that authorizes the reference 
and that is signed by the person who submitted the information.
    (2) The applicant shall submit an accurate and complete English 
translation of each part of the application that is not in English. The 
applicant shall submit a copy of each original literature publication 
for which an English translation is submitted.
    (3) If an applicant who submits a new drug application under section 
505(b) of the act obtains a ``right of reference or use,'' as defined 
under Sec. 314.3(b), to an investigation described in clause (A) of 
section 505(b)(1) of the act, the applicant shall include in its 
application a written statement signed by the owner of the data from 
each such investigation that the applicant may rely on in support of the 
approval of its application, and provide FDA access to, the underlying 
raw data that provide the basis for the report of the investigation 
submitted in its application.
    (h) Patent information. The application is required to contain the 
patent information described under Sec. 314.53.
    (i) Patent certification--(1) Contents. A 505(b)(2) application is 
required to contain the following:
    (i) Patents claiming drug, drug product, or method of use. (A) 
Except as provided in paragraph (i)(2) of this section, a certification 
with respect to each patent issued by the United States Patent and 
Trademark Office that, in the opinion of the applicant and to the best 
of its knowledge, claims a drug (the drug product or drug substance that 
is a component of the drug product) on which investigations that are 
relied upon by the applicant for approval of its application were 
conducted or that claims an approved use for such drug and for which 
information is required to be filed under section 505(b) and (c) of the 
act and Sec. 314.53. For each such patent, the applicant shall provide 
the patent number and certify, in its opinion and to the best of its 
knowledge, one of the following circumstances:
    (1) That the patent information has not been submitted to FDA. The 
applicant shall entitle such a certification ``Paragraph I 
Certification'';
    (2) That the patent has expired. The applicant shall entitle such a 
certification ``Paragraph II Certification'';

[[Page 109]]

    (3) The date on which the patent will expire. The applicant shall 
entitle such a certification ``Paragraph III Certification''; or
    (4) That the patent is invalid, unenforceable, or will not be 
infringed by the manufacture, use, or sale of the drug product for which 
the application is submitted. The applicant shall entitle such a 
certification ``Paragraph IV Certification''. This certification shall 
be submitted in the following form:

I, (name of applicant), certify that Patent No. ____________ (is 
invalid, unenforceable, or will not be infringed by the manufacture, 
use, or sale of) (name of proposed drug product) for which this 
application is submitted.

The certification shall be accompanied by a statement that the applicant 
will comply with the requirements under Sec. 314.52(a) with respect to 
providing a notice to each owner of the patent or their representatives 
and to the holder of the approved application for the drug product which 
is claimed by the patent or a use of which is claimed by the patent and 
with the requirements under Sec. 314.52(c) with respect to the content 
of the notice.
    (B) If the drug on which investigations that are relied upon by the 
applicant were conducted is itself a licensed generic drug of a patented 
drug first approved under section 505(b) of the act, the appropriate 
patent certification under this section with respect to each patent that 
claims the first-approved patented drug or that claims an approved use 
for such a drug.
    (ii) No relevant patents. If, in the opinion of the applicant and to 
the best of its knowledge, there are no patents described in paragraph 
(i)(1)(i) of this section, a certification in the following form:

In the opinion and to the best knowledge of (name of applicant), there 
are no patents that claim the drug or drugs on which investigations that 
are relied upon in this application were conducted or that claim a use 
of such drug or drugs.

    (iii) Method of use patent. (A) If information that is submitted 
under section 505(b) or (c) of the act and Sec. 314.53 is for a method 
of use patent, and the labeling for the drug product for which the 
applicant is seeking approval does not include any indications that are 
covered by the use patent, a statement explaining that the method of use 
patent does not claim any of the proposed indications.
    (B) If the labeling of the drug product for which the applicant is 
seeking approval includes an indication that, according to the patent 
information submitted under section 505(b) or (c) of the act and 
Sec. 314.53 or in the opinion of the applicant, is claimed by a use 
patent, the applicant shall submit an applicable certification under 
paragraph (i)(1)(i) of this section.
    (2) Method of manufacturing patent. An applicant is not required to 
make a certification with respect to any patent that claims only a 
method of manufacturing the drug product for which the applicant is 
seeking approval.
    (3) Licensing agreements. If a 505(b)(2) application is for a drug 
or method of using a drug claimed by a patent and the applicant has a 
licensing agreement with the patent owner, the applicant shall submit a 
certification under paragraph (i)(1)(i)(A)(4) of this section 
(``Paragraph IV Certification'') as to that patent and a statement that 
it has been granted a patent license. If the patent owner consents to an 
immediate effective date upon approval of the 505(b)(2) application, the 
application shall contain a written statement from the patent owner that 
it has a licensing agreement with the applicant and that it consents to 
an immediate effective date.
    (4) Late submission of patent information. If a patent described in 
paragraph (i)(1)(i)(A) of this section is issued and the holder of the 
approved application for the patented drug does not submit the required 
information on the patent within 30 days of issuance of the patent, an 
applicant who submitted a 505(b)(2) application that, before the 
submission of the patent information, contained an appropriate patent 
certification is not required to submit an amended certification. An 
applicant whose 505(b)(2) application is filed after a late submission 
of patent information or whose 505(b)(2) application was previously 
filed but did not contain an appropriate patent certification at the 
time of the patent submission shall submit a certification under 
paragraph (i)(1)(i) or (i)(1)(ii) of this section or a

[[Page 110]]

statement under paragraph (i)(1)(iii) of this section as to that patent.
    (5) Disputed patent information. If an applicant disputes the 
accuracy or relevance of patent information submitted to FDA, the 
applicant may seek a confirmation of the correctness of the patent 
information in accordance with the procedures under Sec. 314.53(f). 
Unless the patent information is withdrawn or changed, the applicant 
must submit an appropriate certification for each relevant patent.
    (6) Amended certifications. A certification submitted under 
paragraphs (i)(1)(i) through (i)(1)(iii) of this section may be amended 
at any time before the effective date of the approval of the 
application. An applicant shall submit an amended certification as an 
amendment to a pending application or by letter to an approved 
application. If an applicant with a pending application voluntarily 
makes a patent certification for an untimely filed patent, the applicant 
may withdraw the patent certification for the untimely filed patent. 
Once an amendment or letter for the change in certification has been 
submitted, the application will no longer be considered to be one 
containing the prior certification.
    (i) After finding of infringement. An applicant who has submitted a 
certification under paragraph (i)(1)(i)(A)(4) of this section and is 
sued for patent infringement within 45 days of the receipt of notice 
sent under Sec. 314.52 shall amend the certification if a final judgment 
in the action is entered finding the patent to be infringed unless the 
final judgment also finds the patent to be invalid. In the amended 
certification, the applicant shall certify under paragraph 
(i)(1)(i)(A)(3) of this section that the patent will expire on a 
specific date.
    (ii) After removal of a patent from the list. If a patent is removed 
from the list, any applicant with a pending application (including a 
tentatively approved application with a delayed effective date) who has 
made a certification with respect to such patent shall amend its 
certification. The applicant shall certify under paragraph (i)(1)(ii) of 
this section that no patents described in paragraph (i)(1)(i) of this 
section claim the drug or, if other relevant patents claim the drug, 
shall amend the certification to refer only to those relevant patents. 
In the amendment, the applicant shall state the reason for the change in 
certification (that the patent is or has been removed from the list). A 
patent that is the subject of a lawsuit under Sec. 314.107(c) shall not 
be removed from the list until FDA determines either that no delay in 
effective dates of approval is required under that section as a result 
of the lawsuit, that the patent has expired, or that any such period of 
delay in effective dates of approval is ended. An applicant shall submit 
an amended certification as an amendment to a pending application. Once 
an amendment for the change has been submitted, the application will no 
longer be considered to be one containing a certification under 
paragraph (i)(1)(i)(A)(4) of this section.
    (iii) Other amendments. (A) Except as provided in paragraphs (i)(4) 
and (i)(6)(iii)(B) of this section, an applicant shall amend a submitted 
certification if, at any time before the effective date of the approval 
of the application, the applicant learns that the submitted 
certification is no longer accurate.
    (B) An applicant is not required to amend a submitted certification 
when information on an otherwise applicable patent is submitted after 
the effective date of approval for the 505(b)(2) application.
    (j) Claimed exclusivity. A new drug product, upon approval, may be 
entitled to a period of marketing exclusivity under the provisions of 
Sec. 314.108. If an applicant believes its drug product is entitled to a 
period of exclusivity, it shall submit with the new drug application 
prior to approval the following information:
    (1) A statement that the applicant is claiming exclusivity.
    (2) A reference to the appropriate paragraph under Sec. 314.108 that 
supports its claim.
    (3) If the applicant claims exclusivity under Sec. 314.108(b)(2), 
information to show that, to the best of its knowledge or belief, a drug 
has not previously been approved under section 505(b) of the act 
containing any active moiety

[[Page 111]]

in the drug for which the applicant is seeking approval.
    (4) If the applicant claims exclusivity under Sec. 314.108(b)(4) or 
(b)(5), the following information to show that the application contains 
``new clinical investigations'' that are ``essential to approval of the 
application or supplement'' and were ``conducted or sponsored by the 
applicant:''
    (i) ``New clinical investigations.'' A certification that to the 
best of the applicant's knowledge each of the clinical investigations 
included in the application meets the definition of ``new clinical 
investigation'' set forth in Sec. 314.108(a).
    (ii) ``Essential to approval.'' A list of all published studies or 
publicly available reports of clinical investigations known to the 
applicant through a literature search that are relevant to the 
conditions for which the applicant is seeking approval, a certification 
that the applicant has thoroughly searched the scientific literature 
and, to the best of the applicant's knowledge, the list is complete and 
accurate and, in the applicant's opinion, such published studies or 
publicly available reports do not provide a sufficient basis for the 
approval of the conditions for which the applicant is seeking approval 
without reference to the new clinical investigation(s) in the 
application, and an explanation as to why the studies or reports are 
insufficient.
    (iii) ``Conducted or sponsored by.'' If the applicant was the 
sponsor named in the Form FDA-1571 for an investigational new drug 
application (IND) under which the new clinical investigation(s) that is 
essential to the approval of its application was conducted, 
identification of the IND by number. If the applicant was not the 
sponsor of the IND under which the clinical investigation(s) was 
conducted, a certification that the applicant or its predecessor in 
interest provided substantial support for the clinical investigation(s) 
that is essential to the approval of its application, and information 
supporting the certification. To demonstrate ``substantial support,'' an 
applicant must either provide a certified statement from a certified 
public accountant that the applicant provided 50 percent or more of the 
cost of conducting the study or provide an explanation of why FDA should 
consider the applicant to have conducted or sponsored the study if the 
applicant's financial contribution to the study is less than 50 percent 
or the applicant did not sponsor the investigational new drug. A 
predecessor in interest is an entity, e.g., a corporation, that the 
applicant has taken over, merged with, or purchased, or from which the 
applicant has purchased all rights to the drug. Purchase of nonexclusive 
rights to a clinical investigation after it is completed is not 
sufficient to satisfy this definition.
    (k) Financial certification or disclosure statement. The application 
shall contain a financial certification or disclosure statement or both 
as required by part 54 of this chapter.
    (l) Format of an original application. (1) The applicant shall 
submit a complete archival copy of the application that contains the 
information required under paragraphs (a) through (f) of this section. 
FDA will maintain the archival copy during the review of the application 
to permit individual reviewers to refer to information that is not 
contained in their particular technical sections of the application, to 
give other agency personnel access to the application for official 
business, and to maintain in one place a complete copy of the 
application. An applicant may submit on microfiche the portions of the 
archival copy of the application described in paragraphs (b) through (d) 
of this section. Information relating to samples and labeling, described 
in paragraph (e) of this section, is required to be submitted in hard 
copy. Tabulations of patient data and case report forms, described in 
paragraph (f) of this section, may be submitted on microfiche only if 
the applicant and FDA agree. If FDA agrees, the applicant may use 
another suitable microform system.
    (2) The applicant shall submit a review copy of the application. 
Each of the technical sections, described in paragraphs (d)(1) through 
(d)(6) of this section, in the review copy is required to be separately 
bound with a copy of the application form required under paragraph (a) 
of this section and a copy of the summary required under paragraph (c) 
of this section.

[[Page 112]]

    (3) The applicant shall submit a field copy of the application that 
contains the technical section described in paragraph (d)(1) of this 
section, a copy of the application form required under paragraph (a) of 
this section, a copy of the summary required under paragraph (c) of this 
section, and a certification that the field copy is a true copy of the 
technical section described in paragraph (d)(1) of this section 
contained in the archival and review copies of the application.
    (4) The applicant may obtain from FDA sufficient folders to bind the 
archival, the review, and the field copies of the application.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0001)

[50 FR 7493, Feb. 22, 1985; 50 FR 14212, Apr. 11, 1985, as amended at 50 
FR 16668, Apr. 26, 1985; 50 FR 21238, May 23, 1985; 52 FR 8847, Mar. 19, 
1987; 55 FR 11580, Mar. 29, 1990; 57 FR 17982, Apr. 28, 1992; 58 FR 
47351, Sept. 8, 1993; 59 FR 13200, Mar. 21, 1994; 59 FR 50361, Oct. 3, 
1994; 59 FR 60051, Nov. 21, 1994; 62 FR 40599, July 29, 1997; 63 FR 
5252, Feb. 2, 1998; 63 FR 6862, Feb. 11, 1998]

    Effective Date Note: 1. At 63 FR 5252, Feb. 2, 1998, Sec. 314.50 was 
amended by redesignating paragraph (k) as paragraph (l) and by adding a 
new paragraph (k), effective Feb. 2, 1999.
    2. At 63 FR 6862, Feb. 11, 1998, Sec. 314.50 was amended by revising 
the second sentence and adding two new sentences after the second 
sentence in paragraph (d)(5)(v), and by adding two new sentences after 
the first sentence in paragraph (d)(5)(vi)(a), effective Aug. 10, 1998. 
For the convenience of the user, the superseded text is set forth as 
follows:

Sec. 314.50  Content and format of an application.

                                * * * * *

    (d) * * *
    (5) * * *
    (v) * * * Evidence is also required to support the dosage and 
administration section of the labeling, including support for the dosage 
and dose interval recommended, and modifications for specific subgroups 
(for example, pediatrics, geriatrics, patients with renal failure).

                                * * * * *



Sec. 314.52  Notice of certification of invalidity or noninfringement of a patent.

    (a) Notice of certification. For each patent which claims the drug 
or drugs on which investigations that are relied upon by the applicant 
for approval of its application were conducted or which claims a use for 
such drug or drugs and which the applicant certifies under 
Sec. 314.50(i)(1)(i)(A)(4) that a patent is invalid, unenforceable, or 
will not be infringed, the applicant shall send notice of such 
certification by registered or certified mail, return receipt requested 
to each of the following persons:
    (1) Each owner of the patent that is the subject of the 
certification or the representative designated by the owner to receive 
the notice. The name and address of the patent owner or its 
representative may be obtained from the United States Patent and 
Trademark Office; and
    (2) The holder of the approved application under section 505(b) of 
the act for each drug product which is claimed by the patent or a use of 
which is claimed by the patent and for which the applicant is seeking 
approval, or, if the application holder does not reside or maintain a 
place of business within the United States, the application holder's 
attorney, agent, or other authorized official. The name and address of 
the application holder or its attorney, agent, or authorized official 
may be obtained from the Division of Drug Information Resources (HFD-
80), Center for Drug Evaluation and Research, Food and Drug 
Administration, 5600 Fishers Lane, Rockville, MD 20857.
    (3) This paragraph does not apply to a use patent that claims no 
uses for which the applicant is seeking approval.
    (b) Sending the notice. The applicant shall send the notice required 
by paragraph (a) of this section when it receives from FDA an 
acknowledgment letter stating that its application has been filed. At 
the same time, the applicant shall amend its application to include a 
statement certifying that the notice has been provided to each person 
identified under paragraph (a) of this section and that the notice met 
the

[[Page 113]]

content requirement under paragraph (c) of this section.
    (c) Content of a notice. In the notice, the applicant shall cite 
section 505(b)(3)(B) of the act and shall include, but not be limited 
to, the following information:
    (1) A statement that a 505(b)(2) application submitted by the 
applicant has been filed by FDA.
    (2) The application number.
    (3) The established name, if any, as defined in section 502(e)(3) of 
the act, of the proposed drug product.
    (4) The active ingredient, strength, and dosage form of the proposed 
drug product.
    (5) The patent number and expiration date, as submitted to the 
agency or as known to the applicant, of each patent alleged to be 
invalid, unenforceable, or not infringed.
    (6) A detailed statement of the factual and legal basis of the 
applicant's opinion that the patent is not valid, unenforceable, or will 
not be infringed. The applicant shall include in the detailed statement:
    (i) For each claim of a patent alleged not to be infringed, a full 
and detailed explanation of why the claim is not infringed.
    (ii) For each claim of a patent alleged to be invalid or 
unenforceable, a full and detailed explanation of the grounds supporting 
the allegation.
    (7) If the applicant does not reside or have a place of business in 
the United States, the name and address of an agent in the United States 
authorized to accept service of process for the applicant.
    (d) Amendment to an application. If an application is amended to 
include the certification described in Sec. 314.50(i), the applicant 
shall send the notice required by paragraph (a) of this section at the 
same time that the amendment to the application is submitted to FDA.
    (e) Documentation of receipt of notice. The applicant shall amend 
its application to document receipt of the notice required under 
paragraph (a) of this section by each person provided the notice. The 
applicant shall include a copy of the return receipt or other similar 
evidence of the date the notification was received. FDA will accept as 
adequate documentation of the date of receipt a return receipt or a 
letter acknowledging receipt by the person provided the notice. An 
applicant may rely on another form of documentation only if FDA has 
agreed to such documentation in advance. A copy of the notice itself 
need not be submitted to the agency.
    (f) Approval. If the requirements of this section are met, the 
agency will presume the notice to be complete and sufficient, and it 
will count the day following the date of receipt of the notice by the 
patent owner or its representative and by the approved application 
holder as the first day of the 45-day period provided for in section 
505(c)(3)(C) of the act. FDA may, if the applicant amends its 
application with a written statement that a later date should be used, 
count from such later date.
[59 FR 50362, Oct. 3, 1994]



Sec. 314.53  Submission of patent information.

    (a) Who must submit patent information. This section applies to any 
applicant who submits to FDA a new drug application or an amendment to 
it under section 505(b) of the act and Sec. 314.50 or a supplement to an 
approved application under Sec. 314.70, except as provided in paragraph 
(d)(2) of this section.
    (b) Patents for which information must be submitted. An applicant 
described in paragraph (a) of this section shall submit information on 
each patent that claims the drug or a method of using the drug that is 
the subject of the new drug application or amendment or supplement to it 
and with respect to which a claim of patent infringement could 
reasonably be asserted if a person not licensed by the owner of the 
patent engaged in the manufacture, use, or sale of the drug product. For 
purposes of this part, such patents consist of drug substance 
(ingredient) patents, drug product (formulation and composition) 
patents, and method of use patents. Process patents are not covered by 
this section and information on process patents may not be submitted to 
FDA. For patents that claim a drug substance or drug product, the 
applicant shall submit information only on those patents that claim a 
drug product that is the

[[Page 114]]

subject of a pending or approved application, or that claim a drug 
substance that is a component of such a product. For patents that claim 
a method of use, the applicant shall submit information only on those 
patents that claim indications or other conditions of use of a pending 
or approved application.
    (c) Reporting requirements--(1) General requirements. An applicant 
described in paragraph (a) of this section shall submit the following 
information for each patent described in paragraph (b) of this section:
    (i) Patent number and the date on which the patent will expire.
    (ii) Type of patent, i.e., drug, drug product, or method of use.
    (iii) Name of the patent owner.
    (iv) If the patent owner or applicant does not reside or have a 
place of business within the United States, the name of an agent 
(representative) of the patent owner or applicant who resides or 
maintains a place of business within the United States authorized to 
receive notice of patent certification under section 505(b)(3) and 
(j)(2)(B) of the act and Secs. 314.52 and 314.95.
    (2) Formulation, composition, or method of use patents--(i) Original 
declaration. For each formulation, composition, or method of use patent, 
in addition to the patent information described in paragraph (c)(1) of 
this section the applicant shall submit the following declaration:

    The undersigned declares that Patent No. ________ covers the 
formulation, composition, and/or method of use of (name of drug 
product). This product is (currently approved under section 505 of the 
Federal Food, Drug, and Cosmetic Act) [or] (the subject of this 
application for which approval is being sought):
 _______________________________________________________________________

    (ii) Amendment of patent information upon approval. Within 30 days 
after the date of approval of its application, if the application 
contained a declaration required under paragraph (c)(2)(i) of this 
section, the applicant shall by letter amend the declaration to identify 
each patent that claims the formulation, composition, or the specific 
indications or other conditions of use that have been approved.
    (3) No relevant patents. If the applicant believes that there are no 
patents which claim the drug or the drug product or which claim a method 
of using the drug product and with respect to which a claim of patent 
infringement could reasonably be asserted if a person not licensed by 
the owner of the patent engaged in the manufacture, use, or sale of the 
drug product, it shall so declare.
    (4) Authorized signature. The declarations required by this section 
shall be signed by the applicant or patent owner, or the applicant's or 
patent owner's attorney, agent (representative), or other authorized 
official.
    (d) When and where to submit patent information--(1) Original 
application. An applicant shall submit with its original application 
submitted under this part, including an application described in section 
505(b)(2) of the act, the information described in paragraph (c) of this 
section on each drug (ingredient), drug product (formulation and 
composition), and method of use patent issued before the application is 
filed with FDA and for which patent information is required to be 
submitted under this section. If a patent is issued after the 
application is filed with FDA but before the application is approved, 
the applicant shall, within 30 days of the date of issuance of the 
patent, submit the required patent information in an amendment to the 
application under Sec. 314.60.
    (2) Supplements. (i) An applicant shall submit patent information 
required under paragraph (c) of this section for a patent that claims 
the drug, drug product, or method of use for which approval is sought in 
any of the following supplements:
    (A) To change the formulation;
    (B) To add a new indication or other condition of use, including a 
change in route of administration;
    (C) To change the strength;
    (D) To make any other patented change regarding the drug, drug 
product, or any method of use.
    (ii) If the applicant submits a supplement for one of the changes 
listed under paragraph (d)(2)(i) of this section and existing patents 
for which information has already been submitted to FDA claim the 
changed product, the applicant shall submit a certification with the 
supplement identifying the

[[Page 115]]

patents that claim the changed product.
    (iii) If the applicant submits a supplement for one of the changes 
listed under paragraph (d)(2)(i) of this section and no patents, 
including previously submitted patents, claim the changed product, it 
shall so certify.
    (iv) The applicant shall comply with the requirements for amendment 
of formulation or composition and method of use patent information under 
paragraphs (c)(2)(ii) and (d)(3) of this section.
    (3) Patent information deadline. If a patent is issued for a drug, 
drug product, or method of use after an application is approved, the 
applicant shall submit to FDA the required patent information within 30 
days of the date of issuance of the patent.
    (4) Copies. The applicant shall submit two copies of each submission 
of patent information, an archival copy and a copy for the chemistry, 
manufacturing, and controls section of the review copy, to the Central 
Document Room, Center for Drug Evaluation and Research, Food and Drug 
Administration, Park Bldg., rm. 2-14, 12420 Parklawn Dr., Rockville, MD 
20857. The applicant shall submit the patent information by letter 
separate from, but at the same time as, submission of the supplement.
    (5) Submission date. Patent information shall be considered to be 
submitted to FDA as of the date the information is received by the 
Central Document Room.
    (6) Identification. Each submission of patent information, except 
information submitted with an original application, and its mailing 
cover shall bear prominent identification as to its contents, i.e., 
``Patent Information,'' or, if submitted after approval of an 
application, ``Time Sensitive Patent Information.''
    (e) Public disclosure of patent information. FDA will publish in the 
list the patent number and expiration date of each patent that is 
required to be, and is, submitted to FDA by an applicant, and for each 
use patent, the approved indications or other conditions of use covered 
by a patent. FDA will publish such patent information upon approval of 
the application, or, if the patent information is submitted by the 
applicant after approval of an application as provided under paragraph 
(d)(2) of this section, as soon as possible after the submission to the 
agency of the patent information. Patent information submitted by the 
last working day of a month will be published in that month's supplement 
to the list. Patent information received by the agency between monthly 
publication of supplements to the list will be placed on public display 
in FDA's Freedom of Information Staff. A request for copies of the file 
shall be sent in writing to the Freedom of Information Staff (HFI-35), 
Food and Drug Administration, rm. 12A-16, 5600 Fishers Lane, Rockville, 
MD 20857.
    (f) Correction of patent information errors. If any person disputes 
the accuracy or relevance of patent information submitted to the agency 
under this section and published by FDA in the list, or believes that an 
applicant has failed to submit required patent information, that person 
must first notify the agency in writing stating the grounds for 
disagreement. Such notification should be directed to the Drug 
Information Services Branch (HFD-84), Center for Drug Evaluation and 
Research, Food and Drug Administration, 5600 Fishers Lane, Rockville, MD 
20857. The agency will then request of the applicable new drug 
application holder that the correctness of the patent information or 
omission of patent information be confirmed. Unless the application 
holder withdraws or amends its patent information in response to FDA's 
request, the agency will not change the patent information in the list. 
If the new drug application holder does not change the patent 
information submitted to FDA, a 505(b)(2) application or an abbreviated 
new drug application under section 505(j) of the act submitted for a 
drug that is claimed by a patent for which information has been 
submitted must, despite any disagreement as to the correctness of the 
patent information, contain an appropriate certification for each listed 
patent.
[59 FR 50363, Oct. 3, 1994]

[[Page 116]]



Sec. 314.54  Procedure for submission of an application requiring investigations for approval of a new indication for, or other change from, a listed drug.

    (a) The act does not permit approval of an abbreviated new drug 
application for a new indication, nor does it permit approval of other 
changes in a listed drug if investigations, other than bioavailability 
or bioequivalence studies, are essential to the approval of the change. 
Any person seeking approval of a drug product that represents a 
modification of a listed drug (e.g., a new indication or new dosage 
form) and for which investigations, other than bioavailability or 
bioequivalence studies, are essential to the approval of the changes 
may, except as provided in paragraph (b) of this section, submit a 
505(b)(2) application. This application need contain only that 
information needed to support the modification(s) of the listed drug.
    (1) The applicant shall submit a complete archival copy of the 
application that contains the following:
    (i) The information required under Sec. 314.50(a), (b), (c), (d)(1), 
(d)(3), (e), and (g), except that Sec. 314.50(d)(1)(ii)(c) shall contain 
the proposed or actual master production record, including a description 
of the equipment, to be used for the manufacture of a commercial lot of 
the drug product.
    (ii) The information required under Sec. 314.50 (d)(2), (d)(4) (if 
an anti-infective drug), (d)(5), (d)(6), and (f) as needed to support 
the safety and effectiveness of the drug product.
    (iii) Identification of the listed drug for which FDA has made a 
finding of safety and effectiveness and on which finding the applicant 
relies in seeking approval of its proposed drug product by established 
name, if any, proprietary name, dosage form, strength, route of 
administration, name of listed drug's application holder, and listed 
drug's approved application number.
    (iv) If the applicant is seeking approval only for a new indication 
and not for the indications approved for the listed drug on which the 
applicant relies, a certification so stating.
    (v) Any patent information required under section 505(b)(1) of the 
act with respect to any patent which claims the drug for which approval 
is sought or a method of using such drug and to which a claim of patent 
infringement could reasonably be asserted if a person not licensed by 
the owner of the patent engaged in the manufacture, use, or sale of the 
drug product.
    (vi) Any patent certification or statement required under section 
505(b)(2) of the act with respect to any relevant patents that claim the 
listed drug or that claim any other drugs on which investigations relied 
on by the applicant for approval of the application were conducted, or 
that claim a use for the listed or other drug.
    (vii) If the applicant believes the change for which it is seeking 
approval is entitled to a period of exclusivity, the information 
required under Sec. 314.50(j).
    (2) The applicant shall submit a review copy that contains the 
technical sections described in Sec. 314.50(d)(1), except that 
Sec. 314.50(d)(1)(ii)(c) shall contain the proposed or actual master 
production record, including a description of the equipment, to be used 
for the manufacture of a commercial lot of the drug product, and 
paragraph (d)(3), and the technical sections described in paragraphs 
(d)(2), (d)(4), (d)(5), (d)(6), and (f) when needed to support the 
modification. Each of the technical sections in the review copy is 
required to be separately bound with a copy of the information required 
under Sec. 314.50 (a), (b), and (c) and a copy of the proposed labeling.
    (3) The information required by Sec. 314.50 (d)(2), (d)(4) (if an 
anti-infective drug), (d)(5), (d)(6), and (f) for the listed drug on 
which the applicant relies shall be satisfied by reference to the listed 
drug under paragraph (a)(1)(iii) of this section.
    (4) The applicant shall submit a field copy of the application that 
contains the technical section described in Sec. 314.50(d)(1), a copy of 
the information required under Sec. 314.50(a) and (c), and certification 
that the field copy is a true copy of the technical section described in 
Sec. 314.50(d)(1) contained in the archival and review copies of the 
application.
    (b) An application may not be submitted under this section for a 
drug

[[Page 117]]

product whose only difference from the reference listed drug is that:
    (1) The extent to which its active ingredient(s) is absorbed or 
otherwise made available to the site of action is less than that of the 
reference listed drug; or
    (2) The rate at which its active ingredient(s) is absorbed or 
otherwise made available to the site of action is unintentionally less 
than that of the reference listed drug.
[57 FR 17982, Apr. 28, 1992; 57 FR 61612, Dec. 28, 1992, as amended at 
58 FR 47351, Sept. 8, 1993; 59 FR 50364, Oct. 3, 1994]



Sec. 314.60  Amendments to an unapproved application.

    (a) Except as provided in paragraph (b) of this section, the 
applicant may submit an amendment to an application that is filed under 
Sec. 314.100, but not yet approved. The submission of a major amendment 
(for example, an amendment that contains significant new data from a 
previously unreported study or detailed newnalyses of previously 
submitted data), whether on the applicant's own initiative or at the 
invitation of the agency, constitutes an agreement by the applicant 
under section 505(c) of the act to extend the date by which the agency 
is required to reach a decision on the application. Ordinarily, the 
agency will extend the review period for a major amendment but only for 
the time necessary to review the new information. However, the agency 
may not extend the review period more than 180 days. If the agency 
extends the review period for the application, the director of the 
division responsible for reviewing the application will notify the 
applicant of the length of the extension. The submission of an amendment 
that is not a major amendment will not extend the review period. An 
amendment that contains new clinical data from a previously unreported 
study shall contain a financial certification or disclosure statement or 
both as required by part 54 of this chapter, or FDA may refuse to accept 
any such amendment.
    (b)(1) An unapproved application may not be amended if all of the 
following conditions apply:
    (i) The unapproved application is for a drug for which a previous 
application has been approved and granted a period of exclusivity in 
accordance with section 505(c)(3)(D)(ii) of the act that has not 
expired;
    (ii) The applicant seeks to amend the unapproved application to 
include a published report of an investigation that was conducted or 
sponsored by the applicant entitled to exclusivity for the drug;
    (iii) The applicant has not obtained a right of reference to the 
investigation described in paragraph (b)(1)(ii) of this section; and
    (iv) The report of the investigation described in paragraph 
(b)(1)(ii) of this section would be essential to the approval of the 
unapproved application.
    (2) The submission of an amendment described in paragraph (b)(1) of 
this section will cause the unapproved application to be deemed to be 
withdrawn by the applicant under Sec. 314.65 on the date of receipt by 
FDA of the amendment. The amendment will be considered a resubmission of 
the application, which may not be accepted except as provided in 
accordance with section 505(c)(3)(D)(ii) of the act.
    (c) The applicant shall submit a field copy of each amendment to 
Sec. 314.50(d)(1). The applicant, other than a foreign applicant, shall 
include in its submission of each such amendment to FDA a statement 
certifying that a field copy of the amendment has been sent to the 
applicant's home FDA district office.
[50 FR 7493, Feb. 22, 1985, as amended at 57 FR 17983, Apr. 28, 1992; 58 
FR 47352, Sept. 8, 1993; 63 FR 5252, Feb. 2, 1998]

    Effective Date Note: At 63 FR 5252, Feb. 2, 1998, Sec. 314.60 was 
amended in paragraph (a) by adding a new sentence at the end of the 
paragraph, effective Feb. 2, 1999.



Sec. 314.65  Withdrawal by the applicant of an unapproved application.

    An applicant may at any time withdraw an application that is not yet 
approved by notifying the Food and Drug Administration in writing. The 
agency will consider an applicant's failure to respond within 10 days to 
an approvable letter under Sec. 314.110 or a not approvable letter under 
Sec. 314.120 to be a request by the applicant to withdraw the 
application. A decision to withdraw the application is without prejudice 
to

[[Page 118]]

refiling. The agency will retain the application and will provide a copy 
to the applicant on request under the fee schedule in Sec. 20.42 of 
FDA's public information regulations.



Sec. 314.70  Supplements and other changes to an approved application.

    (a) Changes to an approved application. The applicant shall notify 
FDA about each change in each condition established in an approved 
application beyond the variations already provided for in the 
application. The notice is required to describe the change fully. 
Depending on the type of change, the applicant shall notify FDA about it 
in a supplemental application under paragraph (b) or (c) of this section 
or by inclusion of the information in the annual report to the 
application under paragraph (d) of this section. Notwithstanding the 
requirements of paragraphs (b) and (c) of this section, an applicant 
shall make a change provided for in those paragraphs (for example, the 
deletion of an ingredient common to many drug products) in accordance 
with a guideline, notice, or regulation published in the Federal 
Register that provides for a less burdensome notification of the change 
(for example, by notification at the time a supplement is submitted or 
in the next annual report). Except for a supplemental application 
providing for a change in the labeling, the applicant, other than a 
foreign applicant, shall include in each supplemental application 
providing for a change under paragraph (b) or (c) of this section a 
statement certifying that a field copy of the supplement has been 
provided to the applicant's home FDA district office.
    (b) Supplements requiring FDA approval before the change is made. An 
applicant shall submit a supplement, and obtain FDA approval of it, 
before making the changes listed below in the conditions in an approved 
application, unless the change is made to comply with an official 
compendium. An applicant may ask FDA to expedite its review of a 
supplement if a delay in making the change described in it would impose 
an extraordinary hardship on the applicant. Such a supplement and its 
mailing cover should be plainly marked: ``Supplement--Expedited Review 
Requested.''
    (1) Drug substance. A change affecting the drug substance to 
accomplish any of the following:
    (i) To relax the limits for a specification;
    (ii) To establish a new regulatory analytical method;
    (iii) To delete a specification or regulatory analytical method;
    (iv) To change the synthesis of the drug substance, including a 
change in solvents and a change in the route of synthesis.
    (v) To use a different facility or establishment to manufacture the 
drug substance, where: (a) the manufacturing process in the new facility 
or establishment differs materially from that in the former facility or 
establishment, or (b) the new facility or establishment has not received 
a satisfactory current good manufacturing practice (CGMP) inspection 
within the previous 2 years covering that manufacturing process.
    (2) Drug product. A change affecting the drug product to accomplish 
any of the following:
    (i) To add or delete an ingredient, or otherwise to change the 
composition of the drug product, other than deletion of an ingredient 
intended only to affect the color of the drug product;
    (ii) To relax the limits for a specification;
    (iii) To establish a new regulatory analytical method;
    (iv) To delete a specification or regulatory analytical method;
    (v) To change the method of manufacture of the drug product, 
including changing or relaxing an in-process control;
    (vi) To use a different facility or establishment, including a 
different contract laboratory or labeler, to manufacture, process, or 
pack the drug product;
    (vii) To change the container and closure system for the drug 
product (for example, glass to high density polyethylene (HDPE), or HDPE 
to polyvinyl chloride) or change a specification or regulatory 
analytical method for the container and closure system;
    (viii) To change the size of the container, except for solid dosage 
forms,

[[Page 119]]

without a change in the container and closure system.
    (ix) To extend the expiration date of the drug product based on data 
obtained under a new or revised stability testing protocol that has not 
been approved in the application.
    (x) To establish a new procedure for reprocessing a batch of the 
drug product that fails to meet specifications.
    (xi) To add a code imprint by printing with ink on a solid oral 
dosage form drug product.
    (xii) To add a code imprint by embossing, debossing, or engraving on 
a modified release solid oral dosage form drug product.
    (3) Labeling. Any change in labeling, except one described in 
paragraph (c)(2) or (d) of this section.
    (c) Supplements for changes that may be made before FDA approval. An 
applicant shall submit a supplement at the time the applicant makes any 
kind of change listed below in the conditions in an approved 
application, unless the change is made to comply with an official 
compendium. A supplement under this paragraph is required to give a full 
explanation of the basis for the change, identify the date on which the 
change is made, and, if the change concerns labeling, include 12 copies 
of final printed labeling. The applicant shall promptly revise all 
promotional labeling and drug advertising to make it consistent with any 
change in the labeling. The supplement and its mailing cover should be 
plainly marked: ``Special Supplement--Changes Being Effected.''
    (1) Adds a new specification or test method or changes in the 
methods, facilities (except a change to a new facility), or controls to 
provide increased assurance that the drug will have the characteristics 
of identity, strength, quality, and purity which it purports or is 
represented to possess;
    (2) Changes labeling to accomplish any of the following:
    (i) To add or strengthen a contraindication, warning, precaution, or 
adverse reaction;
    (ii) To add or strengthen a statement about drug abuse, dependence, 
or overdosage; or
    (iii) To add or strengthen an instruction about dosage and 
administration that is intended to increase the safe use of the product.
    (iv) To delete false, misleading, or unsupported indications for use 
or claims for effectiveness.
    (3) To use a different facility or establishment to manufacture the 
drug substance, where: (i) The manufacturing process in the new facility 
or establishment does not differ materially from that in the former 
facility or establishment, and (ii) the new facility or establishment 
has received a satisfactory current good manufacturing practice (CGMP) 
inspection within the previous 2 years covering that manufacturing 
process.
    (d) Changes described in the annual report. An applicant shall not 
submit a supplement to make any change in the conditions in an approved 
application, unless otherwise required under paragraph (b) or (c) of 
this section, but shall describe the change in the next annual report 
required under Sec. 314.81. Some examples of changes that can be 
described in the annual report are the following:
    (1) Any change made to comply with an official compendium.
    (2) A change in the labeling concerning the description of the drug 
product or in the information about how the drug product is supplied, 
that does not involve a change in the dosage strength or dosage form.
    (3) An editorial or similar minor change in labeling.
    (4) The deletion of an ingredient intended only to affect the color 
of the drug product.
    (5) An extension of the expiration date based upon full shelf-life 
data obtained from a protocol approved in the application.
    (6) A change within the container and closure system for the drug 
product (for example, a change from one high density polyethylene (HDPE) 
to another HDPE), except a change in container size for nonsolid dosage 
forms, based upon a showing of equivalency to the approved system under 
a protocol approved in the application or published in an official 
compendium.
    (7) The addition or deletion of an alternate analytical method.
    (8) A change in the size of a container for a solid dosage form, 
without a

[[Page 120]]

change from one container and closure system to another.
    (9) The addition by embossing, debossing, or engraving of a code 
imprint to a solid oral dosage form drug product other than a modified 
release dosage form, or a minor change in an existing code imprint.
    (e) Patent information. The applicant shall comply with the patent 
information requirements under section 505(c)(2) of the act.
    (f) Claimed exclusivity. If an applicant claims exclusivity under 
Sec. 314.108 upon approval of a supplemental application for a change to 
its previously approved drug product, the applicant shall include with 
its supplemental application the information required under 
Sec. 314.50(j).
    (g) Exception. An applicant proposing to make a change of a type 
described in paragraphs (a), (b)(1), (b)(2), (c)(1), (c)(3), (d)(1), and 
(d)(4) through (d)(9) of this section affecting a recombinant DNA-
derived protein/polypeptide product or a complex or conjugate of a drug 
with a monoclonal antibody regulated under the Federal Food, Drug, and 
Cosmetic Act shall comply with the following:
    (1) Changes requiring supplement submission and approval prior to 
distribution of the product made using the change (major changes). (i) A 
supplement shall be submitted for any change in the product, production 
process, quality controls, equipment, or facilities that has a 
substantial potential to have an adverse effect on the identity, 
strength, quality, purity, or potency of the product as they may relate 
to the safety or effectiveness of the product.
    (ii) These changes include, but are not limited to:
    (A) Changes in the qualitative or quantitative formulation or other 
specifications as provided in the approved application or in the 
regulations;
    (B) Changes requiring completion of an appropriate human study to 
demonstrate the equivalence of the identity, strength, quality, purity, 
or potency of the product as they may relate to the safety or 
effectiveness of the product;
    (C) Changes in the virus or adventitious agent removal or 
inactivation method(s);
    (D) Changes in the source material or cell line;
    (E) Establishment of a new master cell bank or seed; and
    (F) Changes which may affect product sterility assurance, such as 
changes in product or component sterilization method(s) or an addition, 
deletion, or substitution of steps in an aseptic processing operation.
    (iii) The applicant must obtain approval of the supplement from FDA 
prior to distribution of the product made using the change. Except for 
submissions under paragraph (g)(4) of this section, the following shall 
be contained in the supplement:
    (A) A detailed description of the proposed change;
    (B) The product(s) involved;
    (C) The manufacturing site(s) or area(s) affected;
    (D) A description of the methods used and studies performed to 
evaluate the effect of the change on the identity, strength, quality, 
purity, or potency of the product as they may relate to the safety or 
effectiveness of the product;
    (E) The data derived from such studies;
    (F) Relevant validation protocols and data; and
    (G) A reference list of relevant standard operating procedures 
(SOP's).
    (2) Changes requiring supplement submission at least 30 days prior 
to distribution of the product made using the change. (i) A supplement 
shall be submitted for any change in the product, production process, 
quality controls, equipment, or facilities that has a moderate potential 
to have an adverse effect on the identity, strength, quality, purity, or 
potency of the product as they may relate to the safety or effectiveness 
of the product. The supplement shall be labeled ``Supplement--Changes 
Being Effected in 30 Days'' or, if applicable under paragraph (g)(2)(v) 
of this section, ``Supplement--Changes Being Effected.''
    (ii) These changes include, but are not limited to:
    (A) Change in the site of testing from one facility to another;
    (B) An increase or decrease in production scale during finishing 
steps that involves new or different equipment; and

[[Page 121]]

    (C) Replacement of equipment with that of similar, but not 
identical, design and operating principle that does not affect the 
process methodology or process operating parameters.
    (iii) Pending approval of the supplement by FDA, and except as 
provided in paragraph (g)(2)(v) of this section, distribution of the 
product made using the change may begin not less than 30 days after 
receipt of the supplement by FDA. The information listed in paragraph 
(g)(1)(iii)(A) through (g)(1)(iii)(G) of this section shall be contained 
in the supplement.
    (iv) If within 30 days following FDA's receipt of the supplement, 
FDA informs the applicant that either:
    (A) The change requires approval prior to distribution of the 
product in accordance with paragraph (g)(1) of this section; or
    (B) Any of the information required under paragraph (g)(2)(iii) of 
this section is missing; the applicant shall not distribute the product 
made using the change until FDA determines that compliance with this 
section is achieved.
    (v) In certain circumstances, FDA may determine that, based on 
experience with a particular type of change, the supplement for such 
change is usually complete and provides the proper information, and on 
particular assurances that the proposed change has been appropriately 
submitted, the product made using the change may be distributed 
immediately upon receipt of the supplement by FDA. These circumstances 
may include substantial similarity with a type of change regularly 
involving a ``Supplement--Changes Being Effected'' supplement, or a 
situation in which the applicant presents evidence that the proposed 
change has been validated in accordance with an approved protocol for 
such change under paragraph (g)(4) of this section.
    (3) Changes to be described in an annual report (minor changes). (i) 
Changes in the product, production process, quality controls, equipment, 
or facilities that have a minimal potential to have an adverse effect on 
the identity, strength, quality, purity, or potency of the product as 
they may relate to the safety or effectiveness of the product shall be 
documented by the applicant in the next annual report in accordance with 
Sec. 314.81(b)(2)(iv).
    (ii) These changes include, but are not limited to:
    (A) Any change made to comply with an official compendium that is 
consistent with FDA requirements;
    (B) The deletion of an ingredient intended only to affect the color 
of the product;
    (C) An extension of an expiration date based upon full shelf life 
data obtained from a protocol approved in the application;
    (D) A change within the container and closure system for solid 
dosage forms, based upon a showing of equivalency to the approved system 
under a protocol approved in the application or published in an official 
compendium;
    (E) A change in the size of a container for a solid dosage form, 
without a change from one container and closure system to another;
    (F) The addition by embossing, debossing, or engraving of a code 
imprint to a solid dosage form drug product other than a modified 
release dosage form, or a minor change in an existing code imprint; and
    (G) The addition or deletion of an alternate analytical method.
    (4) An applicant may submit one or more protocols describing the 
specific tests and validation studies and acceptable limits to be 
achieved to demonstrate the lack of adverse effect for specified types 
of manufacturing changes on the identity, strength, quality, purity, or 
potency of the product as they may relate to the safety or effectiveness 
of the product. Any such protocols, or change to a protocol, shall be 
submitted as a supplement requiring approval from FDA prior to 
distribution of the product which, if approved, may justify a reduced 
reporting category for the particular change because the use of the 
protocol for that

[[Page 122]]

type of change reduces the potential risk of an adverse effect.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0001)

[50 FR 7493, Feb. 22, 1985; 50 FR 14212, Apr. 11, 1985, as amended at 50 
FR 21238, May 23, 1985; 57 FR 17983, Apr. 28, 1992; 58 FR 47352, Sept. 
8, 1993; 58 FR 47959, Sept. 13, 1993; 59 FR 50364, Oct. 3, 1994; 62 FR 
39900, July 24, 1997]



Sec. 314.71  Procedures for submission of a supplement to an approved application.

    (a) Only the applicant may submit a supplement to an application.
    (b) All procedures and actions that apply to an application under 
Sec. 314.50 also apply to supplements, except that the information 
required in the supplement is limited to that needed to support the 
change. A supplement is required to contain an archival copy and a 
review copy that include an application form and appropriate technical 
sections, samples, and labeling; except that a supplement for a change 
other than a change in labeling is required also to contain a field 
copy.
    (c) All procedures and actions that apply to applications under this 
part, including actions by applicants and the Food and Drug 
Administration, also apply to supplements.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0001)

[50 FR 7493, Feb. 22, 1985, as amended at 50 FR 21238, May 23, 1985; 58 
FR 47352, Sept. 8, 1993]



Sec. 314.72  Change in ownership of an application.

    (a) An applicant may transfer ownership of its application. At the 
time of transfer the new and former owners are required to submit 
information to the Food and Drug Administration as follows:
    (1) The former owner shall submit a letter or other document that 
states that all rights to the application have been transferred to the 
new owner.
    (2) The new owner shall submit an application form signed by the new 
owner and a letter or other document containing the following:
    (i) The new owner's commitment to agreements, promises, and 
conditions made by the former owner and contained in the application;
    (ii) The date that the change in ownership is effective; and
    (iii) Either a statement that the new owner has a complete copy of 
the approved application, including supplements and records that are 
required to be kept under Sec. 314.81, or a request for a copy of the 
application from FDA's files. FDA will provide a copy of the application 
to the new owner under the fee schedule in Sec. 20.42 of FDA's public 
information regulations.
    (b) The new owner shall advise FDA about any change in the 
conditions in the approved application under Sec. 314.70, except the new 
owner may advise FDA in the next annual report about a change in the 
drug product's label or labeling to change the product's brand or the 
name of its manufacturer, packer, or distributor.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0001)

[50 FR 7493, Feb. 22, 1985; 50 FR 14212, Apr. 11, 1985, as amended at 50 
FR 21238, May 23, 1985]



Sec. 314.80  Postmarketing reporting of adverse drug experiences.

    (a) Definitions. The following definitions of terms apply to this 
section:-
    Adverse drug experience. Any adverse event associated with the use 
of a drug in humans, whether or not considered drug related, including 
the following: An adverse event occurring in the course of the use of a 
drug product in professional practice; an adverse event occurring from 
drug overdose whether accidental or intentional; an adverse event 
occurring from drug abuse; an adverse event occurring from drug 
withdrawal; and any failure of expected pharmacological action.
    Disability. A substantial disruption of a person's ability to 
conduct normal life functions.
    Life-threatening adverse drug experience. Any adverse drug 
experience that places the patient, in the view of the initial reporter, 
at immediate risk of death from the adverse drug experience as it 
occurred, i.e., it does not include an adverse drug experience that, had 
it

[[Page 123]]

occurred in a more severe form, might have caused death.
    Serious adverse drug experience. Any adverse drug experience 
occurring at any dose that results in any of the following outcomes: 
Death, a life-threatening adverse drug experience, inpatient 
hospitalization or prolongation of existing hospitalization, a 
persistent or significant disability/incapacity, or a congenital 
anomaly/birth defect. Important medical events that may not result in 
death, be life-threatening, or require hospitalization may be considered 
a serious adverse drug experience when, based upon appropriate medical 
judgment, they may jeopardize the patient or subject and may require 
medical or surgical intervention to prevent one of the outcomes listed 
in this definition. Examples of such medical events include allergic 
bronchospasm requiring intensive treatment in an emergency room or at 
home, blood dyscrasias or convulsions that do not result in inpatient 
hospitalization, or the development of drug dependency or drug abuse.
    Unexpected adverse drug experience. Any adverse drug experience that 
is not listed in the current labeling for the drug product. This 
includes events that may be symptomatically and pathophysiologically 
related to an event listed in the labeling, but differ from the event 
because of greater severity or specificity. For example, under this 
definition, hepatic necrosis would be unexpected (by virtue of greater 
severity) if the labeling only referred to elevated hepatic enzymes or 
hepatitis. Similarly, cerebral thromboembolism and cerebral vasculitis 
would be unexpected (by virtue of greater specificity) if the labeling 
only listed cerebral vascular accidents. ``Unexpected,'' as used in this 
definition, refers to an adverse drug experience that has not been 
previously observed (i.e., included in the labeling) rather than from 
the perspective of such experience not being anticipated from the 
pharmacological properties of the pharmaceutical product.
    (b) Review of adverse drug experiences. Each applicant having an 
approved application under Sec. 314.50 or, in the case of a 505(b)(2) 
application, an effective approved application, shall promptly review 
all adverse drug experience information obtained or otherwise received 
by the applicant from any source, foreign or domestic, including 
information derived from commercial marketing experience, postmarketing 
clinical investigations, postmarketing epidemiological/surveillance 
studies, reports in the scientific literature, and unpublished 
scientific papers. Applicants are not required to resubmit to FDA 
adverse drug experience reports forwarded to the applicant by FDA; 
however, applicants must submit all followup information on such reports 
to FDA. Any person subject to the reporting requirements under paragraph 
(c) of this section shall also develop written procedures for the 
surveillance, receipt, evaluation, and reporting of postmarketing 
adverse drug experiences to FDA.
    (c) Reporting requirements. The applicant shall report to FDA 
adverse drug experience information, as described in this section. The 
applicant shall submit two copies of each report described in this 
section to the Central Document Room, 12229 Wilkins Ave., Rockville, MD 
20852. FDA may waive the requirement for the second copy in appropriate 
instances.
    (1)(i) Postmarketing 15-day ``Alert reports''. The applicant shall 
report each adverse drug experience that is both serious and unexpected, 
whether foreign or domestic, as soon as possible but in no case later 
than 15 calendar days of initial receipt of the information by the 
applicant.
    (ii) Postmarketing 15-day ``Alert reports''--followup. The applicant 
shall promptly investigate all adverse drug experiences that are the 
subject of these postmarketing 15-day Alert reports and shall submit 
followup reports within 15 calendar days of receipt of new information 
or as requested by FDA. If additional information is not obtainable, 
records should be maintained of the unsuccessful steps taken to seek 
additional information. Postmarketing 15-day Alert reports and followups 
to them shall be submitted under separate cover.
    (iii) Submission of reports. The requirements of paragraphs 
(c)(1)(i) and (c)(1)(ii) of this section, concerning the submission of 
postmarketing 15-day

[[Page 124]]

Alert reports, shall also apply to any person other than the applicant 
(nonapplicant) whose name appears on the label of an approved drug 
product as a manufacturer, packer, or distributor. To avoid unnecessary 
duplication in the submission to FDA of reports required by paragraphs 
(c)(1)(i) and (c)(1)(ii) of this section, obligations of a nonapplicant 
may be met by submission of all reports of serious adverse drug 
experiences to the applicant. If a nonapplicant elects to submit adverse 
drug experience reports to the applicant rather than to FDA, the 
nonapplicant shall submit each report to the applicant within 5 calendar 
days of receipt of the report by the nonapplicant, and the applicant 
shall then comply with the requirements of this section. Under this 
circumstance, the nonapplicant shall maintain a record of this action 
which shall include:
    (A) A copy of each adverse drug experience report;
    (B) The date the report was received by the nonapplicant;
    (C) The date the report was submitted to the applicant; and
    (D) The name and address of the applicant.
    (iv) Report identification. Each report submitted under this 
paragraph shall bear prominent identification as to its contents, i.e., 
``15-day Alert report,'' or ``15-day Alert report-followup.''
    (2) Periodic adverse drug experience reports. (i) The applicant 
shall report each adverse drug experience not reported under paragraph 
(c)(1)(i) of this section at quarterly intervals, for 3 years from the 
date of approval of the application, and then at annual intervals. The 
applicant shall submit each quarterly report within 30 days of the close 
of the quarter (the first quarter beginning on the date of approval of 
the application) and each annual report within 60 days of the 
anniversary date of approval of the application. Upon written notice, 
FDA may extend or reestablish the requirement that an applicant submit 
quarterly reports, or require that the applicant submit reports under 
this section at different times than those stated. For example, the 
agency may reestablish a quarterly reporting requirement following the 
approval of a major supplement. Followup information to adverse drug 
experiences submitted in a periodic report may be submitted in the next 
periodic report.
    (ii) Each periodic report is required to contain: (a) a narrative 
summary and analysis of the information in the report and an analysis of 
the 15-day Alert reports submitted during the reporting interval (all 
15-day Alert reports being appropriately referenced by the applicant's 
patient identification number, adverse reaction term(s), and date of 
submission to FDA); (b) a FDA Form 3500A (Adverse Reaction Report) for 
each adverse drug experience not reported under paragraph (c)(1)(i) of 
this section (with an index consisting of a line listing of the 
applicant's patient identification number and adverse reaction term(s)); 
and (c) a history of actions taken since the last report because of 
adverse drug experiences (for example, labeling changes or studies 
initiated).
    (iii) Periodic reporting, except for information regarding 15-day 
Alert reports, does not apply to adverse drug experience information 
obtained from postmarketing studies (whether or not conducted under an 
investigational new drug application), from reports in the scientific 
literature, and from foreign marketing experience.
    (d) Scientific literature. (1) A 15-day Alert report based on 
information from the scientific literature is required to be accompanied 
by a copy of the published article. The 15-day reporting requirements in 
paragraph (c)(1)(i) of this section (i.e., serious, unexpected adverse 
drug experiences) apply only to reports found in scientific and medical 
journals either as case reports or as the result of a formal clinical 
trial.
    (2) As with all reports submitted under paragraph (c)(1)(i) of this 
section, reports based on the scientific literature shall be submitted 
on FDA Form 3500A or comparable format as prescribed by paragraph (f) of 
this section. In cases where the applicant believes that preparing the 
FDA Form 3500A constitutes an undue hardship, the applicant may arrange 
with the Division of Pharmacovigilance and Epidemiology for an 
acceptable alternative reporting format.

[[Page 125]]

    (e) Postmarketing studies. (1) An applicant is not required to 
submit a 15-day Alert report under paragraph (c) of this section for an 
adverse drug experience obtained from a postmarketing study (whether or 
not conducted under an investigational new drug application) unless the 
applicant concludes that there is a reasonable possibility that the drug 
caused the adverse experience.
    (2) The applicant shall separate and clearly mark reports of adverse 
drug experiences that occur during a postmarketing study as being 
distinct from those experiences that are being reported spontaneously to 
the applicant.
    (f) Reporting FDA Form 3500A. (1) Except as provided in paragraph 
(f)(3) of this section, the applicant shall complete FDA Form 3500A for 
each report of an adverse drug experience (foreign events may be 
submitted either on an FDA Form 3500A or, if preferred, on a CIOMS I 
form).
    (2) Each completed FDA Form 3500A should refer only to an individual 
patient or a single attached publication.
    (3) Instead of using FDA Form 3500A, an applicant may use a 
computer-generated FDA Form 3500A or other alternative format (e.g., a 
computer-generated tape or tabular listing) provided that: (i) The 
content of the alternative format is equivalent in all elements of 
information to those specified in FDA Form 3500A; and (ii) The format is 
agreed to in advance by MedWatch: The FDA Medical Products Reporting 
Program.
    (4) Ten copies or fewer of FDA Form 3500A and/or a copy of the 
instructions for completing the form may be obtained from the Division 
of Pharmacovigilance and Epidemiology (HFD-730), Center for Drug 
Evaluation and Research, Food and Drug Administration, 5600 Fishers 
Lane, Rockville, MD 20857. More than 10 copies of the form may be 
obtained by writing to the Consolidated Forms and Publications 
Distribution Center, Washington Commerce Center, 3222 Hubbard Rd., 
Landover, MD 20785.
    (g) Multiple reports. An applicant should not include in reports 
under this section any adverse drug experiences that occurred in 
clinical trials if they were previously submitted as part of the 
approved application. If a report applies to a drug for which an 
applicant holds more than one approved application, the applicant should 
submit the report to the application that was first approved. If a 
report refers to more than one drug marketed by an applicant, the 
applicant should submit the report to the application for the drug 
listed first in the report.
    (h) Patient privacy. An applicant should not include in reports 
under this section the names and addresses of individual patients; 
instead, the applicant should assign a unique code number to each 
report, preferably not more than eight characters in length. The 
applicant should include the name of the reporter from whom the 
information was received. Names of patients, health care professionals, 
hospitals, and geographical identifiers in adverse drug experience 
reports are not releasable to the public under FDA's public information 
regulations in part 20.
    (i) Recordkeeping. The applicant shall maintain for a period of 10 
years records of all adverse drug experiences known to the applicant, 
including raw data and any correspondence relating to adverse drug 
experiences.
    (j) Withdrawal of approval. If an applicant fails to establish and 
maintain records and make reports required under this section, FDA may 
withdraw approval of the application and, thus, prohibit continued 
marketing of the drug product that is the subject of the application.
    (k) Disclaimer. A report or information submitted by an applicant 
under this section (and any release by FDA of that report or 
information) does not necessarily reflect a conclusion by the applicant 
or FDA that the report or information constitutes an admission that the 
drug caused or contributed to an adverse effect. An applicant need not 
admit, and may deny, that the report or information submitted under this 
section constitutes an admission that the drug caused or contributed to 
an adverse effect. For purposes of this

[[Page 126]]

provision, the term ``applicant'' also includes any person reporting 
under paragraph (c)(1)(iii) of this section.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0001)

[50 FR 7493, Feb. 22, 1985; 50 FR 14212, Apr. 11, 1985, as amended at 50 
FR 21238, May 23, 1985; 51 FR 24481, July 3, 1986; 52 FR 37936, Oct. 13, 
1987; 55 FR 11580, Mar. 29, 1990; 57 FR 17983, Apr. 28, 1992; 62 FR 
34168, June 25, 1997; 62 FR 52251, Oct. 7, 1997; 63 FR 14611, Mar. 26, 
1998]

    Effective Date Note: At 62 FR 52251, Oct. 7, 1997, Sec. 314.80 was 
amended by revising paragraphs (a), (c)(1), (f)(1), (f)(3)(ii), and 
(f)(4) and the introductory text of paragraph (c); by adding two new 
sentences at the end of paragraph (b); by removing in paragraph (d)(2) 
the words ``Epidemiology and Surveillance'' and adding in their place 
the words ``Pharmacovigilance and Epidemiology''; by removing in 
paragraphs (c)(2)(ii)(b), (d)(2), (f)(2), and (f)(3) and in the heading 
for paragraph (f) the words ``Form FDA-1639'' or ``FDA-1639'' and adding 
in their place the words ``FDA Form 3500A''; and by removing paragraph 
(j) and redesignating paragraphs (k) and (l) as paragraphs (j) and (k), 
respectively, effective Apr. 6, 1998. For the convenience of the user, 
the superseded text is set forth as follows:

Sec. 314.80  Postmarketing reporting of adverse drug experiences.

    (a) Definitions. The following definitions of terms apply to this 
section:
    Adverse drug experience means any adverse event associated with the 
use of a drug in humans, whether or not considered drug related, 
including the following: an adverse event occurring in the course of the 
use of a drug product in professional practice; an adverse event 
occurring from drug overdose, whether accidental or intentional; an 
adverse event occurring from drug abuse; an adverse event occurring from 
drug withdrawal; and any failure of expected pharmacological action.
    Serious means an adverse drug experience that is fatal or life-
threatening, is permanently disabling, requires inpatient 
hospitalization, or is a congenital anomaly, cancer, or overdose.
    Unexpected means an adverse drug experience that is not listed in 
the current labeling for the drug and includes an event that may be 
symptomatically and pathophysiologically related to an event listed in 
the labeling, but differs from the event because of greater severity or 
specificity. For example, under this definition, hepatic necrosis would 
be unexpected (by virtue of greater severity) if the labeling only 
referred to elevated hepatic enzymes or hepatitis. Similarly, cerebral 
thromboembolism and cerebral vasculitis would be unexpected (by virtue 
of greater specificity) if the labeling only listed cerebral vascular 
accidents.

                                * * * * *

    (c) Reporting requirements. The applicant shall report to FDA 
adverse drug experience information, as described in this section. The 
applicant shall submit two copies of each report described in this 
section to the Central Document Room, Park Bldg., Rm. 214, 12420 
Parklawn Dr., Rockville, MD 20852. FDA may waive the requirement for the 
second copy in appropriate instances.
    (1) Fifteen-day ``Alert reports.'' (i) The applicant shall report 
each adverse drug experience that is both serious and unexpected, 
regardless of source, as soon as possible but in any case within 15 
working days of initial receipt of the information. These reports are 
required to be submitted on Form FDA-1639 (Adverse Reaction Report). The 
applicant shall promptly investigate all adverse drug experiences that 
are the subject of these 15-day Alert reports and shall submit followup 
reports within 15 working days of receipt of new information or as 
requested by FDA. If additional information is not obtainable, a 
followup report may be required that describes briefly the steps taken 
to seek additional information and the reasons why it could not be 
obtained. These 15-day Alert reports and followups to them are required 
to be submitted under separate cover and may not be included, except for 
summary or tabular purposes, in a periodic report.
    (ii) The requirements of paragraph (c)(1)(i) of this section, 
concerning the submission of 15-day Alert reports, shall also apply to 
any person (other than the applicant) whose name appears on the label of 
an approved drug product as a manufacturer, packer, or distributor. 
However, to avoid unnecessary duplication in the submission to FDA of, 
and followup to, reports required by paragraph (c)(1)(i) of this 
section, obligations of a nonapplicant may be met by submission of all 
reports of serious adverse drug experiences to the applicant. If a 
nonapplicant elects to submit adverse drug experience reports to the 
applicant rather than to FDA, it shall submit each report to the 
applicant within 3 working days of its receipt by the nonapplicant, and 
the applicant shall then comply with the requirements of this section. 
Under this circumstance, the nonapplicant shall maintain a record of 
this action which shall include:
    (a) A copy of the drug experience report.
    (b) Date the report was received by the nonapplicant.
    (c) Date the report was submitted to the applicant.
    (d) Name and address of the applicant.

[[Page 127]]

    (iii) Each report submitted under this paragraph shall bear 
prominent identification as to its contents, i.e., ``15-day Alert 
report'' or ``15-day Alert report--followup.''

                                * * * * *

    (f) Reporting Form FDA-1639. (1) Except as provided in paragraph 
(f)(3) of this section, the applicant shall complete a Form FDA-1639 
(Adverse Reaction Report) for each report of an adverse drug experience.

                                * * * * *

    (3) * * *
    (ii) the format is agreed to in advance by the Division of 
Epidemiology and Surveillance (HFD-730).
    (4) Single copies of Form FDA-1639 may be obtained from the Division 
of Epidemiology and Surveillance (HFD-730), Center for Drug Evaluation 
and Research, Food and Drug Administration, 5600 Fishers Lane, 
Rockville, MD 20857. Supplies of Form FDA-1639 may be obtained from the 
PHS Forms and Publications Distribution Center, 12100 Parklawn Dr., 
Rockville, MD 20857.

                                * * * * *

    (j) Guideline. FDA has prepared under Sec. 10.90(b) a guideline for 
the submission of reports of adverse drug experiences and suggested 
followup investigation of reports.

                                * * * * *

    2. At 63 FR 14611, Mar. 26, 1998, Sec. 314.80 was amended by 
correcting the reference to ``paragraph (c)(1)(ii)'' in the last 
sentence in newly redesignated paragraph (k) to read ``paragraph 
(c)(1)(iii)'', effective Apr. 6, 1998.



Sec. 314.81  Other postmarketing reports.

    (a) Applicability. Each applicant shall make the reports for each of 
its approved applications and abbreviated applications required under 
this section and sections 505(k) and 507(g) of the act.
    (b) Reporting requirements. The applicant shall submit to the Food 
and Drug Administration at the specified times two copies of the 
following reports:
    (1) NDA--Field alert report. The applicant shall submit information 
of the following kinds about distributed drug products and articles to 
the FDA district office that is responsible for the facility involved 
within 3 working days of receipt by the applicant. The information may 
be provided by telephone or other rapid communication means, with prompt 
written followup. The report and its mailing cover should be plainly 
marked: ``NDA--Field Alert Report.''
    (i) Information concerning any incident that causes the drug product 
or its labeling to be mistaken for, or applied to, another article.
    (ii) Information concerning any bacteriological contn, or any 
significant chemical, physical, or other change or deterioration in the 
distributed drug product, or any failure of one or more distributed 
batches of the drug product to meet the specifications established for 
it in the application.
    (2) Annual report. The applicant shall submit the following 
information in the order listed each year within 60 days of the 
anniversary date of approval of the application. The applicant shall 
submit the report to the FDA division responsible for reviewing the 
application. Each annual report is required to be accompanied by a 
completed transmittal Form FDA-2252 (Transmittal of Periodic Reports for 
Drugs for Human Use) which may be obtained from the PHS Forms and 
Publications Distribution Center, 12100 Parklawn Dr., Rockville, MD 
20857, and is required to include all the information required under 
this section that the applicant received or otherwise obtained during 
the annual reporting interval which ends on the anniversary date. The 
report is required to contain the following:
    (i) Summary. A brief summary of significant new information from the 
previous year that might affect the safety, effectiveness, or labeling 
of the drug product. The report is also required to contain a brief 
description of actions the applicant has taken or intends to take as a 
result of this new information, for example, submit a labeling 
supplement, add a warning to the labeling, or initiate a new study.
    (ii) Distribution data. Information about the quantity of the drug 
product distributed under the approved application, including that 
distributed to distributors. The information is required to include the 
National Drug Code (NDC) number, the total number of dosage units of 
each strength or potency distributed (e.g., 100,000/5 milligram tablets, 
50,000/10 milliliter vials),

[[Page 128]]

and the quantities distributed for domestic use and the quantities 
distributed for foreign use. Disclosure of financial or pricing data is 
not required.
    (iii) Labeling. Currently used professional labeling, patient 
brochures or package inserts (if any), a representative sample of the 
package labels, and a summary of any changes in labeling that have been 
made since the last report listed by date in the order in which they 
were implemented, or if no changes, a statement of that fact.
    (iv) Chemistry, manufacturing, and controls changes. (a) Reports of 
experiences, investigations, studies, or tests involving chemical or 
physical properties, or any other properties of the drug (such as the 
drug's behavior or properties in relation to microorganisms, including 
both the effects of the drug on microorganisms and the effects of 
microorganisms on the drug). These reports are only required for new 
information that may affect FDA's previous conclusions about the safety 
or effectiveness of the drug product.
    (b) A full description of the manufacturing and controls changes not 
requiring a supplemental application under Sec. 314.70 (b) and (c), 
listed by date in the order in which they were implemented.
    (v) Nonclinical laboratory studies. Copies of unpublished reports 
and summaries of published reports of new toxicological findings in 
animal studies and in vitro studies (e.g., mutagenicity) conducted by, 
or otherwise obtained by, the applicant concerning the ingredients in 
the drug product. The applicant shall submit a copy of a published 
report if requested by FDA.
    (vi) Clinical data. (a) Published clinical trials of the drug (or 
abstracts of them), including clinical trials on safety and 
effectiveness; clinical trials on new uses; biopharmaceutic, 
pharmacokinetic, and clinical pharmacology studies; and reports of 
clinical experience pertinent to safety (for example, epidemiologic 
studies or analyses of experience in a monitored series of patients) 
conducted by or otherwise obtained by the applicant. Review articles, 
papers describing the use of the drug product in medical practice, 
papers and abstracts in which the drug is used as a research tool, 
promotional articles, press clippings, and papers that do not contain 
tabulations or summaries of original data should not be reported.
    (b) Summaries of completed unpublished clinical trials, or 
prepublication manuscripts if available, conducted by, or otherwise 
obtained by, the applicant. Supporting information should not be 
reported. (A study is considered completed 1 year after it is 
concluded.)
    (vii) Status reports. A statement on the current status of any 
postmarketing studies performed by, or on behalf of, the applicant. To 
facilitate communications between FDA and the applicant, the report may, 
at the applicant's discretion, also contain a list of any open 
regulatory business with FDA concerning the drug product subject to the 
application.
    (3) Other reporting--(i) Advertisements and promotional labeling. 
The applicant shall submit specimens of mailing pieces and any other 
labeling or advertising devised for promotion of the drug product at the 
time of initial dissemination of the labeling and at the time of initial 
publication of the advertisement for a prescription drug product. 
Mailing pieces and labeling that are designed to contain samples of a 
drug product are required to be complete, except the sample of the drug 
product may be omitted. Each submission is required to be accompanied by 
a completed transmittal Form FDA-2253 (Transmittal of Advertisements and 
Promotional Labeling for Drugs for Human Use) and is required to include 
a copy of the product's current professional labeling. Form FDA-2253 may 
be obtained from the PHS Forms and Publications Distribution Center, 
12100 Parklawn Dr., Rockville, MD 20857.
    (ii) Special reports. Upon written request the agency may require 
that the applicant submit the reports under this section at different 
times than those stated.
    (iii) Withdrawal of approved drug product from sale. (a) The 
applicant shall submit on Form FDA 2657 (Drug Product Listing), within 
15 working days of the withdrawal from sale of a drug product, the 
following information:
    (1) The National Drug Code (NDC) number.

[[Page 129]]

    (2) The identity of the drug product by established name and by 
proprietary name.
    (3) The new drug application or abbreviated application number.
    (4) The date of withdrawal from sale. It is requested but not 
required that the reason for withdrawal of the drug product from sale be 
included with the information.
    (b) The applicant shall submit each Form FDA-2657 to the Drug 
Listing Branch (HFD-334), Center for Drug Evaluation and Research, Food 
and Drug Administration, 5600 Fishers Lane, Rockville, MD 20857.
    (c) Reporting under paragraph (b)(3)(iii) of this section 
constitutes compliance with the requirements under Sec. 207.30(a) of 
this chapter to report ``at the discretion of the registrant when the 
change occurs.''
    (c) General requirements--(1) Multiple applications. For all reports 
required by this section, the applicant shall submit the information 
common to more than one application only to the application first 
approved, and shall not report separately on each application. The 
submission is required to identify all the applications to which the 
report applies.
    (2) Patient identification. Applicants should not include in reports 
under this section the names and addresses of individual patients; 
instead, the applicant should code the patient names whenever possible 
and retain the code in the applicant's files. The applicant shall 
maintain sufficient patient identification information to permit FDA, by 
using that information alone or along with records maintained by the 
investigator of a study, to identify the name and address of individual 
patients; this will ordinarily occur only when the agency needs to 
investigate the reports further or when there is reason to believe that 
the reports do not represent actual results obtained.
    (d) Withdrawal of approval. If an applicant fails to make reports 
required under this section, FDA may withdraw approval of the 
application and, thus, prohibit continued marketing of the drug product 
that is the subject of the application.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0001)

[50 FR 7493, Feb. 22, 1985; 50 FR 14212, Apr. 11, 1985, as amended at 50 
FR 21238, May 23, 1985; 55 FR 11580, Mar. 29, 1990; 57 FR 17983, Apr. 
28, 1992]



Sec. 314.90  Waivers.

    (a) An applicant may ask the Food and Drug Administration to waive 
under this section any requirement that applies to the applicant under 
Secs. 314.50 through 314.81. An applicant may ask FDA to waive under 
Sec. 314.126(c) any criteria of an adequate and well-controlled study 
described in Sec. 314.126(b). A waiver request under this section is 
required to be submitted with supporting documentation in an 
application, or in an amendment or supplement to an application. The 
waiver request is required to contain one of the following:
    (1) An explanation why the applicant's compliance with the 
requirement is unnecessary or cannot be achieved;
    (2) A description of an alternative submission that satisfies the 
purpose of the requirement; or
    (3) Other information justifying a waiver.
    (b) FDA may grant a waiver if it finds one of the following:
    (1) The applicant's compliance with the requirement is unnecessary 
for the agency to evaluate the application or compliance cannot be 
achieved;
    (2) The applicant's alternative submission satisfies the 
requirement; or
    (3) The applicant's submission otherwise justifies a waiver.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0001)

[50 FR 7493, Feb. 22, 1985, as amended at 50 FR 21238, May 23, 1985]



                   Subpart C--Abbreviated Applications

    Source:  57 FR 17983, Apr. 28, 1992, unless otherwise noted.

[[Page 130]]



Sec. 314.92  Drug products for which abbreviated applications may be submitted.

    (a) Abbreviated applications are suitable for the following drug 
products within the limits set forth under Sec. 314.93:
    (1) Drug products that are the same as a listed drug. A ``listed 
drug'' is defined in Sec. 314.3. For determining the suitability of an 
abbreviated new drug application, the term ``same as'' means identical 
in active ingredient(s), dosage form, strength, route of administration, 
and conditions of use, except that conditions of use for which approval 
cannot be granted because of exclusivity or an existing patent may be 
omitted. If a listed drug has been voluntarily withdrawn from or not 
offered for sale by its manufacturer, a person who wishes to submit an 
abbreviated new drug application for the drug shall comply with 
Sec. 314.122.
    (2) Drug products that are duplicates of, or that meet the monograph 
for, an antibiotic drug for which FDA has approved an application.
    (3) Drug products that have been declared suitable for an 
abbreviated new drug application submission by FDA through the petition 
procedures set forth under Sec. 10.30 of this chapter and Sec. 314.93.
    (b) FDA will publish in the list listed drugs for which abbreviated 
applications may be submitted. The list is available from the 
Superintendent of Documents, U.S. Government Printing Office, 
Washington, DC 20402, 202-783-3238.



Sec. 314.93  Petition to request a change from a listed drug.

    (a) The only changes from a listed drug for which the agency will 
accept a petition under this section are those changes described in 
paragraph (b) of this section. Petitions to submit abbreviated new drug 
applications for other changes from a listed drug will not be approved.
    (b) A person who wants to submit an abbreviated new drug application 
for a drug product which is not identical to a listed drug in route of 
administration, dosage form, and strength, or in which one active 
ingredient is substituted for one of the active ingredients in a listed 
combination drug, must first obtain permission from FDA to submit such 
an abbreviated application.
    (c) To obtain permission to submit an abbreviated new drug 
application for a change described in paragraph (b) of this section, a 
person must submit and obtain approval of a petition requesting the 
change. A person seeking permission to request such a change from a 
reference listed drug shall submit a petition in accordance with 
Sec. 10.20 of this chapter and in the format specified in Sec. 10.30 of 
this chapter. The petition shall contain the information specified in 
Sec. 10.30 of this chapter and any additional information required by 
this section. If any provision of Sec. 10.20 or Sec. 10.30 of this 
chapter is inconsistent with any provision of this section, the 
provisions of this section apply.
    (d) The petitioner shall identify a listed drug and include a copy 
of the proposed labeling for the drug product that is the subject of the 
petition and a copy of the approved labeling for the listed drug. The 
petitioner may, under limited circumstances, identify more than one 
listed drug, for example, when the proposed drug product is a 
combination product that differs from the combination reference listed 
drug with regard to an active ingredient, and the different active 
ingredient is an active ingredient of a listed drug. The petitioner 
shall also include information to show that:
    (1) The active ingredients of the proposed drug product are of the 
same pharmacological or therapeutic class as those of the reference 
listed drug.
    (2) The drug product can be expected to have the same therapeutic 
effect as the reference listed drug when administered to patients for 
each condition of use in the reference listed drug's labeling for which 
the applicant seeks approval.
    (3) If the proposed drug product is a combination product with one 
different active ingredient, including a different ester or salt, from 
the reference listed drug, that the different active ingredient has 
previously been approved in a listed drug or is a drug that does not 
meet the definition of ``new drug'' in section 201(b) of the act.

[[Page 131]]

    (e) No later than 90 days after the date a petition that is 
permitted under paragraph (a) of this section is submitted, FDA will 
approve or disapprove the petition.
    (1) FDA will approve a petition properly submited under this section 
unless it finds that:
    (i) Investigations must be conducted to show the safety and 
effectiveness of the drug product or of any of its active ingredients, 
its route of administration, dosage form, or strength which differs from 
the reference listed drug; or
    (ii) For a petition that seeks to change an active ingredient, the 
drug product that is the subject of the petition is not a combination 
drug; or
    (iii) For a combination drug product that is the subject of the 
petition and has an active ingredient different from the reference 
listed drug:
    (A) The drug product may not be adequately evaluated for approval as 
safe and effective on the basis of the information required to be 
submitted under Sec. 314.94; or
    (B) The petition does not contain information to show that the 
different active ingredient of the drug product is of the same 
pharmacological or therapeutic class as the ingredient of the reference 
listed drug that is to be changed and that the drug product can be 
expected to have the same therapeutic effect as the reference listed 
drug when administered to patients for each condition of use in the 
listed drug's labeling for which the applicant seeks approval; or
    (C) The different active ingredient is not an active ingredient in a 
listed drug or a drug that meets the requirements of section 201(p) of 
the act; or
    (D) The remaining active ingredients are not identical to those of 
the listed combination drug; or
    (iv) Any of the proposed changes from the listed drug would 
jeopardize the safe or effective use of the product so as to necessitate 
significant labeling changes to address the newly introduced safety or 
effectiveness problem; or
    (v) FDA has determined that the reference listed drug has been 
withdrawn from sale for safety or effectiveness reasons under 
Sec. 314.161, or the reference listed drug has been voluntarily 
withdrawn from sale and the agency has not determined whether the 
withdrawal is for safety or effectiveness reasons.
    (2) For purposes of this paragraph, ``investigations must be 
conducted'' means that information derived from animal or clinical 
studies is necessary to show that the drug product is safe or effective. 
Such information may be contained in published or unpublished reports.
    (3) If FDA approves a petition submitted under this section, the 
agency's response may describe what additional information, if any, will 
be required to support an abbreviated new drug application for the drug 
product. FDA may, at any time during the course of its review of an 
abbreviated new drug application, request additional information 
required to evaluate the change approved under the petition.
    (f) FDA may withdraw approval of a petition if the agency receives 
any information demonstrating that the petition no longer satisfies the 
conditions under paragraph (e) of this section.



Sec. 314.94  Content and format of an abbreviated application.

    Abbreviated applications are required to be submitted in the form 
and contain the information required under this section. Three copies of 
the application are required, an archival copy, a review copy, and a 
field copy. FDA will maintain guidelines on the format and content of 
applications to assist applicants in their preparation.
    (a) Abbreviated new drug applications. Except as provided in 
paragraph (b) of this section, the applicant shall submit a complete 
archival copy of the abbreviated new drug application that includes the 
following:
    (1) Application form. The applicant shall submit a completed and 
signed application form that contains the information described under 
Sec. 314.50(a)(1), (a)(3), (a)(4), and (a)(5). The applicant shall state 
whether the submission is an abbreviated application under this section 
or a supplement to an abbreviated application under Sec. 314.97.
    (2) Table of contents. the archival copy of the abbreviated new drug 
application is required to contain a table of

[[Page 132]]

contents that shows the volume number and page number of the contents of 
the submission.
    (3) Basis for abbreviated new drug application submission. An 
abbreviated new drug application must refer to a listed drug. 
Ordinarily, that listed drug will be the drug product selected by the 
agency as the reference standard for conducting bioequivalence testing. 
The application shall contain:
    (i) The name of the reference listed drug, including its dosage form 
and strength. For an abbreviated new drug application based on an 
approverd petition under Sec. 10.30 of this chapter or Sec. 314.93, the 
reference listed drug must be the same as the listed drug approved in 
the petition.
    (ii) A statement as to whether, according to the information 
published in the list, the reference listed drug is entitled to a period 
of marketing exclusivity under section 505(j)(4)(D) of the act.
    (iii) For an abbreviated new drug application based on an approved 
petition under Sec. 10.30 of this chapter or Sec. 314.93, a reference to 
FDA-assigned docket number for the petition and a copy of FDA's 
correspondence approving the petition.
    (4) Conditions of use. (i) A statement that the conditions of use 
prescribed, recommended, or suggested in the labeling proposed for the 
drug product have been previously approved for the reference listed 
drug.
    (ii) A reference to the applicant's annotated proposed labeling and 
to the currently approved labeling for the reference listed drug 
provided under paragraph (a)(8) of this section.
    (5) Active ingredients. (i) For a single-active-ingredient drug 
product, information to show that the active ingredient is the same as 
that of the reference single-active-ingredient listed drug, as follows:
    (A) A statement that the active ingredient of the proposed drug 
product is the same as that of the reference listed drug.
    (B) A reference to the applicant's annotated proposed labeling and 
to the currently approved labeling for the reference listed drug 
provided under paragraph (a)(8) of this section.
    (ii) For a combination drug product, information to show that the 
active ingredients are the same as those of the reference listed drug 
except for any different active ingredient that has been the subject of 
an approved petition, as follows:
    (A) A statement that the active ingredients of the proposed drug 
product are the same as those of the reference listed drug, or if one of 
the active ingredients differs from one of the active ingredients of the 
reference listed drug and the abbreviated application is submitted under 
the approval of a petition under Sec. 314.93 to vary such active 
ingredient, information to show that the other active ingredients of the 
drug product are the same as the other active ingredients of the 
reference listed drug, information to show that the different active 
ingredient is an active ingredient of another listed drug or of a drug 
that does not meet the definition of ``new drug'' in section 201(p) of 
the act, and such other information about the different active 
ingredient that FDA may require.
    (B) A reference to the applicant's annotated proposed labeling and 
to the currently approved labeling for the reference listed drug 
provided under paragraph (a)(8) of this section.
    (6) Route of administration, dosage form, and strength. (i) 
Information to show that the route of administration, dosage form, and 
strength of the drug product are the same as those of the reference 
listed drug except for any differences that have been the subject of an 
approved petition, as follows:
    (A) A statement that the route of administration, dosage form, and 
strength of the proposed drug product are the same as those of the 
reference listed drug.
    (B) A reference to the applicant's annotated proposed labeling and 
to the currently approved labeling for the reference listed drug 
provided under paragraph (a)(8) of this section.
    (ii) If the route of administration, dosage form, or strength of the 
drug product differs from the reference listed drug and the abbreviated 
application is submitted under an approved

[[Page 133]]

petition under Sec. 314.93, such information about the different route 
of administration, dosage form, or strength that FDA may require.
    (7) Bioequivalence. (i) Information that shows that the drug product 
is bioequivalent to the reference listed drug upon which the applicant 
relies; or
    (ii) If the abbreviated new drug application is submitted under a 
petition approved under Sec. 314.93, the results of any bioavailability 
of bioequivalence testing required by the agency, or any other 
information required by the agency to show that the active ingredients 
of the proposed drug product are of the same pharmacological or 
therapeutic class as those in the reference listed drug and that the 
proposed drug product can be expected to have the same therapeutic 
effect as the reference listed drug. If the proposed drug product 
contains a different active ingredient than the reference listed drug, 
FDA will consider the proposed drug product to have the same therapeutic 
effect as the reference listed drug if the applicant provides 
information demonstrating that:
    (A) There is an adequate scientific basis for determining that 
substitution of the specific proposed dose of the different active 
ingredient for the dose of the member of the same pharmacological or 
therapeutic class in the reference listed drug will yield a resulting 
drug product whose safety and effectiveness have not been adversely 
affected.
    (B) The unchanged active ingredients in the proposed drug product 
are bioequivalent to those in the reference listed drug.
    (C) The different active ingredient in the proposed drug product is 
bioequivalent to an approved dosage form containing that ingredient and 
approved for the same indication as the proposed drug product or is 
bioequivalent to a drug product offered for that indication which does 
not meet the definition of ``new drug'' under section 201(p) of the act.
    (iii) For each in vivo bioequivalence study contained in the 
abbreviated new drug application, a description of the analytical and 
statistical methods used in each study and a statement with respect to 
each study that it either was conducted in compliance with the 
institutional review board regulations in part 56 of this chapter, or 
was not subject to the regulations under Sec. 56.104 or Sec. 56.105 of 
this chapter and that each study was conducted in compliance with the 
informed consent regulations in part 50 of this chapter.
    (8) Labeling--(i) Listed drug labeling. A copy of the currently 
approved labeling for the listed drug referred to in the abbreviated new 
drug application, if the abbreviated new drug application relies on a 
reference listed drug.
    (ii) Proposed labeling. Copies of the label and all labeling for the 
drug product (4 copies of draft labeling or 12 copies of final printed 
labeling).
    (iii) A statement that the applicant's proposed labeling is the same 
as the labeling of the reference listed drug except for differences 
annotated and explained under paragraph (a)(8)(iv) of this section.
    (iv) A side-by-side comparison of the applicant's proposed labeling 
with the approved labeling for the reference listed drug with all 
differences annotated and explained. Labeling (including the container 
label and package insert) proposed for the drug product must be the same 
as the labeling approved for the reference listed drug, except for 
changes required because of differences approved under a petition filed 
under Sec. 314.93 or because the drug product and the reference listed 
drug are produced or distributed by different manufacturers. Such 
differences between the applicant's proposed labeling and labeling 
approved for the reference listed drug may include differences in 
expiration date, formulation, bioavailability, or pharmacokinetics, 
labeling revisions made to comply with current FDA labeling guidelines 
or other guidance, or omission of an indication or other aspect of 
labeling protected by patent or accorded exclusivity under section 
505(j)(4)(D) of the act.
    (9) Chemistry, manufacturing, and controls. (i) The information 
required under Sec. 314.50(d)(1), except that Sec. 314.50(d)(1)(ii)(c) 
shall contain the proposed or actual master production record, including 
a description of the

[[Page 134]]

equipment, to be used for the manufacture of a commercial lot of the 
drug product.
    (ii) Inactive ingredients. Unless otherwise stated in paragraphs 
(a)(9)(iii) through (a)(9)(v) of this section, an applicant shall 
identify and characterize the inactive ingredients in the proposed drug 
product and provide information demonstrating that such inactive 
ingredients do not affect the safety of the proposed drug product.
    (iii) Inactive ingredient changes permitted in drug products 
intended for parenteral use. Generally, a drug product intended for 
parenteral use shall contain the same inactive ingredients and in the 
same concentration as the reference listed drug identified by the 
applicant under paragraph (a)(3) of this section. However, an applicant 
may seek approval of a drug product that differs from the reference 
listed drug in preservative, buffer, or antioxidant provided that the 
applicant identifies and characterizes the differences and provides 
information demonstrating that the differences do not affect the safety 
of the proposed drug product.
    (iv) Inactive ingredient changes permitted in drug products intended 
for ophthalmic or otic use. Generally, a drug product intended for 
ophthalmic or otic use shall contain the same inactive ingredients and 
in the same concentration as the reference listed drug identified by the 
applicant under paragraph (a)(3) of this section. However, an applicant 
may seek approval of a drug product that differs from the reference 
listed drug in preservative, buffer, substance to adjust tonicity, or 
thickening agent provided that the applicant identifies and 
characterizes the differences and provides information demonstrating 
that the differences do not affect the safety of the proposed drug 
product, except that, in a product intended for ophthalmic use, an 
applicant may not change a buffer or substance to adjust tonicity for 
the purpose of claiming a therapeutic advantage over or difference from 
the listed drug, e.g., by using a balanced salt solution as a diluent as 
opposed to an isotonic saline solution, or by making a significant 
change in the pH or other change that may raise questions of 
irritability.
    (v) Inactive ingredient changes permitted in drug products intended 
for topical use. Generally, a drug product intended for topical use 
shall contain the same inactive ingredients as the reference listed drug 
identified by the applicant under paragraph (a)(3) of this section. 
However, an applicant may seek approval of a drug product that differs 
from the reference listed drug provided that the applicant identifies 
and characterizes the differences and provides information demonstrating 
that the differences do not affect the safety of the proposed drug 
product.
    (10) Samples. The information required under Sec. 314.50(e)(1) and 
(e)(2)(i). Samples need not be submitted until requested by FDA.
    (11) Other. The information required under Sec. 314.50(g).
    (12) Patent certification--(i) Patents claiming drug, drug product, 
or method of use. (A) Except as provided in paragraph (a)(12)(iv) of 
this section, a certification with respect to each patent issued by the 
United States Patent and Trademark Office that, in the opinion of the 
applicant and to the best of its knowledge, claims the reference listed 
drug or that claims a use of such listed drug for which the applicant is 
seeking approval under section 505(j) of the act and for which 
information is required to be filed under section 505(b) and (c) of the 
act and Sec. 314.53. For each such patent, the applicant shall provide 
the patent number and certify, in its opinion and to the best of its 
knowledge, one of the following circumstances:
    (1) That the patent information has not been submitted to FDA. The 
applicant shall entitle such a certification ``Paragraph I 
Certification'';
    (2) That the patent has expired. The applicant shall entitle such a 
certification ``Paragraph II Certification'';
    (3) The date on which the patent will expire. The applicant shall 
entitle such a certification ``Paragraph III Certification''; or
    (4) That the patent is invalid, unenforceable, or will not be 
infringed by the manufacture, use, or sale of the drug product for which 
the abbreviated application is submitted. The applicant shall entitle 
such a certification

[[Page 135]]

``Paragraph IV Certification''. This certification shall be submitted in 
the following form:

    I, (name of applicant), certify that Patent No. ____________ (is 
invalid, unenforceable, or will not be infringed by the manufacture, 
use, or sale of) (name of proposed drug product) for which this 
application is submitted.


The certification shall be accompanied by a statement that the applicant 
will comply with the requirements under Sec. 314.95(a) with respect to 
providing a notice to each owner of the patent or their representatives 
and to the holder of the approved application for the listed drug, and 
with the requirements under Sec. 314.95(c) with respect to the content 
of the notice.
    (B) If the abbreviated new drug application refers to a listed drug 
that is itself a licensed generic product of a patented drug first 
approved under section 505(b) of the act, the appropriate patent 
certification under paragraph (a)(12)(i) of this section with respect to 
each patent that claims the first-approved patented drug or that claims 
a use for such drug.
    (ii) No relevant patents. If, in the opinion of the applicant and to 
the best of its knowledge, there are no patents described in paragraph 
(a)(12)(i) of this section, a certification in the following form:

    In the opinion and to the best knowledge of (name of applicant), 
there are no patents that claim the listed drug referred to in this 
application or that claim a use of the listed drug.

    (iii) Method of use patent. (A) If patent information is submitted 
under section 505(b) or (c) of the act and Sec. 314.53 for a patent 
claiming a method of using the listed drug, and the labeling for the 
drug product for which the applicant is seeking approval does not 
include any indications that are covered by the use patent, a statement 
explaining that the method of use patent does not claim any of the 
proposed indications.
    (B) If the labeling of the drug product for which the applicant is 
seeking approval includes an indication that, according to the patent 
information submitted under section 505(b) or (c) of the act and 
Sec. 314.53 or in the opinion of the applicant, is claimed by a use 
patent, an applicable certification under paragraph (a)(12)(i) of this 
section.
    (iv) Method of manufacturing patent. An applicant is not required to 
make a certification with respect to any patent that claims only a 
method of manufacturing the listed drug.
    (v) Licensing agreements. If the abbreviated new drug application is 
for a drug or method of using a drug claimed by a patent and the 
applicant has a licensing agreement with the patent owner, a 
certification under paragraph (a)(12)(i)(A)(4) of this section 
(``Paragraph IV Certification'') as to that patent and a statement that 
it has been granted a patent license.
    (vi) Late submission of patent information. If a patent on the 
listed drug is issued and the holder of the approved application for the 
listed drug does not submit the required information on the patent 
within 30 days of issuance of the patent, an applicant who submitted an 
abbreviated new drug application for that drug that contained an 
appropriate patent certification before the submission of the patent 
information is not required to submit an amended certification. An 
applicant whose abbreviated new drug application is submitted after a 
late submission of patent information, or whose pending abbreviated 
application was previously submitted but did not contain an appropriate 
patent certification at the time of the patent submission, shall submit 
a certification under paragraph (a)(12)(i) of this section or a 
statement under paragraph (a)(12)(iii) of this section as to that 
patent.
    (vii) Disputed patent information. If an applicant disputes the 
accuracy or relevance of patent information submitted to FDA, the 
applicant may seek a confirmation of the correctness of the patent 
information in accordance with the procedures under Sec. 314.53(f). 
Unless the patent information is withdrawn or changed, the applicant 
shall submit an appropriate certification for each relevant patent.
    (viii) Amended certifications. A certification submitted under 
paragraphs (a)(12)(i) through (a)(12)(iii) of this section may be 
amended at any time before the effective date of the approval of the 
application. However, an applicant who has submitted a paragraph IV

[[Page 136]]

patent certification may not change it to a paragraph III certification 
if a patent infringement suit has been filed against another paragraph 
IV applicant unless the agency has determined that no applicant is 
entitled to 180-day exclusivity or the patent expires before the lawsuit 
is resolved or expires after the suit is resolved but before the end of 
the 180-day exclusivity period. If an applicant with a pending 
application voluntarily makes a patent certification for an untimely 
filed patent, the applicant may withdraw the patent certification for 
the untimely filed patent. An applicant shall submit an amended 
certification by letter or as an amendment to a pending application or 
by letter to an approved application. Once an amendment or letter is 
submitted, the application will no longer be considered to contain the 
prior certification.
    (A) After finding of infringement. An applicant who has submitted a 
certification under paragraph (a)(12)(i)(A)(4) of this section and is 
sued for patent infringement within 45 days of the receipt of notice 
sent under Sec. 314.95 shall amend the certification if a final judgment 
in the action against the applicant is entered finding the patent to be 
infringed. In the amended certification, the applicant shall certify 
under paragraph (a)(12)(i)(A)(3) of this section that the patent will 
expire on a specific date. Once an amendment or letter for the change 
has been submitted, the application will no longer be considered to be 
one containing a certification under paragraph (a)(12)(i)(A)(4) of this 
section. If a final judgment finds the patent to be invalid and 
infringed, an amended certification is not required.
    (B) After removal of a patent from the list. If a patent is removed 
from the list, any applicant with a pending application (including a 
tentatively approved application with a delayed effective date) who has 
made a certification with respect to such patent shall amend its 
certification. The applicant shall certify under paragraph (a)(12)(ii) 
of this section that no patents described in paragraph (a)(12)(i) of 
this section claim the drug or, if other relevant patents claim the 
drug, shall amend the certification to refer only to those relevant 
patents. In the amendment, the applicant shall state the reason for the 
change in certification (that the patent is or has been removed from the 
list). A patent that is the subject of a lawsuit under Sec. 314.107(c) 
shall not be removed from the list until FDA determines either that no 
delay in effective dates of approval is required under that section as a 
result of the lawsuit, that the patent has expired, or that any such 
period of delay in effective dates of approval is ended. An applicant 
shall submit an amended certification. Once an amendment or letter for 
the change has been submitted, the application will no longer be 
considered to be one containing a certification under paragraph 
(a)(12)(i)(A)(4) of this section.
    (C) Other amendments. (1) Except as provided in paragraphs 
(a)(12)(vi) and (a)(12)(viii)(C)(2) of this section, an applicant shall 
amend a submitted certification if, at any time before the effective 
date of the approval of the application, the applicant learns that the 
submitted certification is no longer accurate.
    (2) An applicant is not required to amend a submitted certification 
when information on a patent on the listed drug is submitted after the 
effective date of approval of the abbreviated application.
    (13) Financial certification or disclosure statement. An abbreviated 
application shall contain a financial certification or disclosure 
statement as required by part 54 of this chapter.
    (b) Drug products subject to the Drug Efficacy Study Implementation 
(DESI) review. If the abbreviated new drug application is for a 
duplicate of a drug product that is subject to FDA's DESI review (a 
review of drug products approved as safe between 1938 and 1962) or other 
DESI-like review and the drug product evaluated in the review is a 
listed drug, the applicant shall comply with the provisions of paragraph 
(a) of this section.
    (c) Abbreviated antibiotic application. For applications submitted 
under section 507 of the act, the applicant shall submit a complete 
archival copy of the abbreviated application that contains the 
information described under Sec. 314.50 (a)(1), (a)(3), (a)(4), and 
(a)(5), (b), (d)(1) and (d)(3), (e), and (g). The applicant

[[Page 137]]

shall state whether the submission is an abbreviated application under 
this section or a supplement to an abbreviated application under 
Sec. 314.97.
    (d) Format of an abbreviated application. (1) The applicant shall 
submit a complete archival copy of the abbreviated application as 
required under paragraphs (a) and (c) of this section. FDA will maintain 
the archival copy during the review of the application to permit 
individual reviewers to refer to information that is not contained in 
their particular technical sections of the application, to give other 
agency personnel access to the application for official business, and to 
maintain in one place a complete copy of the application. An applicant 
may submit all or portions of the archival copy of the abbreviated 
application in any form (e.g., microfiche, optical disc, and magnetic 
tape) that the applicant and FDA agree is acceptable.
    (2) For abbreviated new drug applications, the applicant shall 
submit a review copy of the abbreviated application that contains two 
separate sections. One section shall contain the information described 
under paragraphs (a)(2) through (a)(6), (a)(8), and (a)(9) of this 
section 505(j)(2)(A)(vii) of the act and one copy of the analytical 
methods and descriptive information needed by FDA's laboratories to 
perform tests on samples of the proposed drug product and to validate 
the applicant's analytical methods. The other section shall contain the 
information described under paragraphs (a)(3), (a)(7), and (a)(8) of 
this section. Each of the sections in the review copy is required to 
contain a copy of the application form described under Sec. 314.50(a).
    (3) For abbreviated antibiotic applications, the applicant shall 
submit a review copy that contains the technical sections described in 
Sec. 314.50 (d)(1) and (d)(3). Each of the technical sections in the 
review copy is required to be separate with a copy of the application 
form required under Sec. 314.50(a).
    (4) The applicant may obtain from FDA sufficient folders to bind the 
archival, the review, and the field copies of the abbreviated 
application.
    (5) The applicant shall submit a field copy of the abbreviated 
application that contains the technical section described in paragraph 
(a)(9) of this section, a copy of the application form required under 
paragraph (a)(1) of this section, and a certification that the field 
copy is a true copy of the technical section described in paragraph 
(a)(9) of this section contained in the archival and review copies of 
the abbreviated application.
[57 FR 17983, Apr. 28, 1992; 57 FR 29353, July 1, 1992, as amended at 58 
FR 47352, Sept. 8, 1993; 59 FR 50364, Oct. 3, 1994; 63 FR 5252, Feb. 2, 
1998]

    Effective Date Note: At 63 FR 5252, Feb. 2, 1998, Sec. 314.94 was 
amended by adding new paragraph (a)(13), effective Feb. 2, 1999.



Sec. 314.95  Notice of certification of invalidity or noninfringement of a patent.

    (a) Notice of certification. For each patent that claims the listed 
drug or that claims a use for such listed drug for which the applicant 
is seeking approval and that the applicant certifies under 
Sec. 314.94(a)(12) is invalid, unenforceable, or will not be infringed, 
the applicant shall send notice of such certification by registered or 
certified mail, return receipt requested to each of the following 
persons:
    (1) Each owner of the patent which is the subject of the 
certification or the representative designated by the owner to receive 
the notice. The name and address of the patent owner or its 
representative may be obtained from the United States Patent and 
Trademark Office; and
    (2) The holder of the approved application under section 505(b) of 
the act for the listed drug that is claimed by the patent and for which 
the applicant is seeking approval, or, if the application holder does 
not reside or maintain a place of business within the United States, the 
application holder's attorney, agent, or other authorized official. The 
name and address of the application holder or its attorney, agent, or 
authorized official may be obtained from the Division of Drug 
Information Resources (HFD-80), Center for Drug Evaluation and Research, 
Food and Drug Administration, 5600 Fishers Lane, Rockville, MD 20857.
    (3) This paragraph does not apply to a use patent that claims no 
uses for

[[Page 138]]

which the applicant is seeking approval.
    (b) Sending the notice. The applicant shall send the notice required 
by paragraph (a) of this section when it receives from FDA an 
acknowledgment letter stating that its abbreviated new drug application 
is sufficiently complete to permit a substantive review. At the same 
time, the applicant shall amend its abbreviated new drug application to 
include a statement certifying that the notice has been provided to each 
person identified under paragraph (a) of this section and that the 
notice met the content requirements under paragraph (c) of this section.
    (c) Contents of a notice. In the notice, the applicant shall cite 
section 505(j)(2)(B)(ii) of the act and shall include, but not be 
limited to, the following information:
    (1) A statement that FDA has received an abbreviated new drug 
application submitted by the applicant containing any required 
bioavailability or bioequivalence data or information.
    (2) The abbreviated application number.
    (3) The established name, if any, as defined in section 502(e)(3) of 
the act, of the proposed drug product.
    (4) The active ingredient, strength, and dosage form of the proposed 
drug product.
    (5) The patent number and expiration date, as submitted to the 
agency or as known to the applicant, of each patent alleged to be 
invalid, unenforceable, or not infringed.
    (6) A detailed statement of the factual and legal basis of the 
applicant's opinion that the patent is not valid, unenforceable, or will 
not be infringed. The applicant shall include in the detailed statement:
    (i) For each claim of a patent alleged not to be infringed, a full 
and detailed explanation of why the claim is not infringed.
    (ii) For each claim of a patent alleged to be invalid or 
unenforceable, a full and detailed explanation of the grounds supporting 
the allegation.
    (7) If the applicant does not reside or have a place of business in 
the United States, the name and address of an agent in the United States 
authorized to accept service of process for the applicant.
    (d) Amendment to an abbreviated application. If an abbreviated 
application is amended to include the certification described in 
Sec. 314.94(a)(12)(i)(A)(4), the applicant shall send the notice 
required by paragraph (a) of this section at the same time that the 
amendment to the abbreviated application is submitted to FDA.
    (e) Documentation of receipt of notice. The applicant shall amend 
its abbreviated application to document receipt of the notice required 
under paragraph (a) of this section by each person provided the notice. 
The applicant shall include a copy of the return receipt or other 
similar evidence of the date the notification was received. FDA will 
accept as adequate documentation of the date of receipt a return receipt 
or a letter acknowledging receipt by the person provided the notice. An 
applicant may rely on another form of documentation only if FDA has 
agreed to such documentation in advance. A copy of the notice itself 
need not be submitted to the agency.
    (f) Approval. If the requirements of this section are met, FDA will 
presume the notice to be complete and sufficient, and it will count the 
day following the date of receipt of the notice by the patent owner or 
its representative and by the approved application holder as the first 
day of the 45-day period provided for in section 505(j)(4)(B)(iii) of 
the act. FDA may, if the applicant provides a written statement to FDA 
that a later date should be used, count from such later date.
[59 FR 50366, Oct. 3, 1994]



Sec. 314.96  Amendments to an unapproved abbreviated application.

    (a) Abbreviated new drug application. (1) An applicant may amend an 
abbreviated new drug application that is submitted under Sec. 314.94, 
but not yet approved, to revise existing information or provide 
additional information.
    (2) Submission of an amendment containing significant data or 
information constitutes an agreement between FDA and the applicant to 
extend the review period only for the time necessary to review the 
significant data or information and for no more than 180 days.

[[Page 139]]

    (3) Submission of an amendment containing significant data or 
information to resolve deficiencies in the application as set forth in a 
not approvable letter issued under Sec. 314.120 constitutes an agreement 
between FDA and the applicant under section 505(j)(4)(A) of the act to 
extend the date by which the agency is required to reach a decision on 
the abbreviated new drug application only for the time necessary to 
review the significant data or information and for no more than 180 
days.
    (b) The applicant shall submit a field copy of each amendment to 
Sec. 314.94(a)(9). The applicant, other than a foreign applicant, shall 
include in its submission of each such amendment to FDA a statement 
certifying that a field copy of the amendment has been sent to the 
applicant's home FDA district office.
    (c) Abbreviated antibiotic application. The applicant shall comply 
with the provisions of Sec. 314.60.
[57 FR 17983, Apr. 28, 1992, as amended at 58 FR 47352, Sept. 8, 1993]



Sec. 314.97  Supplements and other changes to an approved abbreviated application.

    The applicant shall comply with the requirements of Secs. 314.70 and 
314.71 regarding the submission of supplemental applications and other 
changes to an approved abbreviated application.



Sec. 314.98  Postmarketing reports.

    (a) Except as provided in paragraph (b) of this section, each 
applicant having an approved abbreviated antibiotic application under 
Sec. 314.94 or approved abbreviated new drug application under 
Sec. 314.94 that is effective shall comply with the requirements of 
Sec. 314.80 regarding the reporting and recordkeeping of adverse drug 
experiences.
    (b) Each applicant shall submit one copy of each report required 
under Sec. 314.80 to the Division of Epidemiology and Surveillance (HFD-
730), Center for Drug Evaluation and Research, Food and Drug 
Administration, 5600 Fishers Lane, Rockville, MD 20857.
    (c) Each applicant shall make the reports required under Sec. 314.81 
and sections 505(k) and 507(g) of the act for each of its approved 
abbreviated applications.



Sec. 314.99  Other responsibilities of an applicant of an abbreviated application.

    (a) An applicant shall comply with the requirements of Sec. 314.65 
regarding withdrawal by the applicant of an unapproved abbreviated 
application and Sec. 314.72 regarding a change in ownership of an 
abbreviated application.
    (b) An applicant may ask FDA to waive under this section any 
requirement that applies to the applicant under Secs. 314.92 through 
314.99. The applicant shall comply with the requirements for a waiver 
under Sec. 314.90.



   Subpart D--FDA Action on Applications and Abbreviated Applications

    Source:  50 FR 7493, Feb. 22, 1985, unless otherwise noted. 
Redesignated at 57 FR 17983, Apr. 28, 1992.



Sec. 314.100  Timeframes for reviewing applications and abbreviated applications.

    (a) Within 180 days of receipt of an application for a new drug 
under section 505(b) of the act, or of an abbreviated application for a 
new drug under section 505(j) of the act, or of an application or 
abbreviated application for an antibiotic drug under section 507 of the 
act, FDA will review it and send the applicant either an approval letter 
under Sec. 314.105, or an approvable letter under Sec. 314.110, or a not 
approvable letter under Sec. 314.120. This 180-day period is called the 
``review clock.''
    (b) During the review period, an applicant may withdraw an 
application under Sec. 314.65 or an abbreviated application under 
Sec. 314.99 and later resubmit it. FDA will treat the resubmission as a 
new application or abbreviated application.
    (c) The review clock may be extended by mutual agreement between FDA 
and an applicant or as provided in Secs. 314.60 and 314.96, as the 
result of a major amendment.
[57 FR 17987, Apr. 28, 1992]

[[Page 140]]



Sec. 314.101  Filing an application and an abbreviated antibiotic application and receiving an abbreviated new drug application.

    (a)(1) Within 60 days after FDA receives an application or 
abbreviated antibiotic application, the agency will determine whether 
the application or abbreviated antibiotic application may be filed. The 
filing of an application or abbreviated antibiotic application means 
that FDA has made a threshold determination that the application or 
abbreviated antibiotic application is sufficiently complete to permit a 
substantive review.
    (2) If FDA finds that none of the reasons in paragraphs (d) and (e) 
of this section for refusing to file the application or abbreviated 
antibiotic apply, the agency will file the application or abbreviated 
antibiotic application and notify the applicant in writing. The date of 
filing will be the date 60 days after the date FDA received the 
application or abbreviated antibiotic application. The date of filing 
begins the 180-day period described in section 505(c) of the act. This 
180-day period is called the ``filing clock.''
    (3) If FDA refuses to file the application or abbreviated antibiotic 
application, the agency will notify the applicant in writing and state 
the reason under paragraph (d) or (e) of this section for the refusal. 
If FDA refuses to file the application or abbreviated antibiotic 
application under paragraph (d) of this section, the applicant may 
request in writing within 30 days of the date of the agency's 
notification an informal conference with the agency about whether the 
agency should file the application or abbreviated antibiotic 
application. If, following the informal conference, the applicant 
requests that FDA file the application or abbreviated antibiotic 
application (with or without amendments to correct the deficiencies), 
the agency will file the application or abbreviated antibiotic 
application over protest under paragraph (a)(2) of this section, notify 
the applicant in writing, and review it as filed. If the application or 
abbreviated antibiotic application is filed over protest, the date of 
filing will be the date 60 days after the date the applicant requested 
the informal conference. The applicant need not resubmit a copy of an 
application or abbreviated antibiotic application that is filed over 
protest. If FDA refuses to file the application or abbreviated 
antibiotic application under paragraph (e) of this section, the 
applicant may amend the application or abbreviated antibiotic 
application and resubmit it, and the agency will make a determination 
under this section whether it may be filed.
    (b)(1) An abbreviated new drug application will be reviewed after it 
is submitted to determine whether the abbreviated application may be 
received. Receipt of an abbreviated new drug application means that FDA 
has made a threshold determination that the abbreviated application is 
sufficiently complete to permit a substantive review.
    (2) If FDA finds that none of the reasons in paragraphs (d) and (e) 
of this section for considering the abbreviated new drug application not 
to have been received applies, the agency will receive the abbreviated 
new drug application and notify the applicant in writing.
    (3) If FDA considers the abbreviated new drug application not to 
have been received under paragraph (d) or (e) of this section, FDA will 
notify the applicant, ordinarily by telephone. The applicant may then:
    (i) Withdraw the abbreviated new drug application under Sec. 314.99; 
or
    (ii) Amend the abbreviated new drug application to correct the 
deficiencies; or
    (iii) Take no action, in which case FDA will refuse to receive the 
abbreviated new drug application.
    (c) [Reserved]
    (d) FDA may refuse to file an application or abbreviated antibiotic 
application or may not consider an abbreviated new drug application to 
be received if any of the following applies:
    (1) The application or abbreviated application does not contain a 
completed application form.
    (2) The application or abbreviated application is not submitted in 
the form required under Sec. 314.50 or Sec. 314.94.
    (3) The application or abbreviated application is incomplete because 
it does

[[Page 141]]

not on its face contain information required under section 505(b), 
section 505(j), or section 507 of the act and Sec. 314.50 or 
Sec. 314.94.
    (4) The applicant fails to submit a complete environmental 
assessment, which addresses each of the items specified in the 
applicable format under Sec. 25.40 of this chapter or fails to provide 
sufficient information to establish that the requested action is subject 
to categorical exclusion under Sec. 25.30 or Sec. 25.31 of this chapter.
    (5) The application or abbreviated application does not contain an 
accurate and complete English translation of each part of the 
application that is not in English.
    (6) The application does not contain a statement for each 
nonclinical laboratory study that it was conducted in compliance with 
the requirements set forth in part 58 of this chapter, or, for each 
study not conducted in compliance with part 58 of this chapter, a brief 
statement of the reason for the noncompliance.
    (7) The application does not contain a statement for each clinical 
study that it was conducted in compliance with the institutional review 
board regulations in part 56 of this chapter, or was not subject to 
those regulations, and that it was conducted in compliance with the 
informed consent regulations in part 50 of this chapter, or, if the 
study was subject to but was not conducted in compliance with those 
regulations, the application does not contain a brief statement of the 
reason for the noncompliance.
    (8) The drug product that is the subject of the submission is 
already covered by an approved application or abbreviated application 
and the applicant of the submission:
    (i) Has an approved application or abbreviated application for the 
same drug product; or
    (ii) Is merely a distributor and/or repackager of the already 
approved drug product.
    (9) The application is submitted as a 505(b)(2) application for a 
drug that is a duplicate of a listed drug and is eligible for approval 
under section 505(j) of the act.
    (e) The agency will refuse to file an application or abbreviated 
antibiotic application or will consider an abbreviated new drug 
application not to have been received if any of the following applies:
    (1) The drug product is subject to licensing by FDA under the Public 
Health Service Act (42 U.S.C. 201 et seq.) and subchapter F of this 
chapter.
    (2) In the case of a 505(b)(2) application or an abbreviated new 
drug application, the drug product contains the same active moiety as a 
drug that:
    (i) Was approved after September 24, 1984, in an application under 
section 505(b) of the act, and
    (ii) Is entitled to a 5-year period of exclusivity under section 
505(c)(3)(D)(ii) and (j)(4)(D)(ii) of the act and Sec. 314.108(b)(2), 
unless the 5-year exclusivity period has elapsed or unless 4 years of 
the 5-year period have elapsed and the application or abbreviated 
application contains a certification of patent invalidity or 
noninfringement described in Sec. 314.50(i)(1)(i)(A)(4) or 
Sec. 314.94(a)(12)(i)(A)(4).
    (f)(1) Within 180 days after the date of filing, plus the period of 
time the review period was extended (if any), FDA will either:
    (i) Approve the application or abbreviated antibiotic application; 
or
    (ii) Issue a notice of opportunity for hearing if the applicant 
asked FDA to provide it an opportunity for a hearing on an application 
or abbreviated antibiotic application in response to an approvable 
letter or a not approvable letter.
    (2) Within 180 days after the date of receipt, plus the period of 
time the review clock was extended (if any), FDA will either approve or 
disapprove the abbreviated new drug application. If FDA disapproves the 
abbreviated new drug application, FDA will issue a notice of opportunity 
for hearing if the applicant asked FDA to provide it an opportunity for 
a hearing on an abbreviated new drug application in response to a not 
approvable letter.
    (3) This paragraph does not apply to applications or abbreviated 
applications that have been withdrawn from FDA review by the applicant.
[57 FR 17987, Apr. 28, 1992; 57 FR 29353, July 1, 1992, as amended at 59 
FR 50366, Oct. 3, 1994; 62 FR 40599, July 29, 1997]

[[Page 142]]



Sec. 314.102  Communications between FDA and applicants.

    (a) General principles. During the course of reviewing an 
application or an abbreviated application, FDA shall communicate with 
applicants about scientific, medical, and procedural issues that arise 
during the review process. Such communication may take the form of 
telephone conversations, letters, or meetings, whichever is most 
appropriate to discuss the particular issue at hand. Communications 
shall be appropriately documented in the application in accordance with 
Sec. 10.65 of this chapter. Further details on the procedures for 
communication between FDA and applicants are contained in a staff manual 
guide that is publicly available.
    (b) Notification of easily correctable deficiencies. FDA reviewers 
shall make every reasonable effort to communicate promptly to applicants 
easily correctable deficiencies found in an application or an 
abbreviated application when those deficiencies are discovered, 
particularly deficiencies concerning chemistry, manufacturing, and 
controls issues. The agency will also inform applicants promptly of its 
need for more data or information or for technical changes in the 
application or the abbreviated application needed to facilitate the 
agency's review. This early communication is intended to permit 
applicants to correct such readily identified deficiencies relatively 
early in the review process and to submit an amendment before the review 
period has elapsed. Such early communication would not ordinarily apply 
to major scientific issues, which require consideration of the entire 
pending application or abbreviated application by agency managers as 
well as reviewing staff. Instead, major scientific issues will 
ordinarily be addressed in an action letter.
    (c) Ninety-day conference. Approximately 90 days after the agency 
receives the application, FDA will provide applicants with an 
opportunity to meet with agency reviewing officials. The purpose of the 
meeting will be to inform applicants of the general progress and status 
of their applications, and to advise applicants of deficiencies that 
have been identified by that time and that have not already been 
communicated. This meeting will be available on applications for all new 
chemical entities and major new indications of marketed drugs. Such 
meetings will be held at the applicant's option, and may be held by 
telephone if mutually agreed upon. Such meetings would not ordinarily be 
held on abbreviated applications because they are not submitted for new 
chemical entities or new indications.
    (d) End of review conference. At the conclusion of FDA's review of 
an application or an abbreviated application as designated by the 
issuance of an approvable or not approvable letter, FDA will provide 
applicants with an opportunity to meet with agency reviewing officials. 
The purpose of the meeting will be to discuss what further steps need to 
be taken by the applicant before the application or abbreviated 
application can be approved. This meeting will be available on all 
applications or abbreviated applications, with priority given to 
applications for new chemical entities and major new indications for 
marketed drugs and for the first duplicates for such drugs. Requests for 
such meetings shall be directed to the director of the division 
responsible for reviewing the application or abbreviated application.
    (e) Other meetings. Other meetings between FDA and applicants may be 
held, with advance notice, to discuss scientific, medical, and other 
issues that arise during the review process. Requests for meetings shall 
be directed to the director of the division responsible for reviewing 
the application or abbreviated application. FDA will make every attempt 
to grant requests for meetings that involve important issues and that 
can be scheduled at mutually convenient times. However, ``drop-in'' 
visits (i.e., an unannounced and unscheduled visit by a company 
representative) are discouraged except for urgent matters, such as to 
discuss an important new safety issue.
[57 FR 17988, Apr. 28, 1992; 57 FR 29353, July 1, 1992]



Sec. 314.103  Dispute resolution.

    (a) General. FDA is committed to resolving differences between 
applicants

[[Page 143]]

and FDA reviewing divisions with respect to technical requirements for 
applications or abbreviated applications as quickly and amicably as 
possible through the cooperative exchange of information and views.
    (b) Administrative and procedural issues. When administrative or 
procedural disputes arise, the applicant should first attempt to resolve 
the matter with the division responsible for reviewing the application 
or abbreviated application, beginning with the consumer safety officer 
assigned to the application or abbreviated application. If resolution is 
not achieved, the applicant may raise the matter with the person 
designated as ombudsman, whose function shall be to investigate what has 
happened and to facilitate a timely and equitable resolution. 
Appropriate issues to raise with the ombudsman include resolving 
difficulties in scheduling meetings, obtaining timely replies to 
inquiries, and obtaining timely completion of pending reviews. Further 
details on this procedure are contained in a staff manual guide that is 
publicly available under FDA's public information regulations in part 
20.
    (c) Scientific and medical disputes. (1) Because major scientific 
issues are ordinarily communicated to applicants in an approvable or not 
approvable letter pursuant to Sec. 314.110 or Sec. 314.120, 
respectively, the ``end-of-review conference'' described in 
Sec. 314.102(d) will provide a timely forum for discussing and 
resolving, if possible, scientific and medical issues on which the 
applicant disagrees with the agency. In addition, the ``ninety-day 
conference'' described in Sec. 314.102(c) will provide a timely forum 
for discussing and resolving, if possible, issues identified by that 
date.
    (2) When scientific or medical disputes arise at other times during 
the review process, applicants should discuss the matter directly with 
the responsible reviewing officials. If necessary, applicants may 
request a meeting with the appropriate reviewing officials and 
management representatives in order to seek a resolution. Ordinarily, 
such meetings would be held first with the Division Director, then with 
the Office Director, and finally with the Center Director if the matter 
is still unresolved. Requests for such meetings shall be directed to the 
director of the division responsible for reviewing the application or 
abrreviated application. FDA will make every attempt to grant requests 
for meetings that involve important issues and that can be scheduled at 
mutually convenient times.
    (3) In requesting a meeting designed to resolve a scientific or 
medical dispute, applicants may suggest that FDA seek the advice of 
outside experts, in which case FDA may, in its discretion, invite to the 
meeting one or more of its advisory committee members or other 
consultants, as designated by the agency. Applicants may also bring 
their own consultants. For major scientific and medical policy issues 
not resolved by informal meetings, FDA may refer the matter to one of 
its standing advisory committees for its consideration and 
recommendations.
[50 FR 7493, Feb. 22, 1985; 50 FR 14212, Apr. 11, 1985, as amended at 57 
FR 17989, Apr. 28, 1992]



Sec. 314.104  Drugs with potential for abuse.

    The Food and Drug Administration will inform the Drug Enforcement 
Administration under section 201(f) of the Controlled Substances Act (21 
U.S.C. 801) when an application or abbreviated application is submitted 
for a drug that appears to have an abuse potential.
[57 FR 17989, Apr. 28, 1992]



Sec. 314.105  Approval of an application and an abbreviated application.

    (a) The Food and Drug Administration will approve an application or 
an abbreviated antibiotic application and sent the applicant an approval 
letter if none of the reasons in Sec. 314.125 for refusing to approve 
the application or abbreviated antibiotic application applies. An 
approval becomes effective on the date of the issuance of the approval 
letter, except with regard to an approval under section 505(b)(2) of the 
act with a delayed effective date. An approval with a delayed effective 
date is tentative and does not become final until the effective date. 
When FDA sends an applicant an approval letter for an antibiotic, it 
will promulgate a regulation under Sec. 314.300 providing for

[[Page 144]]

certification of the drug, if necessary. A new drug product or 
antibiotic approved under this paragraph may not be marketed until an 
approval is effective. Marketing of an antibiotic need not await the 
promulgation of a regulation under Sec. 314.300.
    (b) FDA will approve an application or abbreviated antibiotic 
application and issue the applicant an approval letter (rather than an 
approvable letter under Sec. 314.110) on the basis of draft labeling if 
the only deficiencies in the application or abbreviated antibiotic 
application concern editorial or similar minor deficiencies in the draft 
labeling. Such approval will be conditioned upon the applicant 
incorporating the specified labeling changes exactly as directed, and 
upon the applicant submitting to FDA a copy of the final printed 
labeling prior to marketing.
    (c) FDA will approve an application after it determines that the 
drug meets the statutory standards for safety and effectiveness, 
manufacturing and controls, and labeling, and an abbreviated application 
after it determines that the drug meets the statutory standards for 
manufacturing and controls, labeling, and, where applicable, 
bioequivalence. While the statutory standards apply to all drugs, the 
many kinds of drugs that are subject to the statutory standards and the 
wide range of uses for those drugs demand flexibility in applying the 
standards. Thus FDA is required to exercise its scientific judgment to 
determine the kind and quantity of data and information an applicant is 
required to provide for a particular drug to meet the statutory 
standards. FDA makes its views on drug products and classes of drugs 
available through guidelines, recommendations, and other statements of 
policy.
    (d) FDA will approve an abbreviated new drug application and send 
the applicant an approval letter if none of the reasons in Sec. 314.127 
for refusing to approve the abbreviated new drug application applies. 
The approval becomes effective on the date of the issuance of the 
agency's approval letter unless the approval letter provides for a 
delayed effective date. An approval with a delayed effective date is 
tentative and does not become final until the effective date. A new drug 
product approved under this paragraph may not be introduced or delivered 
for introduction into interstate commerce until approval of the 
abbreviated new drug application is effective. Ordinarily, the effective 
date of approval will be stated in the approval letter.
[57 FR 17989, Apr. 28, 1992]



Sec. 314.106  Foreign data.

    (a) General. The acceptance of foreign data in an application 
generally is governed by Sec. 312.120 of this chapter.
    (b) As sole basis for marketing approval. An application based 
solely on foreign clinical data meeting U.S. criteria for marketing 
approval may be approved if: (1) The foreign data are applicable to the 
U.S. population and U.S. medical practice; (2) the studies have been 
performed by clinical investigators of recognized competence; and (3) 
the data may be considered valid without the need for an on-site 
inspection by FDA or, if FDA considers such an inspection to be 
necessary, FDA is able to validate the data through an on-site 
inspection or other appropriate means. Failure of an application to meet 
any of these criteria will result in the application not being 
approvable based on the foreign data alone. FDA will apply this policy 
in a flexible manner according to the nature of the drug and the data 
being considered.
    (c) Consultation between FDA and applicants. Applicants are 
encouraged to meet with agency officials in a ``presubmission'' meeting 
when approval based solely on foreign data will be sought.
[50 FR 7493, Feb. 22, 1985, as amended at 55 FR 11580, Mar. 29, 1990]



Sec. 314.107  Effective date of approval of a 505(b)(2) application or abbreviated new drug application under section 505(j) of the act.

    (a) General. A drug product may be introduced or delivered for 
introduction into interstate commerce when approval of the application 
or abbreviated application for the drug product becomes effective. 
Except as provided in this section, approval of an application or 
abbreviated application for a drug product becomes effective on the date 
FDA issues an approval letter

[[Page 145]]

under Sec. 314.105 for the application or abbreviated application.
    (b) Effect of patent on the listed drug. If approval of an 
abbreviated new drug application submitted under section 505(j) of the 
act or of a 505(b)(2) application is granted, that approval will become 
effective in accordance with the following:
    (1) Date of approval letter. Except as provided in paragraphs 
(b)(3), (b)(4), and (c) of this section, approval will become effective 
on the date FDA issues an approval letter under Sec. 314.105 if the 
applicant certifies under Sec. 314.50(i) or Sec. 314.94(a)(12) that:
    (i) There are no relevant patents; or
    (ii) The applicant is aware of a relevant patent but the patent 
information required under section 505 (b) or (c) of the act has not 
been submitted to FDA; or
    (iii) The relevant patent has expired; or
    (iv) The relevant patent is invalid, unenforceable, or will not be 
infringed.
    (2) Patent expiration. If the applicant certifies under 
Sec. 314.50(i) or Sec. 314.94(a)(12) that the relevant patent will 
expire on a specified date, approval will become effective on the 
specified date.
    (3) Disposition of patent litigation. (i)(A) Except as provided in 
paragraphs (b)(3)(ii), (b)(3)(iii), and (b)(3)(iv) of this section, if 
the applicant certifies under Sec. 314.50(i) or Sec. 314.94(a)(12) that 
the relevant patent is invalid, unenforceable, or will not be infringed, 
and the patent owner or its representative or the exclusive patent 
licensee brings suit for patent infringement within 45 days of receipt 
by the patent owner of the notice of certification from the applicant 
under Sec. 314.52 or Sec. 314.95, approval may be made effective 30 
months after the date of the receipt of the notice of certification by 
the patent owner or by the exclusive licensee (or their representatives) 
unless the court has extended or reduced the period because of a failure 
of either the plaintiff or defendant to cooperate reasonably in 
expediting the action; or
    (B) If the patented drug product qualifies for 5 years of exclusive 
marketing under Sec. 314.108(b)(2) and the patent owner or its 
representative or the exclusive patent licensee brings suit for patent 
infringement during the 1-year period beginning 4 years after the date 
the patented drug was approved and within 45 days of receipt by the 
patent owner of the notice of certification, the approval may be made 
effective at the expiration of the 7\1/2\ years from the date of 
approval of the application for the patented drug product.
    (ii) If before the expiration of the 30-month period, or 7\1/2\ 
years where applicable, the court issues a final order that the patent 
is invalid, unenforceable, or not infringed, approval may be made 
effective on the date the court enters judgment;
    (iii) If before the expiration of the 30-month period, or 7\1/2\ 
years where applicable, the court issues a final order or judgment that 
the patent has been infringed, approval may be made effective on the 
date the court determines that the patent will expire or otherwise 
orders; or
    (iv) If before the expiration of the 30-month period, or 7\1/2\ 
years where applicable, the court grants a preliminary injunction 
prohibiting the applicant from engaging in the commercial manufacture or 
sale of the drug product until the court decides the issues of patent 
validity and infringement, and if the court later decides that the 
patent is invalid, unenforceable, or not infringed, approval may be made 
effective on the date the court enters a final order or judgment that 
the patent is invalid, unenforceable, or not infringed.
    (v) In order for an approval to be made effective under paragraph 
(b)(3) of this section, the applicant must receive an approval letter 
from the agency indicating that the application has received final 
approval. Tentative approval of an application does not constitute 
``approval'' of an application and cannot, absent a final approval 
letter from the agency, result in an effective approval under paragraph 
(b)(3) of this section.
    (4) Multiple certifications. If the applicant has submitted 
certifications under Sec. 314.50(i) or Sec. 314.94(a)(12) for more than 
one patent, the date of approval will be calculated for each 
certification, and the approval will become effective on the last 
applicable date.

[[Page 146]]

    (c) Subsequent abbreviated new drug application submission. (1) If 
an abbreviated new drug application contains a certification that a 
relevant patent is invalid, unenforceable, or will not be infringed and 
the application is for a generic copy of the same listed drug for which 
one or more substantially complete abbreviated new drug applications 
were previously submitted containing a certification that the same 
patent was invalid, unenforceable, or would not be infringed and the 
applicant submitting the first application has successfully defended 
against a suit for patent infringement brought within 45 days of the 
patent owner's receipt of notice submitted under Sec. 314.95, approval 
of the subsequent abbreviated new drug application will be made 
effective no sooner than 180 days from whichever of the following dates 
is earlier:
    (i) The date the applicant submitting the first application first 
commences commercial marketing of its drug product; or
    (ii) The date of a decision of the court holding the relevant patent 
invalid, unenforceable, or not infringed.
    (2) For purposes of paragraph (c)(1) of this section, the 
``applicant submitting the first application'' is the applicant that 
submits an application that is both substantially complete and contains 
a certification that the patent was invalid, unenforceable, or not 
infringed prior to the submission of any other application for the same 
listed drug that is both substantially complete and contains the same 
certification. A ``substantially complete'' application must contain the 
results of any required bioequivalence studies, or, if applicable, a 
request for a waiver of such studies.
    (3) For purposes of paragraph (c)(1) of this section, if FDA 
concludes that the applicant submitting the first application is not 
actively pursuing approval of its abbreviated application, FDA will make 
the approval of subsequent abbreviated applications immediately 
effective if they are otherwise eligible for an immediately effective 
approval.
    (4) For purposes of paragraph (c)(1)(i) of this section, the 
applicant submitting the first application shall, if sued for patent 
infringement, notify FDA of the date that it commences commercial 
marketing of its drug product. Commercial marketing commences with the 
first date of introduction or delivery for introduction into interstate 
commerce outside the control of the manufacturer of a drug product, 
except for investigational use under part 312 of this chapter, but does 
not include transfer of the drug product for reasons other than sale 
within the control of the manufacturer or application holder. If an 
applicant does not promptly notify FDA of such date, the effective date 
of approval shall be deemed to be the date of the commencement of first 
commercial marketing.
    (d) Delay due to exclusivity. The agency will also delay the 
effective date of the approval of an abbreviated new drug application 
under section 505(j) of the act or a 505(b)(2) application if delay is 
required by the exclusivity provisions in Sec. 314.108. When the 
effective date of an application is delayed under both this section and 
Sec. 314.108, the effective date will be the later of the 2 days 
specified under this section and Sec. 314.108.
    (e) Court actions. (1) References to actions of ``the court'' in 
paragraphs (b) and (c) of this section are to the court that enters 
final judgment from which no appeal can be or has been taken.
    (2) For purposes of establishing the effective date of approval 
based on a court judgment, the following dates shall be deemed to be the 
date of the final decision of the court on the patent issues:
    (i) If the district court enters a decision that the patent is 
invalid, unenforceable, or not infringed, and the decision is not 
appealed, the date on which the right to appeal lapses.
    (ii) If the district court enters a decision that the patent is 
invalid, unenforceable, or not infringed, and the decision is appealed, 
the date of the first decision or order by a higher court holding or 
affirming the decision of the district court that the patent is invalid, 
unenforceable, or not infringed.
    (iii) If the district court enters a decision that the patent is 
infringed, and the decision is appealed, the date on

[[Page 147]]

which the district court enters a judgment that the patent is invalid, 
unenforceable, or not infringed pursuant to a mandate issued by a court 
of appeals.
    (iv) The applicant shall submit a copy of the entry of the order or 
judgment to the Office of Generic Drugs (HFD-600), or to the appropriate 
division in the Office of Drug Evaluation I (HFD-100) or Office of Drug 
Evaluation II (HFD-500), whichever is applicable, within 10 working days 
of a final judgment.
    (f) Computation of 45-day time clock. (1) The 45-day clock described 
in paragraph (b)(3) of this section begins on the day after the date of 
receipt of the applicant's notice of certification by the patent owner 
or its representative, and by the approved application holder. When the 
45th day falls on Saturday, Sunday, or a Federal holiday, the 45th day 
will be the next day that is not a Saturday, Sunday, or a Federal 
holiday.
    (2) The abbreviated new drug applicant or the 505(b)(2) applicant 
shall notify FDA immediately of the filing of any legal action filed 
within 45 days of receipt of the notice of certification. If the 
applicant submitting the abbreviated new drug application or the 
505(b)(2) application or patent owner or its representative does not 
notify FDA in writing before the expiration of the 45-day time period or 
the completion of the agency's review of the application, whichever 
occurs later, that a legal action for patent infringement was filed 
within 45 days of receipt of the notice of certification, approval of 
the abbreviated new drug application or the 505(b)(2) application will 
be made effective immediately upon expiration of the 45 days or upon 
completion of the agency's review and approval of the application, 
whichever is later. The notification to FDA of the legal action shall 
include:
    (i) The abbreviated new drug application or 505(b)(2) application 
number.
    (ii) The name of the abbreviated new drug or 505(b)(2) application 
applicant.
    (iii) The established name of the drug product or, if no established 
name exists, the name(s) of the active ingredient(s), the drug product's 
strength, and dosage form.
    (iv) A certification that an action for patent infringement 
identified by number, has been filed in an appropriate court on a 
specified date.
    The applicant of an abbreviated new drug application shall send the 
notification to FDA's Office of Generic Drugs (HFD-600). A 505(b)(2) 
applicant shall send the notification to the appropriate division in the 
Center for Drug Evaluation and Research reviewing the application. A 
patent owner or its representative may also notify FDA of the filing of 
any legal action for patent infringement. The notice should contain the 
information and be sent to the offices or divisions described in this 
paragraph.
    (3) If the patent owner or approved application holder who is an 
exclusive patent licensee waives its opportunity to file a legal action 
for patent infringement within 45 days of a receipt of the notice of 
certification and the patent owner or approved application holder who is 
an exclusive patent licensee submits to FDA a valid waiver before the 45 
days elapse, approval of the abbreviated new drug application or the 
505(b)(2) application will be made effective upon completion of the 
agency's review and approval of the application. FDA will only accept a 
waiver in the following form:

    (Name of patent owner or exclusive patent licensee) has received 
notice from (name of applicant) under (section 505(b)(3) or 505(j)(2)(B) 
of the act) and does not intend to file an action for patent 
infringement against (name of applicant) concerning the drug (name of 
drug) before (date on which 45 days elapses. (Name of patent owner or 
exclusive patent licensee) waives the opportunity provided by (section 
505(c)(3)(C) or 505(j)(B)(iii) of the act) and does not object to FDA's 
approval of (name of applicant)'s (505(b)(2) or abbreviated new drug 
application) for (name of drug) with an immediate effective date on or 
after the date of this letter.
[59 FR 50367, Oct. 3, 1994]



Sec. 314.108  New drug product exclusivity.

    (a) Definitions. The following definitions of terms apply to this 
section:
    Active moiety means the molecule or ion, excluding those appended 
portions of the molecule that cause the drug to be an ester, salt 
(including a salt with hydrogen or coordination bonds), or

[[Page 148]]

other noncovalent derivative (such as a complex, chelate, or clathrate) 
of the molecule, responsible for the physiological or pharmacological 
action of the drug substance.
    Approved under section 505(b) means an application submitted under 
section 505(b) and approved on or after October 10, 1962, or an 
application that was ``deemed approved'' under section 107(c)(2) of Pub. 
L. 87-781.
    Clinical investigation means any experiment other than a 
bioavailability study in which a drug is administered or dispensed to, 
or used on, human subjects.
    Conducted or sponsored by the applicant with regard to an 
investigation means that before or during the investigation, the 
applicant was named in Form FDA-1571 filed with FDA as the sponsor of 
the investigational new drug application under which the investigation 
was conducted, or the applicant or the applicant's predecessor in 
interest, provided substantial support for the investigation. To 
demonstrate ``substantial support,'' an applicant must either provide a 
certified statement from a certified public accountant that the 
applicant provided 50 percent or more of the cost of conducting the 
study or provide an explanation why FDA should consider the applicant to 
have conducted or sponsored the study if the applicant's financial 
contribution to the study is less than 50 percent or the applicant did 
not sponsor the investigational new drug. A predecessor in interest is 
an entity, e.g., a corporation, that the applicant has taken over, 
merged with, or purchased, or from which the applicant has purchased all 
rights to the drug. Purchase of nonexclusive rights to a clinical 
investigation after it is completed is not sufficient to satisfy this 
definition.
    Date of approval means the date on the letter from FDA stating that 
the new drug application is approved, whether or not final printed 
labeling or other materials must yet be submitted as long as approval of 
such labeling or materials is not expressly required. ``Date of 
approval'' refers only to a final approval and not to a tentative 
approval that may become effective at a later date.
    Essential to approval means, with regard to an investigation, that 
there are no other data available that could support approval of the 
application.
    FDA means the Food and Drug Administration.
    New chemical entity means a drug that contains no active moiety that 
has been approved by FDA in any other application submitted under 
section 505(b) of the act.
    New clinical investigation means an investigation in humans the 
results of which have not been relied on by FDA to demonstrate 
substantial evidence of effectiveness of a previously approved drug 
product for any indication or of safety for a new patient population and 
do not duplicate the results of another investigation that was relied on 
by the agency to demonstrate the effectiveness or safety in a new 
patient population of a previously approved drug product. For purposes 
of this section, data from a clinical investigation previously submitted 
for use in the comprehensive evaluation of the safety of a drug product 
but not to support the effectiveness of the drug product would be 
considered new.
    (b) Submission of and effective date of approval of an abbreviated 
new drug application submitted under section 505(j) of the act or a 
505(b)(2) application. (1) [Reserved]
    (2) If a drug product that contains a new chemical entity was 
approved after September 24, 1984, in an application submitted under 
section 505(b) of the act, no person may submit a 505(b)(2) application 
or abbreviated new drug application under section 505(j) of the act for 
a drug product that contains the same active moiety as in the new 
chemical entity for a period of 5 years from the date of approval of the 
first approved new drug application, except that the 505(b)(2) 
application or abbreviated application may be submitted after 4 years if 
it contains a certification of patent invalidity or noninfringement 
described in Sec. 314.50(i)(1)(i)(A)(4) or Sec. 314.94(a)(12)(i)(A)(4).
    (3) The approval of a 505(b)(2) application or abbreviated 
application described in paragraph (b)(2) of this section will become 
effective as provided in Sec. 314.107(b)(1) or (b)(2), unless the

[[Page 149]]

owner of a patent that claims the drug, the patent owner's 
representative, or exclusive licensee brings suit for patent 
infringement against the applicant during the 1-year period beginning 48 
months after the date of approval of the new drug application for the 
new chemical entity and within 45 days after receipt of the notice 
described at Sec. 314.52 or Sec. 314.95, in which case, approval of the 
505(b)(2) application or abbreviated application will be made effective 
as provided in Sec. 314.107(b)(3).
    (4) If an application:
    (i) Was submitted under section 505(b) of the act;
    (ii) Was approved after September 24, 1984;
    (iii) Was for a drug product that contains an active moiety that has 
been previously approved in another application under section 505(b) of 
the act; and
    (iv) Contained reports of new clinical investigations (other than 
bioavailability studies) conducted or sponsored by the applicant that 
were essential to approval of the application, the agency will not make 
effective for a period of 3 years after the date of approval of the 
application the approval of a 505(b)(2) application or an abbreviated 
new drug application for the conditions of approval of the original 
application, or an abbreviated new drug application submitted pursuant 
to an approved petition under section 505(j)(2)(C) of the act that 
relies on the information supporting the conditions of approval of an 
original new drug application.
    (5) If a supplemental application:
    (i) Was approved after September 24, 1984; and
    (ii) Contained reports of new clinical investigations (other than 
bioavailability studies) that were conducted or sponsored by the 
applicant that were essential to approval of the supplemental 
application, the agency will not make effective for a period of 3 years 
after the date of approval of the supplemental application the approval 
of a 505(b)(2) application or an abbreviated new drug application for a 
change, or an abbreviated new drug application submitted pursuant to an 
approved petition under section 505(j)(2)(C) of the act that relies on 
the information supporting a change approved in the supplemental new 
drug application.
[59 FR 50368, Oct. 3, 1994]



Sec. 314.110  Approvable letter to the applicant.

    (a) In selected circumstances, it is useful at the end of the review 
period for the Food and Drug Administration to indicate to the applicant 
that the application or abbreviated application is basically approvable 
providing certain issues are resolved. An approvable letter may be 
issued in such circumstances. FDA will send the applicant an approvable 
letter if the application or abbreviated application substantially meets 
the requirements of this part and the agency believes that it can 
approve the application or abbreviated application if specific 
additional information or material is submitted or specific conditions 
(for example, certain changes in labeling) are agreed to by the 
applicant. The approvable letter will describe the information or 
material FDA requires or the conditions the applicant is asked to meet. 
As a practical matter, the approvable letter will serve in most 
instances as a mechanism for resolving outstanding issues on drugs that 
are about to be approved and marketed. For an application or an 
abbreviated antibiotic application, the applicant shall, within 10 days 
after the date of the approvable letter:
    (1) Amend the application or abbreviated antibiotic application or 
notify FDA of an intent to file an amendment. The filing of an amendment 
or notice of intent to file an amendment constitutes an agreement by the 
applicant to extend the review period for 45 days after the date FDA 
receives the amendment. The extension is to permit the agency to review 
the amendment;
    (2) Withdraw the application or abbreviated antibiotic application. 
FDA will consider the applicant's failure to respond within 10 days to 
an approvable letter to be a request by the applicant to withdraw the 
application under Sec. 314.65 or the abbreviated antibiotic application 
under Sec. 314.99. A decision to withdraw an application or abbreviated 
antibiotic application is without prejudice to a refiling;

[[Page 150]]

    (3) For a new drug application or abbreviated antibiotic 
application, ask the agency to provide the applicant an opportunity for 
a hearing on the question of whether there are grounds for denying 
approval of the application under section 505(d) of the act. The 
applicant shall submit the request to the Associate Director for Policy 
(HFD-5), Center for Drug Evaluation and Research, Food and Drug 
Administration, 5600 Fishers Lane, Rockville, MD 20857. Within 60 days 
of the date of the approvable letter, or within a different time period 
to which FDA and the applicant agree, the agency will either approve the 
application or abbreviated antibiotic application under Sec. 314.105 or 
refuse to approve the application or abbreviated antibiotic application 
under Sec. 314.125 and give the applicant written notice of an 
opportunity for a hearing under Sec. 314.200 and section 505(c)(2) of 
the act on the question of whether there are grounds for denying 
approval of the application under section 505(d) of the act;
    (4) For an antibiotic, file a petition or notify FDA of an intent to 
file a petition proposing the issuance, amendment, or repeal of a 
regulation under Sec. 314.300 and section 507(f) of the act; or
    (5) Notify FDA that the applicant agrees to an extension of the 
review period under section 505(c) of the act, so that the applicant can 
determine whether to respond further under paragraph (a)(1), (a)(2), 
(a)(3), or (a)(4) of this section. The applicant's notice is required to 
state the length of the extension. FDA will honor any reasonable request 
for such an extension. FDA will consider the applicant's failure to 
respond further within the extended review period to be a request to 
withdraw the application under Sec. 314.65 or the abbreviated antibiotic 
application under Sec. 314.99. A decision to withdraw an application or 
abbreviated antibiotic application is without prejudice to a refiling.
    (b) FDA will send the applicant of an abbreviated new drug 
application an approvable letter only if the application substantially 
meets the requirements of this part and the agency believes that it can 
approve the abbreviated application if minor deficiencies (e.g., 
labeling deficiencies) are corrected. The approvable letter will 
describe the deficiencies and state a time period within which the 
applicant must respond. Unless the applicant corrects the deficiencies 
by amendment within the specified time period, FDA will refuse to 
approve the abbreviated application under Sec. 314.127. Within 10 days 
after the date of the approvable letter, the applicant may also ask the 
agency to provide the applicant an opportunity for a hearing on the 
question of whether there are grounds for denying approval of the 
abbreviated new drug application. Applicants who request a hearing shall 
submit the request to the Associate Director for Policy (HFD-5), Center 
for Drug Evaluation and Research, Food and Drug Administration, 5600 
Fishers Lane, Rockville, MD 20857.
[57 FR 17989, Apr. 28, 1992, as amended at 62 FR 43639, Aug. 15, 1997]



Sec. 314.120  Not approvable letter to the applicant.

    (a) The Food and Drug Administration will send the applicant a not 
approvable letter if the agency believes that the application or 
abbreviated antibiotic application may not be approved for one of the 
reasons given in Sec. 314.125 or the abbreviated new drug application 
may not be approved for one of the reasons given in Sec. 314.127. The 
not approvable letter will describe the deficiencies in the application 
or abbreviated application. Except as provided in paragraph (b) of this 
section, within 10 days after the date of the not approvable letter, the 
applicant shall:
    (1) Amend the application or abbreviated application or notify FDA 
of an intent to file an amendment. The filing of an amendment or a 
notice of intent to file an amendment constitutes an agreement by the 
applicant to extend the review period under Sec. 314.60 or Sec. 314.96;
    (2) Withdraw the application or abbreviated application. Except as 
provided in paragraph (b) of this section, FDA will consider the 
applicant's failure to respond within 10 days to a not approvable letter 
to be a request by the applicant to withdraw the application

[[Page 151]]

under Sec. 314.65 or abbreviated application under Sec. 314.99. A 
decision to withdraw the application or abbreviated application is 
without prejudice to refiling;
    (3) For a new drug application or an abbreviated application, ask 
the agency to provide the applicant an opportunity for a hearing on the 
question of whether there are grounds for denying approval of the 
application under section 505(d) or (j)(3) of the act. The applicant 
shall submit the request to the Associate Director for Policy (HFD-5), 
Center for Drug Evaluation and Research, Food and Drug Administration, 
5600 Fishers Lane, Rockville, MD 20857. Within 60 days of the date of 
the not approvable letter, or within a different time period to which 
FDA and the applicant agree, the agency will either approve the 
application or abbreviated application under Sec. 314.105 or refuse to 
approve the application or abbreviated antibiotic application under 
Sec. 314.125 or abbreviated new drug application under Sec. 314.127 and 
give the applicant written notice of an opportunity for a hearing under 
Sec. 314.200 and section 505(c)(1)(B) or (j)(4)(C) of the act on the 
question of whether there are grounds for denying approval of the 
application under section 505(d) or (j)(3) of the act;
    (4) For an antibiotic application, file a petition or notify FDA of 
an intent to file a petition proposing the issuance, amendment, or 
repeal of a regulation under Sec. 314.300 and section 507(f) of the act; 
or
    (5) Notify FDA that the applicant agrees to an extension of the 
review period under section 505(c)(1) or (j)(4)(A) of the act, so that 
the applicant can determine whether to respond further under paragraph 
(a)(1), (a)(2), (a)(3), or (a)(4) of this section. The applicant's 
notice is required to state the length of the extension. FDA will honor 
any reasonable request for such an extension. FDA will consider the 
applicant's failure to respond further within the extended review period 
to be a request to withdraw the application under Sec. 314.65 or 
abbreviated application under Sec. 314.99. A decision to withdraw an 
application or abbreviated application is without prejudice to a 
refiling.
    (b) With the exception of a request for an opportunity for a hearing 
under paragraph (a)(3) of this section, the 10-day time period in this 
section for responding to a not approvable letter does not apply to 
abbreviated new drug applications. FDA may consider the applicant's 
failure to respond within 180 days to a not approvable letter to be a 
request by the applicant to withdraw the abbreviated new drug 
application under Sec. 314.99.
[57 FR 17990, Apr. 28, 1992, as amended at 62 FR 43639, Aug. 15, 1997]



Sec. 314.122  Submitting an abbreviated application for, or a 505(j)(2)(C) petition that relies on, a listed drug that is no longer marketed.

    (a) An abbreviated new drug application that refers to, or a 
petition under section 505(j)(2)(C) of the act and Sec. 314.93 that 
relies on, a listed drug that has been voluntarily withdrawn from sale 
in the United States must be accompanied by a petition seeking a 
determination whether the listed drug was withdrawn for safety or 
effectiveness reasons. The petition must be submitted under 
Secs. 10.25(a) and 10.30 of this chapter and must contain all evidence 
available to the petitioner concerning the reasons for the withdrawal 
from sale.
    (b) When a petition described in paragraph (a) of this section is 
submitted, the agency will consider the evidence in the petition and any 
other evidence before the agency, and determine whether the listed drug 
is withdrawn from sale for safety or effectiveness reasons, in 
accordance with the procedures in Sec. 314.161.
    (c) An abbreviated new drug application described in paragraph (a) 
of this section will be disapproved, under Sec. 314.127(a)(11), and a 
505(j)(2)(C) petition described in paragraph (a) of this section will be 
disapproved, under Sec. 314.93(e)(1)(iv), unless the agency determines 
that the withdrawal of the listed drug was not for safety or 
effectiveness reasons.
    (d) Certain drug products approved for safety and effectiveness that 
were no longer marketed on September 24, 1984, are not included in the 
list. Any person who wishes to obtain marketing approval for such a drug 
product under an abbreviated new drug application must petition FDA for 
a determination

[[Page 152]]

whether the drug product was withdrawn from the market for safety or 
effectiveness reasons and request that the list be amended to include 
the drug product. A person seeking such a determination shall use the 
petition procedures established in Sec. 10.30 of this chapter. The 
petitioner shall include in the petition information to show that the 
drug product was approved for safety and effectiveness and all evidence 
available to the petitioner concerning the reason that marketing of the 
drug product ceased.
[57 FR 17990, Apr. 28, 1992; 57 FR 29353, July 1, 1992]



Sec. 314.125  Refusal to approve an application or abbreviated antibiotic application.

    (a) The Food and Drug Administration will refuse to approve the 
application or abbreviated antibiotic application and for a new drug 
give the applicant written notice of an opportunity for a hearing under 
Sec. 314.200 on the question of whether there are grounds for denying 
approval of the application under section 505(d) of the act, or for an 
antibiotic publish a proposed regulation based on an acceptable petition 
under Sec. 314.300, if:
    (1) FDA sends the applicant an approvable or a not approvable letter 
under Sec. 314.110 or Sec. 314.120;
    (2) The applicant requests an opportunity for hearing for a new drug 
on the question of whether the application is approvable or files a 
petition for an antibiotic proposing the issuance, amendment, or repeal 
of a regulation; and
    (3) FDA finds that any of the reasons given in paragraph (b) of this 
section apply.
    (b) FDA may refuse to approve an application or abbreviated 
antibiotic application for any of the following reasons:
    (1) The methods to be used in, and the facilities and controls used 
for, the manufacture, processing, packing, or holding of the drug 
substance or the drug product are inadequate to preserve its identity, 
strength, quality, purity, stability, and bioavailability.
    (2) The investigations required under section 505(b) or 507 of the 
act do not include adequate tests by all methods reasonably applicable 
to show whether or not the drug is safe for use under the conditions 
prescribed, recommended, or suggested in its proposed labeling.
    (3) The results of the tests show that the drug is unsafe for use 
under the conditions prescribed, recommended, or suggested in its 
proposed labeling or the results do not show that the drug product is 
safe for use under those conditions.
    (4) There is insufficient information about the drug to determine 
whether the product is safe for use under the conditions prescribed, 
recommended, or suggested in its proposed labeling.
    (5) There is a lack of substantial evidence consisting of adequate 
and well-controlled investigations, as defined in Sec. 314.126, that the 
drug product will have the effect it purports or is represented to have 
under the conditions of use prescribed, recommended, or suggested in its 
proposed labeling.
    (6) The proposed labeling is false or misleading in any particular.
    (7) The application or abbreviated antibiotic application contains 
an untrue statement of a material fact.
    (8) The drug product's proposed labeling does not comply with the 
requirements for labels and labeling in part 201.
    (9) The application or abbreviated antibiotic application does not 
contain bioavailability or bioequivalence data required under part 320 
of this chapter.
    (10) A reason given in a letter refusing to file the application or 
abbreviated antibiotic application under Sec. 314.101(d), if the 
deficiency is not corrected.
    (11) The drug will be manufactured or processed in whole or in part 
in an establishment that is not registered and not exempt from 
registration under section 510 of the act and part 207.
    (12) The applicant does not permit a properly authorized officer or 
employee of the Department of Health and Human Services an adequate 
opportunity to inspect the facilities, controls, and any records 
relevant to the application or abbreviated antibiotic application.
    (13) The methods to be used in, and the facilities and controls used 
for, the manufacture, processing, packing, or holding of the drug 
substance or the

[[Page 153]]

drug product do not comply with the current good manufacturing practice 
regulations in parts 210 and 211.
    (14) The application or abbreviated antibiotic application does not 
contain an explanation of the omission of a report of any investigation 
of the drug product sponsored by the applicant, or an explanation of the 
omission of other information about the drug pertinent to an evaluation 
of the application or abbreviated antibiotic application that is 
received or otherwise obtained by the applicant from any source.
    (15) A nonclinical laboratory study that is described in the 
application or abbreviated antibiotic application and that is essential 
to show that the drug is safe for use under the conditions prescribed, 
recommended, or suggested in its proposed labeling was not conducted in 
compliance with the good laboratory practice regulations in part 58 of 
this chapter and no reason for the noncompliance is provided or, if it 
is, the differences between the practices used in conducting the study 
and the good laboratory practice regulations do not support the validity 
of the study.
    (16) Any clinical investigation involving human subjects described 
in the application or abbreviated antibiotic application, subject to the 
institutional review board regulations in part 58 of this chapter or 
informed consent regulations in part 50 of this chapter, was not 
conducted in compliance with those regulations such that the rights or 
safety of human subjects were not adequately protected.
    (17) The applicant or contract research organization that conducted 
a bioavailability or bioequivalence study described in Sec. 320.38 or 
Sec. 320.63 of this chapter that is contained in the application or 
abbreviated antibiotic application refuses to permit an inspection of 
facilities or records relevant to the study by a properly authorized 
officer or employee of the Department of Health and Human Services or 
refuses to submit reserve samples of the drug products used in the study 
when requested by FDA.
    (18) For a new drug, the application failed to contain the patent 
information required by section 505(b)(1) of the act.
    (c) For drugs intended to treat life-threatening or severely-
debilitating illnesses that are developed in accordance with 
Secs. 312.80 through 312.88 of this chapter, the criteria contained in 
paragraphs (b) (3), (4), and (5) of this section shall be applied 
according to the considerations contained in Sec. 312.84 of this 
chapter.
[50 FR 7493, Feb. 22, 1985, as amended at 53 FR 41524, Oct. 21, 1988; 57 
FR 17991, Apr. 28, 1992; 58 FR 25926, Apr. 28, 1993]



Sec. 314.126  Adequate and well-controlled studies.

    (a) The purpose of conducting clinical investigations of a drug is 
to distinguish the effect of a drug from other influences, such as 
spontaneous change in the course of the disease, placebo effect, or 
biased observation. The characteristics described in paragraph (b) of 
this section have been developed over a period of years and are 
recognized by the scientific community as the essentials of an adequate 
and well-controlled clinical investigation. The Food and Drug 
Administration considers these characteristics in determining whether an 
investigation is adequate and well-controlled for purposes of sections 
505 and 507 of the act. Reports of adequate and well-controlled 
investigations provide the primary basis for determining whether there 
is ``substantial evidence'' to support the claims of effectiveness for 
new drugs and antibiotics. Therefore, the study report should provide 
sufficient details of study design, conduct, and analysis to allow 
critical evaluation and a determination of whether the characteristics 
of an adequate and well-controlled study are present.
    (b) An adequate and well-controlled study has the following 
characteristics:
    (1) There is a clear statement of the objectives of the 
investigation and a summary of the proposed or actual methods of 
analysis in the protocol for the study and in the report of its results. 
In addition, the protocol should contain a description of the proposed 
methods of analysis, and the study report should contain a description 
of the methods of analysis ultimately used. If

[[Page 154]]

the protocol does not contain a description of the proposed methods of 
analysis, the study report should describe how the methods used were 
selected.
    (2) The study uses a design that permits a valid comparison with a 
control to provide a quantitative assessment of drug effect. The 
protocol for the study and report of results should describe the study 
design precisely; for example, duration of treatment periods, whether 
treatments are parallel, sequential, or crossover, and whether the 
sample size is predetermined or based upon some interim analysis. 
Generally, the following types of control are recognized:
    (i) Placebo concurrent control. The test drug is compared with an 
inactive preparation designed to resemble the test drug as far as 
possible. A placebo-controlled study may include additional treatment 
groups, such as an active treatment control or a dose-comparison 
control, and usually includes randomization and blinding of patients or 
investigators, or both.
    (ii) Dose-comparison concurrent control. At least two doses of the 
drug are compared. A dose-comparison study may include additional 
treatment groups, such as placebo control or active control. Dose-
comparison trials usually include randomization and blinding of patients 
or investigators, or both.
    (iii) No treatment concurrent control. Where objective measurements 
of effectiveness are available and placebo effect is negligible, the 
test drug is compared with no treatment. No treatment concurrent control 
trials usually include randomization.
    (iv) Active treatment concurrent control. The test drug is compared 
with known effective therapy; for example, where the condition treated 
is such that administration of placebo or no treatment would be contrary 
to the interest of the patient. An active treatment study may include 
additional treatment groups, however, such as a placebo control or a 
dose-comparison control. Active treatment trials usually include 
randomization and blinding of patients or investigators, or both. If the 
intent of the trial is to show similarity of the test and control drugs, 
the report of the study should assess the ability of the study to have 
detected a difference between treatments. Similarity of test drug and 
active control can mean either that both drugs were effective or that 
neither was effective. The analysis of the study should explain why the 
drugs should be considered effective in the study, for example, by 
reference to results in previous placebo-controlled studies of the 
active control drug.
    (v) Historical control. The results of treatment with the test drug 
are compared with experience historically derived from the adequately 
documented natural history of the disease or condition, or from the 
results of active treatment, in comparable patients or populations. 
Because historical control populations usually cannot be as well 
assessed with respect to pertinent variables as can concurrent control 
populations, historical control designs are usually reserved for special 
circumstances. Examples include studies of diseases with high and 
predictable mortality (for example, certain malignancies) and studies in 
which the effect of the drug is self-evident (general anesthetics, drug 
metabolism).
    (3) The method of selection of subjects provides adequate assurance 
that they have the disease or condition being studied, or evidence of 
susceptibility and exposure to the condition against which prophylaxis 
is directed.
    (4) The method of assigning patients to treatment and control groups 
minimizes bias and is intended to assure comparability of the groups 
with respect to pertinent variables such as age, sex, severity of 
disease, duration of disease, and use of drugs or therapy other than the 
test drug. The protocol for the study and the report of its results 
should describe how subjects were assigned to groups. Ordinarily, in a 
concurrently controlled study, assignment is by randomization, with or 
without stratification.
    (5) Adequate measures are taken to minimize bias on the part of the 
subjects, observers, and analysts of the data. The protocol and report 
of the study should describe the procedures used to accomplish this, 
such as blinding.
    (6) The methods of assessment of subjects' response are well-defined 
and reliable. The protocol for the study and

[[Page 155]]

the report of results should explain the variables measured, the methods 
of observation, and criteria used to assess response.
    (7) There is an analysis of the results of the study adequate to 
assess the effects of the drug. The report of the study should describe 
the results and the analytic methods used to evaluate them, including 
any appropriate statistical methods. The analysis should assess, among 
other things, the comparability of test and control groups with respect 
to pertinent variables, and the effects of any interim data analyses 
performed.
    (c) The Director of the Center for Drug Evaluation and Research may, 
on the Director's own initiative or on the petition of an interested 
person, waive in whole or in part any of the criteria in paragraph (b) 
of this section with respect to a specific clinical investigation, 
either prior to the investigation or in the evaluation of a completed 
study. A petition for a waiver is required to set forth clearly and 
concisely the specific criteria from which waiver is sought, why the 
criteria are not reasonably applicable to the particular clinical 
investigation, what alternative procedures, if any, are to be, or have 
been employed, and what results have been obtained. The petition is also 
required to state why the clinical investigations so conducted will 
yield, or have yielded, substantial evidence of effectiveness, 
notwithstanding nonconformance with the criteria for which waiver is 
requested.
    (d) For an investigation to be considered adequate for approval of a 
new drug, it is required that the test drug be standardized as to 
identity, strength, quality, purity, and dosage form to give 
significance to the results of the investigation.
    (e) Uncontrolled studies or partially controlled studies are not 
acceptable as the sole basis for the approval of claims of 
effectiveness. Such studies carefully conducted and documented, may 
provide corroborative support of well-controlled studies regarding 
efficacy and may yield valuable data regarding safety of the test drug. 
Such studies will be considered on their merits in the light of the 
principles listed here, with the exception of the requirement for the 
comparison of the treated subjects with controls. Isolated case reports, 
random experience, and reports lacking the details which permit 
scientific evaluation will not be considered.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0001)

[50 FR 7493, Feb. 22, 1985, as amended at 50 FR 21238, May 23, 1985; 55 
FR 11580, Mar. 29, 1990]



Sec. 314.127  Refusal to approve an abbreviated new drug application.

    (a) FDA will refuse to approve an abbreviated application for a new 
drug under section 505(j) of the act for any of the following reasons:
    (1) The methods used in, or the facilities and controls used for, 
the manufacture, processing, and packing of the drug product are 
inadequate to ensure and preserve its identity, strength, quality, and 
purity.
    (2) Information submitted with the abbreviated new drug application 
is insufficient to show that each of the proposed conditions of use has 
been previously approved for the listed drug referred to in the 
application.
    (3)(i) If the reference listed drug has only one active ingredient, 
information submitted with the abbreviated new drug application is 
insufficient to show that the active ingredient is the same as that of 
the reference listed drug;
    (ii) If the reference listed drug has more than one active 
ingredient, information submitted with the abbreviated new drug 
application is insufficient to show that the active ingredients are the 
same as the active ingredients of the reference listed drug; or
    (iii) If the reference listed drug has more than one active 
ingredient and if the abbreviated new drug application is for a drug 
product that has an active ingredient different from the reference 
listed drug:
    (A) Information submitted with the abbreviated new drug application 
is insufficient to show:
    (1) That the other active ingredients are the same as the active 
ingredients of the reference listed drug; or
    (2) That the different active ingredient is an active ingredient of 
a listed

[[Page 156]]

drug or a drug that does not meet the requirements of section 201(p) of 
the act; or
    (B) No petition to submit an abbreviated application for the drug 
product with the different active ingredient was approved under 
Sec. 314.93.
    (4)(i) If the abbreviated new drug application is for a drug product 
whose route of administration, dosage form, or strength purports to be 
the same as that of the listed drug referred to in the abbreviated new 
drug application, information submitted in the abbreviated new drug 
application is insufficient to show that the route of administration, 
dosage form, or strength is the same as that of the reference listed 
drug; or
    (ii) If the abbreviated new drug application is for a drug product 
whose route of administration, dosage form, or strength is different 
from that of the listed drug referred to in the application, no petition 
to submit an abbreviated new drug application for the drug product with 
the different route of administration, dosage form, or strength was 
approved under Sec. 314.93.
    (5) If the abbreviated new drug application was submitted under the 
approval of a petition under Sec. 314.93, the abbreviated new drug 
application did not contain the information required by FDA with respect 
to the active ingredient, route of administration, dosage form, or 
strength that is not the same as that of the reference listed drug.
    (6)(i) Information submitted in the abbreviated new drug application 
is insufficient to show that the drug product is bioequivalent to the 
listed drug referred to in the abbreviated new drug application; or
    (ii) If the abbreviated new drug application was submitted under a 
petition approved under Sec. 314.93, information submitted in the 
abbreviated new drug application is insufficient to show that the active 
ingredients of the drug product are of the same pharmacological or 
therapeutic class as those of the reference listed drug and that the 
drug product can be expected to have the same therapeutic effect as the 
reference listed drug when administered to patients for each condition 
of use approved for the reference listed drug.
    (7) Information submitted in the abbreviated new drug application is 
insufficient to show that the labeling proposed for the drug is the same 
as the labeling approved for the listed drug referred to in the 
abbreviated new drug application except for changes required because of 
differences approved in a petition under Sec. 314.93 or because the drug 
product and the reference listed drug are produced or distributed by 
different manufacturers or because aspects of the listed drug's labeling 
are protected by patent, or by exclusivity, and such differences do not 
render the proposed drug product less safe or effective than the listed 
drug for all remaining, nonprotected conditions of use.
    (8)(i) Information submitted in the abbreviated new drug application 
of any other information available to FDA shows that:
    (A) The inactive ingredients of the drug product are unsafe for use, 
as described in paragraph (a)(8)(ii) of this section, under the 
conditions prescribed, recommended, or suggested in the labeling 
proposed for the drug product; or
    (B) The composition of the drug product is unsafe, as described in 
paragraph (a)(8)(ii) of this section, under the conditions prescribed, 
recommended, or suggested in the proposed labeling because of the type 
or quantity of inactive ingredients included or the manner in which the 
inactive ingredients are included.
    (ii)(A) FDA will consider the inactive ingredients or composition of 
a drug product unsafe and refuse to approve an abbreviated new drug 
application under paragraph (a)(8)(i) of this section if, on the basis 
of information available to the agency, there is a reasonable basis to 
conclude that one or more of the inactive ingredients of the proposed 
drug or its composition raises serious questions of safety. From its 
experience with reviewing inactive ingredients, and from other 
information available to it, FDA may identify changes in inactive 
ingredients or composition that may adversely affect a drug product's 
safety. The inactive ingredients or composition of a proposed drug 
product will be considered to raise serious questions of safety if the 
product incorporates one or more of these

[[Page 157]]

changes. Examples of the changes that may raise serious questions of 
safety include, but are not limited to, the following:
    (1) A change in an inactive ingredient so that the product does not 
comply with an official compendium.
    (2) A change in composition to include an inactive ingredient that 
has not been previously approved in a drug product for human use by the 
same route of administration.
    (3) A change in the composition of a parenteral drug product to 
include an inactive ingredient that has not been previously approved in 
a parenteral drug product.
    (4) A change in composition of a drug product for ophthalmic use to 
include an inactive ingredient that has not been previously approved in 
a drug for ophthalmic use.
    (5) The use of a delivery or a modified release mechanism never 
before approved for the drug.
    (6) A change in composition to include a significantly greater 
content of one or more inactive ingredients than previously used in the 
drug product.
    (7) If the drug product is intended for topical administration, a 
change in the properties of the vehicle or base that might increase 
absorption of certain potentially toxic active ingredients thereby 
affecting the safety of the drug product, or a change in the lipophilic 
properties of a vehicle or base, e.g., a change from an oleaginous to a 
water soluble vehicle or base.
    (B) FDA will consider an inactive ingredient in, or the composition 
of, a drug product intended for parenteral use to be unsafe and will 
refuse to approve the abbreviated new drug application unless it 
contains the same inactive ingredients, other than preservatives, 
buffers, and antioxidants, in the same concentration as the listed drug, 
and, if it differs from the listed drug in a preservative, buffer, or 
antioxidant, the application contains sufficient information to 
demonstrate that the difference does not affect the safety of the drug 
product.
    (C) FDA will consider an inactive ingredient in, or the composition 
of, a drug product intended for ophthalmic or otic use unsafe and will 
refuse to approve the abbreviated new drug application unless it 
contains the same inactive ingredients, other than preservatives, 
buffers, substances to adjust tonicity, or thickening agents, in the 
same concentration as the listed drug, and if it differs from the listed 
drug in a preservative, buffer, substance to adjust tonicity, or 
thickening agent, the application contains sufficient information to 
demonstrate that the difference does not affect the safety of the drug 
product and the labeling does not claim any therapeutic advantage over 
or difference from the listed drug.
    (9) Approval of the listed drug referred to in the abbreviated new 
drug application has been withdrawn or suspended for grounds described 
in Sec. 314.150(a) or FDA has published a notice of opportunity for 
hearing to withdraw approval of the reference listed drug under 
Sec. 314.150(a).
    (10) Approval of the listed drug referred to in the abbreviated new 
drug application has been withdrawn under Sec. 314.151 or FDA has 
proposed to withdraw approval of the reference listed drug under 
Sec. 314.151(a).
    (11) FDA has determined that the reference listed drug has been 
withdrawn from sale for safety or effectiveness reasons under 
Sec. 314.161, or the reference listed drug has been voluntarily 
withdrawn from sale and the agency has not determined whether the 
withdrawal is for safety or effectiveness reasons, or approval of the 
reference listed drug has been suspended under Sec. 314.153, or the 
agency has issued an initial decision proposing to suspend the reference 
listed drug under Sec. 314.153(a)(1).
    (12) The abbreviated new drug application does not meet any other 
requirement under section 505(j)(2)(A) of the act.
    (13) The abbreviated new drug application contains an untrue 
statement of material fact.
    (b) FDA may refuse to approve an abbreviated application for a new 
drug if the applicant or contract research organization that conducted a 
bioavailability or bioequivalence study described in Sec. 320.63 of this 
chapter that is contained in the abbreviated new drug application 
refuses to permit an

[[Page 158]]

inspection of facilities or records relevant to the study by a properly 
authorized officer of employee of the Department of Health and Human 
Services or refuses to submit reserve samples of the drug products used 
in the study when requested by FDA.
[57 FR 17991, Apr. 28, 1992; 57 FR 29353, July 1, 1992, as amended at 58 
FR 25927, Apr. 28, 1993]



Sec. 314.150  Withdrawal of approval of an application or abbreviated application.

    (a) The Food and Drug Administration will notify the applicant, and, 
if appropriate, all other persons who manufacture or distribute 
identical, related, or similar drug products as defined in Secs. 310.6 
and 314.151(a) of this chapter and for a new drug afford an opportunity 
for a hearing on a proposal to withdraw approval of the application or 
abbreviated new drug application under section 505(e) of the act and 
under the procedure in Sec. 314.200, or, for an antibiotic, rescind a 
certification or release, or amend or repeal a regulation providing for 
certification under section 507 of the act and under the procedure in 
Sec. 314.300, if any of the following apply:
    (1) The Secretary of Health and Human Services has suspended the 
approval of the application or abbreviated application for a new drug on 
a finding that there is an imminent hazard to the public health. FDA 
will promptly afford the applicant an expedited hearing following 
summary suspension on a finding of imminent hazard to health.
    (2) FDA finds:
    (i) That clinical or other experience, tests, or other scientific 
data show that the drug is unsafe for use under the conditions of use 
upon the basis of which the application or abbreviated application was 
approved; or
    (ii) That new evidence of clinical experience, not contained in the 
application or not available to FDA until after the application or 
abbreviated application was approved, or tests by new methods, or tests 
by methods not deemed reasonably applicable when the application or 
abbreviated application was approved, evaluated together with the 
evidence available when the application or abbreviated application was 
approved, reveal that the drug is not shown to be safe for use under the 
conditions of use upon the basis of which the application or abbreviated 
application was approved; or
    (iii) Upon the basis of new information before FDA with respect to 
the drug, evaluated together with the evidence available when the 
application or abbreviated application was approved, that there is a 
lack of substantial evidence from adequate and well-controlled 
investigations as defined in Sec. 314.126, that the drug will have the 
effect it is purported or represented to have under the conditions of 
use prescribed, recommended, or suggested in its labeling; or
    (iv) That the application or abbreviated application contains any 
untrue statement of a material fact; or
    (v) That the patent information prescribed by section 505(c) of the 
act was not submitted within 30 days after the receipt of written notice 
from FDA specifying the failure to submit such information; or
    (b) FDA may notify the applicant, and, if appropriate, all other 
persons who manufacture or distribute identical, related, or similar 
drug products as defined in Sec. 310.6, and for a new drug afford an 
opportunity for a hearing on a proposal to withdraw approval of the 
application or abbreviated new drug application under section 505(e) of 
the act and under the procedure in Sec. 314.200, or, for an antibiotic, 
rescind a certification or release, or amend or repeal a regulation 
providing for certification under section 507 of the act and the 
procedure in Sec. 314.300, if the agency finds:
    (1) That the applicant has failed to establish a system for 
maintaining required records, or has repeatedly or deliberately failed 
to maintain required records or to make required reports under section 
505(k) or 507(g) of the act and Sec. 314.80, Sec. 314.81, or 
Sec. 314.98, or that the applicant has refused to permit access to, or 
copying or verification of, its records.
    (2) That on the basis of new information before FDA, evaluated 
together with the evidence available when the application or abbreviated 
application was approved, the methods used in, or

[[Page 159]]

the facilities and controls used for, the manufacture, processing, and 
packing of the drug are inadequate to ensure and preserve its identity, 
strength, quality, and purity and were not made adequate within a 
reasonable time after receipt of written notice from the agency.
    (3) That on the basis of new information before FDA, evaluated 
together with the evidence available when the application or abbreviated 
application was approved, the labeling of the drug, based on a fair 
evaluation of all material facts, is false or misleading in any 
particular, and the labeling was not corrected by the applicant within a 
reasonable time after receipt of written notice from the agency.
    (4) That the applicant has failed to comply with the notice 
requirements of section 510(j)(2) of the act.
    (5) That the applicant has failed to submit bioavailability or 
bioequivalence data required under part 320 of this chapter.
    (6) The application or abbreviated application does not contain an 
explanation of the omission of a report of any investigation of the drug 
product sponsored by the applicant, or an explanation of the omission of 
other information about the drug pertinent to an evaluation of the 
application or abbreviated application that is received or otherwise 
obtained by the applicant from any source.
    (7) That any nonclinical laboratory study that is described in the 
application or abbreviated application and that is essential to show 
that the drug is safe for use under the conditions prescribed, 
recommended, or suggested in its labeling was not conducted in 
compliance with the good laboratory practice regulations in part 58 of 
this chapter and no reason for the noncompliance was provided or, if it 
was, the differences between the practices used in conducting the study 
and the good laboratory practice regulations do not support the validity 
of the study.
    (8) Any clinical investigation involving human subjects described in 
the application or abbreviated application, subject to the institutional 
review board regulations in part 56 of this chapter or informed consent 
regulations in part 50 of this chapter, was not conducted in compliance 
with those regulations such that the rights or safety of human subjects 
were not adequately protected.
    (9) That the applicant or contract research organization that 
conducted a bioavailability or bioequivalence study described in 
Sec. 320.38 or Sec. 320.63 of this chapter that is contained in the 
application or abbreviated application refuses to permit an inspection 
of facilities or records relevant to the study by a properly authorized 
officer or employee of the Department of Health and Human Services or 
refuses to submit reserve samples of the drug products used in the study 
when requested by FDA.
    (10) That the labeling for the drug product that is the subject of 
the abbreviated new drug application is no longer consistent with that 
for the listed drug referred to in the abbreviated new drug application, 
except for differences approved in the abbreviated new drug application 
or those differences resulting from:
    (i) A patent on the listed drug issued after approval of the 
abbreviated new drug application; or
    (ii) Exclusivity accorded to the listed drug after approval of the 
abbreviated new drug application that do not render the drug product 
less safe or effective than the listed drug for any remaining, 
nonprotected condition(s) of use.
    (c) FDA will withdraw approval of an application or abbreviated 
application if the applicant requests its withdrawal because the drug 
subject to the application or abbreviated application is no longer being 
marketed, provided none of the conditions listed in paragraphs (a) and 
(b) of this section applies to the drug. FDA will consider a written 
request for a withdrawal under this paragraph to be a waiver of an 
opportunity for hearing otherwise provided for in this section. 
Withdrawal of approval of an application or abbreviated application 
under this paragraph is without prejudice to refiling.
    (d) FDA may notify an applicant that it believes a potential problem 
associated with a drug is sufficiently serious that the drug should be 
removed from the market and may ask the applicant to waive the 
opportunity for hearing

[[Page 160]]

otherwise provided for under this section, to permit FDA to withdraw 
approval of the application or abbreviated application for the product, 
and to remove voluntarily the product from the market. If the applicant 
agrees, the agency will not make a finding under paragraph (b) of this 
section, but will withdraw approval of the application or abbreviated 
application in a notice published in the Federal Register that contains 
a brief summary of the agency's and the applicant's views of the reasons 
for withdrawal.
[57 FR 17993, Apr. 28, 1992, as amended at 58 FR 25927, Apr. 28, 1993]



Sec. 314.151  Withdrawal of approval of an abbreviated new drug application under section 505(j)(5) of the act.

    (a) Approval of an abbreviated new drug application approved under 
Sec. 314.105(d) may be withdrawn when the agency withdraws approval, 
under Sec. 314.150(a) or under this section, of the approved drug 
referred to in the abbreviated new drug application. If the agency 
proposed to withdraw approval of a listed drug under Sec. 314.150(a), 
the holder of an approved application for the listed drug has a right to 
notice and opportunity for hearing. The published notice of opportunity 
for hearing will identify all drug products approved under 
Sec. 314.105(d) whose applications are subject to withdrawal under this 
section if the listed drug is withdrawn, and will propose to withdraw 
such drugs. Holders of approved applications for the identified drug 
products will be provided notice and an opportunity to respond to the 
proposed withdrawal of their applications as described in paragraphs (b) 
and (c) of this section.
    (b)(1) The published notice of opportunity for hearing on the 
withdrawal of the listed drug will serve as notice to holders of 
identified abbreviated new drug applications of the grounds for the 
proposed withdrawal.
    (2) Holders of applications for drug products identified in the 
notice of opportunity for hearing may submit written comments on the 
notice of opportunity for hearing issued on the proposed withdrawal of 
the listed drug. If an abbreviated new drug application holder submits 
comments on the notice of opportunity for hearing and a hearing is 
granted, the abbreviated new drug application holder may participate in 
the hearing as a nonparty participant as provided for in Sec. 12.89 of 
this chapter.
    (3) Except as provided in paragraphs (c) and (d) of this section, 
the approval of an abbreviated new drug application for a drug product 
identified in the notice of opportunity for hearing on the withdrawal of 
a listed drug will be withdrawn when the agency has completed the 
withdrawal of approval of the listed drug.
    (c)(1) If the holder of an application for a drug identified in the 
notice of opportunity for hearing has submitted timely comments but does 
not have an opportunity to participate in a hearing because a hearing is 
not requested or is settled, the submitted comments will be considered 
by the agency, which will issue an initial decision. The initial 
decision will respond to the comments, and contain the agency's decision 
whether there are grounds to withdraw approval of the listed drug and of 
the abbreviated new drug applications on which timely comments were 
submitted. The initial decision will be sent to each abbreviated new 
drug application holder that has submitted comments.
    (2) Abbreviated new drug application holders to whom the initial 
decision was sent may, within 30 days of the issuance of the initial 
decision, submit written objections.
    (3) The agency may, at its discretion, hold a limited oral hearing 
to resolve dispositive factual issues that cannot be resolved on the 
basis of written submissions.
    (4) If there are no timely objections to the initial decision, it 
will become final at the expiration of 30 days.
    (5) If timely objections are submitted, they will be reviewed and 
responded to in a final decision.
    (6) The written comments received, the initial decision, the 
evidence relied on in the comments and in the initial decision, the 
objections to the initial decision, and, if a limited oral hearing has 
been held, the transcript of that hearing and any documents submitted 
therein, shall form the record upon which the agency shall make a final 
decision.

[[Page 161]]

    (7) Except as provided in paragraph (d) of this section, any 
abbreviated new drug application whose holder submitted comments on the 
notice of opportunity for hearing shall be withdrawn upon the issuance 
of a final decision concluding that the listed drug should be withdrawn 
for grounds as described in Sec. 314.150(a). The final decision shall be 
in writing and shall constitute final agency action, reviewable in a 
judicial proceeding.
    (8) Documents in the record will be publicly available in accordance 
with Sec. 10.20(j) of this chapter. Documents available for examination 
or copying will be placed on public display in the Dockets Management 
Branch (HFA-305), Food and Drug Administration, room. 1-23, 12420 
Parklawn Dr., Rockville, MD 20857, promptly upon receipt in that office.
    (d) If the agency determines, based upon information submitted by 
the holder of an abbreviated new drug application, that the grounds for 
withdrawal of the listed drug are not applicable to a drug identified in 
the notice of opportunity for hearing, the final decision will state 
that the approval of the abbreviated new drug application for such drug 
is not withdrawn.
[57 FR 17994, Apr. 28, 1992]



Sec. 314.152  Notice of withdrawal of approval of an application or abbreviated application for a new drug.

    If the Food and Drug Administration withdraws approval of an 
application or abbreviated application for a new drug, FDA will publish 
a notice in the Federal Register announcing the withdrawal of approval. 
If the application or abbreviated application was withdrawn for grounds 
described in Sec. 314.150(a) or Sec. 314.151, the notice will announce 
the removal of the drug from the list of approved drugs published under 
section 505(j)(6) of the act and shall satisfy the requirement of 
Sec. 314.162(b).
[57 FR 17994, Apr. 28, 1992]



Sec. 314.153  Suspension of approval of an abbreviated new drug application.

    (a) Suspension of approval. The approval of an abbreviated new drug 
application approved under Sec. 314.105(d) shall be suspended for the 
period stated when:
    (1) The Secretary of the Department of Health and Human Services, 
under the imminent hazard authority of section 505(e) of the act or the 
authority of this paragraph, suspends approval of a listed drug referred 
to in the abbreviated new drug application, for the period of the 
suspension;
    (2) The agency, in the notice described in paragraph (b) of this 
section, or in any subsequent written notice given an abbreviated new 
drug application holder by the agency, concludes that the risk of 
continued marketing and use of the drug is inappropriate, pending 
completion of proceedings to withdraw or suspend approval under 
Sec. 314.151 or paragraph (b) of this section; or
    (3) The agency, under the procedures set forth in paragraph (b) of 
this section, issues a final decision stating the determination that the 
abbreviated application is suspended because the listed drug on which 
the approval of the abbreviated new drug application depends has been 
withdrawn from sale for reasons of safety or effectiveness or has been 
suspended under paragraph (b) of this section. The suspension will take 
effect on the date stated in the decision and will remain in effect 
until the agency determines that the marketing of the drug has resumed 
or that the withdrawal is not for safety or effectiveness reasons.
    (b) Procedures for suspension of abbreviated new drug applications 
when a listed drug is voluntarily withdrawn for safety or effectiveness 
reasons. (1) If a listed drug is voluntarily withdrawn from sale, and 
the agency determines that the withdrawal from sale was for reasons of 
safety or effectiveness, the agency will send each holder of an approved 
abbreviated new drug application that is subject to suspension as a 
result of this determination a copy of the agency's initial decision 
setting forth the reasons for the determination. The initial decision 
will also be placed on file with the Dockets Management Branch (HFA-
305), Food and Drug Administration, room 1-23, 12420 Parklawn Dr., 
Rockville, MD 20857.
    (2) Each abbreviated new drug application holder will have 30 days 
from

[[Page 162]]

the issuance of the initial decision to present, in writing, comments 
and information bearing on the initial decision. If no comments or 
information is received, the initial decision will become final at the 
expiration of 30 days.
    (3) Comments and information received within 30 days of the issuance 
of the initial decision will be considered by the agency and responded 
to in a final decision.
    (4) The agency may, in its discretion, hold a limited oral hearing 
to resolve dispositive factual issues that cannot be resolved on the 
basis of written submissions.
    (5) If the final decision affirms the agency's initial decision that 
the listed drug was withdrawn for reasons of safety or effectiveness, 
the decision will be published in the Federal Register in compliance 
with Sec. 314.152, and will, except as provided in paragraph (b)(6) of 
this section, suspend approval of all abbreviated new drug applications 
identified under paragraph (b)(1) of this section and remove from the 
list the listed drug and any drug whose approval was suspended under 
this paragraph. The notice will satisfy the requirement of 
Sec. 314.162(b). The agency's final decision and copies of materials on 
which it relies will also be filed with the Dockets Management Branch 
(address in paragraph (b)(1) of this section).
    (6) If the agency determines in its final decision that the listed 
drug was withdrawn for reasons of safety or effectiveness but, based 
upon information submitted by the holder of an abbreviated new drug 
application, also determines that the reasons for the withdrawal of the 
listed drug are not relevant to the safety and effectiveness of the drug 
subject to such abbreviated new drug application, the final decision 
will state that the approval of such abbreviated new drug application is 
not suspended.
    (7) Documents in the record will be publicly available in accordance 
with Sec. 10.20(j) of this chapter. Documents available for examination 
or copying will be placed on public display in the Dockets Management 
Branch (address in paragraph (b)(1) of this section) promptly upon 
receipt in that office.
[57 FR 17995, Apr. 28, 1992]



Sec. 314.160  Approval of an application or abbreviated application for which approval was previously refused, suspended, or withdrawn.

    Upon the Food and Drug Administration's own initiative or upon 
request of an applicant, FDA may, on the basis of new data, approve an 
application or abbreviated application which it had previously refused, 
suspended, or withdrawn approval. FDA will publish a notice in the 
Federal Register announcing the approval.
[57 FR 17995, Apr. 28, 1992]



Sec. 314.161  Determination of reasons for voluntary withdrawal of a listed drug.

    (a) A determination whether a listed drug that has been voluntarily 
withdrawn from sale was withdrawn for safety or effectiveness reasons 
may be made by the agency at any time after the drug has been 
voluntarily withdrawn from sale, but must be made:
    (1) Prior to approving an abbreviated new drug application that 
refers to the listed drug;
    (2) Whenever a listed drug is voluntarily withdrawn from sale and 
abbreviated new drug applications that referred to the listed drug have 
been approved; and
    (3) When a person petitions for such a determination under 
Secs. 10.25(a) and 10.30 of this chapter.
    (b) Any person may petition under Secs. 10.25(a) and 10.30 of this 
chapter for a determination whether a listed drug has been voluntarily 
withdrawn for safety or effectiveness reasons. Any such petition must 
contain all evidence available to the petitioner concerning the reason 
that the drug is withdrawn from sale.
    (c) If the agency determines that a listed drug is withdrawn from 
sale for safety or effectiveness reasons, the agency will, except as 
provided in paragraph (d) of this section, publish a notice of the 
determination in the Federal Register.
    (d) If the agency determines under paragraph (a) of this section 
that a listed drug is withdrawn from sale for safety and effectiveness 
reasons and there are approved abbreviated new drug applications that 
are subject to suspension under section 505(j)(5) of the

[[Page 163]]

act, FDA will initiate a proceeding in accordance with Sec. 314.153(b).
    (e) A drug that the agency determines is withdrawn for safety or 
effectiveness reasons will be removed from the list, under Sec. 314.162. 
The drug may be relisted if the agency has evidence that marketing of 
the drug has resumed or that the withdrawal is not for safety or 
effectiveness reasons. A determination that the drug is not withdrawn 
for safety or effectiveness reasons may be made at any time after its 
removal from the list, upon the agency's initiative, or upon the 
submission of a petition under Secs. 10.25(a) and 10.30 of this chapter. 
If the agency determines that the drug is not withdrawn for safety or 
effectiveness reasons, the agency shall publish a notice of this 
determination in the Federal Register. The notice will also announce 
that the drug is relisted, under Sec. 314.162(c). The notice will also 
serve to reinstate approval of all suspended abbreviated new drug 
applications that referred to the listed drug.
[57 FR 17995, Apr. 28, 1992]



Sec. 314.162  Removal of a drug product from the list.

    (a) FDA will remove a previously approved new drug product from the 
list for the period stated when:
    (1) The agency withdraws or suspends approval of a new drug 
application or an abbreviated new drug application under Sec. 314.150(a) 
or Sec. 314.151 or under the imminent hazard authority of section 505(e) 
of the act, for the same period as the withdrawal or suspension of the 
application; or
    (2) The agency, in accordance with the procedures in Sec. 314.153(b) 
or Sec. 314.161, issues a final decision stating that the listed drug 
was withdrawn from sale for safety or effectiveness reasons, or 
suspended under Sec. 314.153(b), until the agency determines that the 
withdrawal from the market has ceased or is not for safety or 
effectiveness reasons.
    (b) FDA will publish in the Federal Register a notice announcing the 
removal of a drug from the list.
    (c) At the end of the period specified in paragraph (a)(1) or (a)(2) 
of this section, FDA will relist a drug that has been removed from the 
list. The agency will publish in the Federal Register a notice 
announcing the relisting of the drug.
[57 FR 17996, Apr. 28, 1992]



Sec. 314.170  Adulteration and misbranding of an approved drug.

    All drugs, including those the Food and Drug Administration 
approves, or provides for certification of, under sections 505, 506, and 
507 of the act and this part, are subject to the adulteration and 
misbranding provisions in sections 501, 502, and 503 of the act. FDA is 
authorized to regulate approved new drugs and approved antibiotic drugs 
by regulations issued through informal rulemaking under sections 501, 
502, and 503 of the act.



               Subpart E--Hearing Procedures for New Drugs

    Source:  50 FR 7493, Feb. 22, 1985, unless otherwise noted. 
Redesignated at 57 FR 17983, Apr. 28, 1992.



Sec. 314.200  Notice of opportunity for hearing; notice of participation and request for hearing; grant or denial of hearing.

    (a) Notice of opportunity for hearing. The Director of the Center 
for Drug Evaluation and Research, Food and Drug Administration, will 
give the applicant, and all other persons who manufacture or distribute 
identical, related, or similar drug products as defined in Sec. 310.6 of 
this chapter, notice and an opportunity for a hearing on the Center's 
proposal to refuse to approve an application or to withdraw the approval 
of an application or abbreviated application under section 505(e) of the 
act. The notice will state the reasons for the action and the proposed 
grounds for the order.
    (1) The notice may be general (that is, simply summarizing in a 
general way the information resulting in the notice) or specific (that 
is, either referring to specific requirements in the statute and 
regulations with which there is a lack of compliance, or providing a 
detailed description and analysis of the specific facts resulting in the 
notice).
    (2) FDA will publish the notice in the Federal Register and will 
state that

[[Page 164]]

the applicant, and other persons subject to the notice under Sec. 310.6, 
who wishes to participate in a hearing, has 30 days after the date of 
publication of the notice to file a written notice of participation and 
request for hearing. The applicant, or other persons subject to the 
notice under Sec. 310.6, who fails to file a written notice of 
participation and request for hearing within 30 days, waives the 
opportunity for a hearing.
    (3) It is the responsibility of every manufacturer and distributor 
of a drug product to review every notice of opportunity for a hearing 
published in the Federal Register to determine whether it covers any 
drug product that person manufactures or distributes. Any person may 
request an opinion of the applicability of a notice to a specific 
product that may be identical, related, or similar to a product listed 
in a notice by writing to the Division of Drug Labeling Compliance (HFD-
310), Center for Drug Evaluation and Research, Food and Drug 
Administration, 5600 Fishers Lane, Rockville, MD 20857. A person shall 
request an opinion within 30 days of the date of publication of the 
notice to be eligible for an opportunity for a hearing under the notice. 
If a person requests an opinion, that person's time for filing an 
appearance and request for a hearing and supporting studies and analyses 
begins on the date the person receives the opinion from FDA.
    (b) FDA will provide the notice of opportunity for a hearing to 
applicants and to other persons subject to the notice under Sec. 310.6, 
as follows:
    (1) To any person who has submitted an application or abbreviated 
application, by delivering the notice in person or by sending it by 
registered or certified mail to the last address shown in the 
application or abbreviated application.
    (2) To any person who has not submitted an application or 
abbreviated application but who is subject to the notice under 
Sec. 310.6 of this chapter, by publication of the notice in the Federal 
Register.
    (c)(1) Notice of participation and request for a hearing, and 
submission of studies and comments. The applicant, or any other person 
subject to the notice under Sec. 310.6, who wishes to participate in a 
hearing, shall file with the Dockets Management Branch (HFA-305), Food 
and Drug Administration, rm. 1-23, 12420 Parklawn Dr., Rockville, MD 
20857, (i) within 30 days after the date of the publication of the 
notice (or of the date of receipt of an opinion requested under 
paragraph (a)(3) of this section) a written notice of participation and 
request for a hearing and (ii) within 60 days after the date of 
publication of the notice, unless a different period of time is 
specified in the notice of opportunity for a hearing, the studies on 
which the person relies to justify a hearing as specified in paragraph 
(d) of this section. The applicant, or other person, may incorporate by 
reference the raw data underlying a study if the data were previously 
submitted to FDA as part of an application, abbreviated application, or 
other report.
    (2) FDA will not consider data or analyses submitted after 60 days 
in determining whether a hearing is warranted unless they are derived 
from well-controlled studies begun before the date of the notice of 
opportunity for hearing and the results of the studies were not 
available within 60 days after the date of publication of the notice. 
Nevertheless, FDA may consider other studies on the basis of a showing 
by the person requesting a hearing of inadvertent omission and hardship. 
The person requesting a hearing shall list in the request for hearing 
all studies in progress, the results of which the person intends later 
to submit in support of the request for a hearing. The person shall 
submit under paragraph (c)(1)(ii) of this section a copy of the complete 
protocol, a list of the participating investigators, and a brief status 
report of the studies.
    (3) Any other interested person who is not subject to the notice of 
opportunity for a hearing may also submit comments on the proposal to 
withdraw approval of the application or abbreviated application. The 
comments are requested to be submitted within the time and under the 
conditions specified in this section.
    (d) The person requesting a hearing is required to submit under 
paragraph (c)(1)(ii) of this section the studies (including all 
protocols and underlying raw data) on which the person relies to

[[Page 165]]

justify a hearing with respect to the drug product. Except, a person who 
requests a hearing on the refusal to approve an application is not 
required to submit additional studies and analyses if the studies upon 
which the person relies have been submitted in the application and in 
the format and containing the summaries required under Sec. 314.50.
    (1) If the grounds for FDA's proposed action concern the 
effectiveness of the drug, each request for hearing is required to be 
supported only by adequate and well-controlled clinical studies meeting 
all of the precise requirements of Sec. 314.126 and, for combination 
drug products, Sec. 300.50, or by other studies not meeting those 
requirements for which a waiver has been previously granted by FDA under 
Sec. 314.126. Each person requesting a hearing shall submit all adequate 
and well-controlled clinical studies on the drug product, including any 
unfavorable analyses, views, or judgments with respect to the studies. 
No other data, information, or studies may be submitted.
    (2) The submission is required to include a factual analysis of all 
the studies submitted. If the grounds for FDA's proposed action concern 
the effectiveness of the drug, the analysis is required to specify how 
each study accords, on a point-by-point basis, with each criterion 
required for an adequate well-controlled clinical investigation 
established under Sec. 314.126 and, if the product is a combination drug 
product, with each of the requirements for a combination drug 
established in Sec. 300.50, or the study is required to be accompanied 
by an appropriate waiver previously granted by FDA. If a study concerns 
a drug or dosage form or condition of use or mode of administration 
other than the one in question, that fact is required to be clearly 
stated. Any study conducted on the final marketed form of the drug 
product is required to be clearly identified.
    (3) Each person requesting a hearing shall submit an analysis of the 
data upon which the person relies, except that the required information 
relating either to safety or to effectiveness may be omitted if the 
notice of opportunity for hearing does not raise any issue with respect 
to that aspect of the drug; information on compliance with Sec. 300.50 
may be omitted if the drug product is not a combination drug product. A 
financial certification or disclosure statement or both as required by 
part 54 of this chapter must accompany all clinical data submitted. FDA 
can most efficiently consider submissions made in the following format.

    I. Safety data.
    A. Animal safety data.
    1. Individual active components.
    a. Controlled studies.
    b. Partially controlled or uncontrolled studies.
    2. Combinations of the individual active components.
    a. Controlled studies.
    b. Partially controlled or uncontrolled studies.
    B. Human safety data.
    1. Individual active components.
    a. Controlled studies.
    b. Partially controlled or uncontrolled studies.
    c. Documented case reports.
    d. Pertinent marketing experiences that may influence a 
determination about the safety of each individual active component.
    2. Combinations of the individual active components.
    a. Controlled studies.
    b. Partially controlled or uncontrolled studies.
    c. Documented case reports.
    d. Pertinent marketing experiences that may influence a 
determination about the safety of each individual active component.
    II. Effectiveness data.
    A. Individual active components: Controlled studies, with an 
analysis showing clearly how each study satisfies, on a point-by-point 
basis, each of the criteria required by Sec. 314.126.
    B. Combinations of individual active components.
    1. Controlled studies with an analysis showing clearly how each 
study satisfies on a point-by-point basis, each of the criteria required 
by Sec. 314.126.
    2. An analysis showing clearly how each requirement of Sec. 300.50 
has been satisfied.
    III. A summary of the data and views setting forth the medical 
rationale and purpose for the drug and its ingredients and the 
scientific basis for the conclusion that the drug and its ingredients 
have been proven safe and/or effective for the intended use. If there is 
an absence of controlled studies in the material submitted or the 
requirements of any element of Sec. 300.50 or Sec. 314.126 have not been 
fully met, that fact is required to be stated clearly and a waiver 
obtained under Sec. 314.126 is required to be submitted.

[[Page 166]]

    IV. A statement signed by the person responsible for such submission 
that it includes in full (or incorporates by reference as permitted in 
Sec. 314.200(c)(2)) all studies and information specified in 
Sec. 314.200(d).

    (Warning: A willfully false statement is a criminal offense, 18 
U.S.C. 1001.)

    (e) Contentions that a drug product is not subject to the new drug 
requirements. A notice of opportunity for a hearing encompasses all 
issues relating to the legal status of each drug product subject to it, 
including identical, related, and similar drug products as defined in 
Sec. 310.6. A notice of appearance and request for a hearing under 
paragraph (c)(1)(i) of this section is required to contain any 
contention that the product is not a new drug because it is generally 
recognized as safe and effective within the meaning of section 201(p) of 
the act, or because it is exempt from part or all of the new drug 
provisions of the act under the exemption for products marketed before 
June 25, 1938, contained in section 201(p) of the act or under section 
107(c) of the Drug Amendments of 1962, or for any other reason. Each 
contention is required to be supported by a submission under paragraph 
(c)(1)(ii) of this section and the Commissioner of Food and Drugs will 
make an administrative determination on each contention. The failure of 
any person subject to a notice of opportunity for a hearing, including 
any person who manufactures or distributes an identical, related, or 
similar drug product as defined in Sec. 310.6, to submit a notice of 
participation and request for hearing or to raise all such contentions 
constitutes a waiver of any contentions not raised.
    (1) A contention that a drug product is generally recognized as safe 
and effective within the meaning of section 201(p) of the act is 
required to be supported by submission of the same quantity and quality 
of scientific evidence that is required to obtain approval of an 
application for the product, unless FDA has waived a requirement for 
effectiveness (under Sec. 314.126) or safety, or both. The submission 
should be in the format and with the analyses required under paragraph 
(d) of this section. A person who fails to submit the required 
scientific evidence required under paragraph (d) waives the contention. 
General recognition of safety and effectiveness shall ordinarily be 
based upon published studies which may be corroborated by unpublished 
studies and other data and information.
    (2) A contention that a drug product is exempt from part or all of 
the new drug provisions of the act under the exemption for products 
marketed before June 25, 1938, contained in section 201(p) of the act, 
or under section 107(c) of the Drug Amendments of 1962, is required to 
be supported by evidence of past and present quantitative formulas, 
labeling, and evidence of marketing. A person who makes such a 
contention should submit the formulas, labeling, and evidence of 
marketing in the following format.

    I. Formulation.
    A. A copy of each pertinent document or record to establish the 
exact quantitative formulation of the drug (both active and inactive 
ingredients) on the date of initial marketing of the drug.
    B. A statement whether such formulation has at any subsequent time 
been changed in any manner. If any such change has been made, the exact 
date, nature, and rationale for each change in formulation, including 
any deletion or change in the concentration of any active ingredient 
and/or inactive ingredient, should be stated, together with a copy of 
each pertinent document or record to establish the date and nature of 
each such change, including, but not limited to, the formula which 
resulted from each such change. If no such change has been made, a copy 
of representative documents or records showing the formula at 
representative points in time should be submitted to support the 
statement.
    II. Labeling.
    A. A copy of each pertinent document or record to establish the 
identity of each item of written, printed, or graphic matter used as 
labeling on the date the drug was initially marketed.
    B. A statement whether such labeling has at any subsequent time been 
discontinued or changed in any manner. If such discontinuance or change 
has been made, the exact date, nature, and rationale for each 
discontinuance or change and a copy of each pertinent document or record 
to establish each such discontinuance or change should be submitted, 
including, but not limited to, the labeling which resulted from each 
such discontinuance or change. If no such discontinuance or change has 
been made, a copy of representative documents or records showing 
labeling at representative points in time should be submitted to support 
the statement.

[[Page 167]]

    III. Marketing.
    A. A copy of each pertinent document or record to establish the 
exact date the drug was initially marketed.
    B. A statement whether such marketing has at any subsequent time 
been discontinued. If such marketing has been discontinued, the exact 
date of each such discontinuance should be submitted, together with a 
copy of each pertinent document or record to establish each such date.
    IV. Verification.
    A statement signed by the person responsible for such submission, 
that all appropriate records have been searched and to the best of that 
person's knowledge and belief it includes a true and accurate 
presentation of the facts.

    (Warning: A willfully false statement is a criminal offense, 18 
U.S.C. 1001.)

    (3) The Food and Drug Administration will not find a drug product, 
including any active ingredient, which is identical, related, or 
similar, as described in Sec. 310.6, to a drug product, including any 
active ingredient for which an application is or at any time has been 
effective or deemed approved, or approved under section 505 of the act, 
to be exempt from part or all of the new drug provisions of the act.
    (4) A contention that a drug product is not a new drug for any other 
reason is required to be supported by submission of the factual records, 
data, and information that are necessary and appropriate to support the 
contention.
    (5) It is the responsibility of every person who manufactures or 
distributes a drug product in reliance upon a ``grandfather'' provision 
of the act to maintain files that contain the data and information 
necessary fully to document and support that status.
    (f) Separation of functions. Separation of functions commences upon 
receipt of a request for hearing. The Director of the Center for Drug 
Evaluation and Research, Food and Drug Administration, will prepare an 
analysis of the request and a proposed order ruling on the matter. The 
analysis and proposed order, the request for hearing, and any proposed 
order denying a hearing and response under paragraph (g) (2) or (3) of 
this section will be submitted to the Office of the Commissioner of Food 
and Drugs for review and decision. When the Center for Drug Evaluation 
and Research recommends denial of a hearing on all issues on which a 
hearing is requested, no representative of the Center will participate 
or advise in the review and decision by the Commissioner. When the 
Center for Drug Evaluation and Research recommends that a hearing be 
granted on one or more issues on which a hearing is requested, 
separation of functions terminates as to those issues, and 
representatives of the Center may participate or advise in the review 
and decision by the Commissioner on those issues. The Commissioner may 
modify the text of the issues, but may not deny a hearing on those 
issues. Separation of functions continues with respect to issues on 
which the Center for Drug Evaluation and Research has recommended denial 
of a hearing. The Commissioner will neither evaluate nor rule on the 
Center's recommendation on such issues and such issues will not be 
included in the notice of hearing. Participants in the hearing may make 
a motion to the presiding officer for the inclusion of any such issue in 
the hearing. The ruling on such a motion is subject to review in 
accordance with Sec. 12.35(b). Failure to so move constitutes a waiver 
of the right to a hearing on such an issue. Separation of functions on 
all issues resumes upon issuance of a notice of hearing. The Office of 
the General Counsel, Department of Health and Human Services, will 
observe the same separation of functions.
    (g) Summary judgment. A person who requests a hearing may not rely 
upon allegations or denials but is required to set forth specific facts 
showing that there is a genuine and substantial issue of fact that 
requires a hearing with respect to a particular drug product specified 
in the request for hearing.
    (1) Where a specific notice of opportunity for hearing (as defined 
in paragraph (a)(1) of this section) is used, the Commissioner will 
enter summary judgment against a person who requests a hearing, making 
findings and conclusions, denying a hearing, if it conclusively appears 
from the face of the data, information, and factual analyses in the 
request for the hearing that there is no genuine and substantial issue 
of fact which precludes the refusal to approve the application or 
abbreviated application or the withdrawal of approval of the application 
or

[[Page 168]]

abbreviated application; for example, no adequate and well-controlled 
clinical investigations meeting each of the precise elements of 
Sec. 314.126 and, for a combination drug product, Sec. 300.50 of this 
chapter, showing effectiveness have been identified. Any order entering 
summary judgment is required to set forth the Commissioner's findings 
and conclusions in detail and is required to specify why each study 
submitted fails to meet the requirements of the statute and regulations 
or why the request for hearing does not raise a genuine and substantial 
issue of fact.
    (2) When following a general notice of opportunity for a hearing (as 
defined in paragraph (a)(1) of this section) the Director of the Center 
for Drug Evaluation and Research concludes that summary judgment against 
a person requesting a hearing should be considered, the Director will 
serve upon the person requesting a hearing by registered mail a proposed 
order denying a hearing. This person has 60 days after receipt of the 
proposed order to respond with sufficient data, information, and 
analyses to demonstrate that there is a genuine and substantial issue of 
fact which justifies a hearing.
    (3) When following a general or specific notice of opportunity for a 
hearing a person requesting a hearing submits data or information of a 
type required by the statute and regulations, and the Director of the 
Center for Drug Evaluation and Research concludes that summary judgment 
against the person should be considered, the Director will serve upon 
the person by registered mail a proposed order denying a hearing. The 
person has 60 days after receipt of the proposed order to respond with 
sufficient data, information, and analyses to demonstrate that there is 
a genuine and substantial issue of fact which justifies a hearing.
    (4) If review of the data, information, and analyses submitted show 
that the grounds cited in the notice are not valid, for example, that 
substantial evidence of effectiveness exists, the Commissioner will 
enter summary judgment for the person requesting the hearing, and 
rescind the notice of opportunity for hearing.
    (5) If the Commissioner grants a hearing, it will begin within 90 
days after the expiration of the time for requesting the hearing unless 
the parties otherwise agree in the case of denial of approval, and as 
soon as practicable in the case of withdrawal of approval.
    (6) The Commissioner will grant a hearing if there exists a genuine 
and substantial issue of fact or if the Commissioner concludes that a 
hearing would otherwise be in the public interest.
    (7) If the manufacturer or distributor of an identical, related, or 
similar drug product requests and is granted a hearing, the hearing may 
consider whether the product is in fact identical, related, or similar 
to the drug product named in the notice of opportunity for a hearing.
    (8) A request for a hearing, and any subsequent grant or denial of a 
hearing, applies only to the drug products named in such documents.
    (h) FDA will issue a notice withdrawing approval and declaring all 
products unlawful for drug products subject to a notice of opportunity 
for a hearing, including any identical, related, or similar drug product 
under Sec. 310.6, for which an opportunity for a hearing is waived or 
for which a hearing is denied. The Commissioner may defer or stay the 
action pending a ruling on any related request for a hearing or pending 
any related hearing or other administrative or judicial proceeding.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0001)

[50 FR 7493, Feb. 22, 1985; 50 FR 14212, Apr. 11, 1985, as amended at 50 
FR 21238, May 23, 1985; 55 FR 11580, Mar. 29, 1990; 57 FR 17996, Apr. 
28, 1992; 59 FR 14364, Mar. 28, 1994; 63 FR 5252, Feb. 2, 1998]

    Effective Date Note: At 63 FR 5252, Feb. 2, 1998, Sec. 314.200 was 
amended in paragraph (d)(3) by adding a new sentence after the first 
sentence, effective Feb. 2, 1999.



Sec. 314.201  Procedure for hearings.

    Parts 10 through 16 apply to hearings relating to new drugs under 
section 505 (d) and (e) of the act.



Sec. 314.235  Judicial review.

    (a) The Commissioner of Food and Drugs will certify the transcript 
and

[[Page 169]]

record. In any case in which the Commissioner enters an order without a 
hearing under Sec. 314.200(g), the record certified by the Commissioner 
is required to include the requests for hearing together with the data 
and information submitted and the Commissioner's findings and 
conclusion.
    (b) A manufacturer or distributor of an identical, related, or 
similar drug product under Sec. 310.6 may seek judicial review of an 
order withdrawing approval of a new drug application, whether or not a 
hearing has been held, in a United States court of appeals under section 
505(h) of the act.



          Subpart F--Administrative Procedures for Antibiotics

    Source:  50 FR 7493, Feb. 22, 1985, unless otherwise noted. 
Redesignated at 57 FR 17983, Apr. 28, 1992.



Sec. 314.300  Procedure for the issuance, amendment, or repeal of regulations.

    (a) The procedures in part 10 apply to the issuance, amendment, or 
repeal of regulations under section 507 of the act.
    (b)(1) The Commissioner of Food and Drugs, on his or her own 
initiative or on the application or request of any interested person, 
may publish in the Federal Register a notice of proposed rulemaking and 
order to issue, amend, or repeal any regulation contemplated by section 
507 of the act. The notice and order may be general (that is, simply 
summarizing in a general way the information resulting in the notice and 
order) or specific (that is, either referring to specific requirements 
in the statute and regulations with which there is a lack of compliance, 
or providing a detailed description and analysis of the specific facts 
resulting in the notice and order).
    (2) The Food and Drug Administration will give interested persons an 
opportunity to submit written comments and to request an informal 
conference on the proposal, unless the notice and opportunity for 
comment and informal conference have already been provided in connection 
with the announcement of the reports of the National Academy of 
Sciences/National Research Council, Drug Efficacy Study Group, to 
persons who will be adversely affected, or as provided in Secs. 10.40(e) 
and 12.20(c)(2). A person is required to request an informal conference 
within 30 days of the notice of proposed rulemaking unless otherwise 
specified in the notice. If an informal conference is requested and 
granted, those persons participating in the conference may submit 
comments, within 30 days of the conference, unless otherwise specified 
in the proposal.
    (3) It is the responsibility of every manufacturer and distributor 
of an antibiotic drug product to review every proposal published in the 
Federal Register to determine whether it covers any drug product that 
person manufactures or distributes.
    (4) After considering the written comments, the results of any 
conference, and the data available, the Commissioner will publish an 
order in the Federal Register acting on the proposal, with an 
opportunity for any person who will be adversely affected to file 
objections, to request a hearing, and to show reasonable grounds for the 
hearing. Any person who wishes to participate in a hearing, shall file 
with the Dockets Management Branch (HFA-305), Food and Drug 
Administration, rm. 1-23, 12420 Parklawn Dr., Rockville, MD 20857, (i) 
within 30 days after the date of the publication of the order a written 
notice of participation and request for a hearing and (ii) within 60 
days after the date of publication of the order, unless a different 
period of time is specified in the order, the studies on which the 
person relies to justify a hearing as specified in paragraph (b)(6) of 
this section. The person may incorporate by reference the raw data 
underlying a study if the data were previously submitted to FDA as part 
of an application or other report.
    (5) FDA will not consider data or analysis submitted after 60 days 
in determining whether a hearing is warranted unless they are derived 
from well-controlled studies begun before the date of the order and the 
results of the studies were not available within 60 days after the date 
of publication of the order. Nevertheless, FDA may consider other 
studies on the basis of a showing by the person requesting a hearing of 
inadvertent omission and

[[Page 170]]

hardship. The person requesting a hearing shall list in the request for 
hearing all studies in progress, the results of which the person intends 
later to submit in support of the request for hearing. The person shall 
submit under paragraph (b)(4)(ii) of this section a copy of the complete 
protocol, a list of the participating investigators, and a brief status 
report of the studies.
    (6) The person requesting a hearing is required to submit as 
required under Sec. 314.200(c)(1)(ii) the studies (including all 
protocols and underlying raw data) on which the person relies to justify 
a hearing with respect to the drug product. A financial certification or 
disclosure statement or both as required by part 54 of this chapter must 
accompany all clinical data submitted with the request for hearing. 
Except, a person who requests a hearing on a proposal is not required to 
submit additional studies and analyses if the studies upon which the 
person relies have been submitted in an application and in the format 
and containing the summaries required under Sec. 314.50.
    (i) If the grounds for FDA proposed action concern the effectiveness 
of the drug, each request for hearing is required to be supported only 
by adequate and well-controlled clinical studies meeting all of the 
precise requirements of Sec. 314.126 and, for combination drug products, 
Sec. 300.50, or by other studies not meeting those requirements for 
which a waiver has been previously granted by FDA under Sec. 314.126. 
Each person requesting a hearing shall submit all adequate and well-
controlled clinical studies on the drug product, any unfavorable 
analyses, views, or judgments with respect to the studies. No other 
data, information, or studies may be submitted.
    (ii) The submission is required to include a factual analysis of all 
the studies submitted. If the grounds for FDA proposed action concern 
the effectiveness of the drug, the analysis is required to specify how 
each study accords, on a point-by-point basis, with each criterion 
required for an adequate well-controlled clinical investigation 
established under Sec. 314.126 and, if the product is a combination drug 
product, with each of the requirements for a combination drug 
established in Sec. 300.50, or the study is required to be accompanied 
by an appropriate waiver previously granted by FDA. If a study concerns 
a drug entity or dosage form or condition of use or mode of 
administration other than the one in question, that fact is required to 
be clearly stated. Any study conducted on the final marketed form of the 
drug product is required to be clearly identified.
    (iii) Each person requesting a hearing shall submit an analysis of 
the data upon which the person relies, except that the required 
information relating either to safety or to effectiveness may be omitted 
if the notice of opportunity for hearing does not raise any issue with 
respect to that aspect of the drug; information on compliance with 
Sec. 300.50 may be omitted if the drug product is not a combination drug 
product. FDA can most efficiently consider submissions made in the 
following format.

    I. Safety data.
    A. Animal safety data.
    1. Individual active components.
    a. Controlled studies.
    b. Partially controlled or uncontrolled studies.
    2. Combinations of the individual active components.
    a. Controlled studies.
    b. Partially controlled or uncontrolled studies.
    B. Human safety data.
    1. Individual active components.
    a. Controlled studies.
    b. Partially controlled or uncontrolled studies.
    c. Documented case reports.
    d. Pertinent marketing experiences that may influence a 
determination about the safety of each individual active component.
    2. Combinations of the individual active components.
    a. Controlled studies.
    b. Partially controlled or uncontrolled studies.
    c. Documented case reports.
    d. Pertinent marketing experiences that may influence a 
determination about the safety of each individual active component.
    II. Effectiveness data.
    A. Individual active components: Controlled studies, with an 
analysis showing clearly how each study satisfies, on a point-by-point 
basis, each of the criteria required by Sec. 314.126.
    B. Combinations of individual active components.
    1. Controlled studies with an analysis showing clearly how each 
study satisfies on

[[Page 171]]

a point-by-point basis, each of the criteria required by Sec. 314.126.
    2. An analysis showing clearly how each requirement of Sec. 300.50 
has been satisfied.
    III. A summary of the data and views setting forth the medical 
rationale and purpose for the drug and its ingredients and the 
scientific basis for the conclusion that the drug and its ingredients 
have been proven safe and/or effective for the intended use. If there is 
an absence of controlled studies in the material submitted or the 
requirements of any element of Sec. 300.50 or Sec. 314.126 have not been 
fully met, that fact is required to be stated clearly and a waiver 
obtained under Sec. 314.126 is required to be submitted.
    IV. A statement signed by the person responsible for such submission 
that it includes in full (or incorporates by reference as permitted in 
Sec. 314.200(c)(2)) all studies and information specified in 
Sec. 314.200(d).

    (Warning: A willfully false statement is a criminal offense, 18 
U.S.C. 1001.)

    (7) Separation of functions. Separation of functions commences upon 
receipt of a request for hearing. The Director of the Center for Drug 
Evaluation and Research will prepare an analysis of the request and a 
proposed order ruling on the matter. The analysis and proposed order, 
the request for hearing, and any proposed order denying a hearing and 
response under paragraph (b)(8) (ii) or (iii) of this section will be 
submitted to the Office of the Commissioner for review and decision. 
When the Center for Drug Evaluation and Research recommends denial of a 
hearing on all issues on which a hearing is requested, no representative 
of the Center will participate or advise in the review and decision by 
the Commissioner. When the Center for Drug Evaluation and Research 
recommends that a hearing be granted on one or more issues on which a 
hearing is requested, separation of functions terminates as to those 
issues, and representatives of the Center may participate or advise in 
the review and decision by the Commissioner on those issues. The 
Commissioner may modify the text of the issues, but may not deny a 
hearing on those issues. Separation of functions continues with respect 
to issues on which the Center for Drug Evaluatioon and Research has 
recommended denial of a hearing. The Commissioner will neither evaluate 
nor rule on the Center's recommendation on such issues and such issues 
will not be included in the notice of hearing. Participants in the 
hearing may make a motion to the presiding officer for the inclusion of 
any such issue in the hearing. The ruling on such a motion is subject to 
review in accordance with Sec. 12.35(b). Failure to so move constitutes 
a waiver of the right to a hearing on such an issue. Separation of 
functions on all issues resumes upon issuance of a notice of hearing. 
The Office of the General Counsel, Department of Health and Human 
Services, will observe the same separation of functions.
    (8) Summary judgment. A person who requests a hearing may not rely 
upon allegations or denials but is required to set forth specific facts 
showing that there is a genuine and substantial issue of fact that 
requires a hearing with respect to a particular drug product specified 
in the request for hearing.
    (i) Where a specific notice of opportunity for hearing (as defined 
in paragraph (b)(1) of this section) is used, the Commissioner will 
enter summary judgment against a person who requests a hearing, making 
findings and conclusions, denying a hearing, if it conclusively appears 
from the face of the data, information, and factual analyses in the 
request for the hearing that there is no genuine and substantial issue 
of fact which precludes the refusal to approve the application or the 
withdrawal of approval of the application; for example, no adequate and 
well-controlled clinical investigations meeting each of the precise 
elements of Sec. 314.126 and, for a combination drug product, 
Sec. 300.50, showing effectiveness have been identified. Any order 
entering summary judgment is required to set forth the Commissioner's 
findings and conclusions in detail and is required to specify why each 
study submitted fails to meet the requirements of the statute and 
regulations or why the request for hearing does not raise a genuine and 
substantial issue of fact.
    (ii) When following a general notice of opportunity for a hearing 
(as defined in paragraph (b)(1) of this section) the Director of the 
Center for Drug Evaluation and Research concludes that summary judgment 
against a person requesting a hearing should be considered, the Director 
will serve upon the

[[Page 172]]

person requesting a hearing by registered mail a proposed order denying 
a hearing. This person has 60 days after receipt of the proposed order 
to respond with sufficient data, information, and analyses to 
demonstrate that there is a genuine and substantial issue of fact which 
justifies a hearing.
    (iii) When following a general or specific notice of opportunity for 
a hearing a person requesting a hearing submits data or information of a 
type required by the statute and regulations, and the Director of the 
Center for Drug Evaluation and Research concludes that summary judgment 
against the person should be considered, the Director will serve upon 
the person by registered mail a proposed order denying a hearing. The 
person has 60 days after receipt of the proposed order to respond with 
sufficient data, information, and analyses to demonstrate that there is 
a genuine and substantial issue of fact which justifies a hearing.
    (iv) If review of the data, information, and analyses submitted show 
that the basis for the order is not valid, for example, that substantial 
evidence of effectiveness exists, the Commissioner will enter summary 
judgment for the person requesting the hearing, and revoke the order. If 
a hearing is not requested, the order will become effective as 
published.
    (v) If the Commissioner grants a hearing, it will be conducted under 
part 12.
    (vi) The Commissioner will grant a hearing if there exists a genuine 
and substantial issue of fact or if the Commissioner concludes that a 
hearing would otherwise be in the public interest.
    (9) The repeal of any regulation constitutes a revocation of all 
outstanding certificates based upon such regulation. However, the 
Commissioner may, in his or her discretion, defer or stay such action 
pending a ruling on any related request for a hearing or pending any 
related hearing or other administrative or judicial proceeding.
    (c) Whenever any interested person submits an application or request 
under section 507 of the act and part 314 and FDA sends the person an 
approvable letter under Sec. 314.110 or a not approvable letter under 
Sec. 314.120, the person may file a petition proposing the issuance, 
amendment, or repeal of the regulation under the provisions of section 
507(f) of the act and part 10. The Commissioner shall cause the 
regulation proposed in the petition to be published in the Federal 
Register within 60 days of the receipt of an acceptable petition and 
further proceedings shall be in accord with the provisions of sections 
507(f) and 701 (f) and (g) of the act and part 10.
    (d) (1) FDA will not promulgate a regulation providing for the 
certification of any batch of any drug composed wholly or in part of any 
kind of penicillin, streptomycin, chlortetracycline, chloramphenicol, 
bacitracin, or any other antibiotic drug, or any derivative thereof, 
intended for human use and no existing regulation will be continued in 
effect unless it is established by substantial evidence that the drug 
will have such characteristics of identity, strength, quality, and 
purity necessary to adequately ensure safety and efficacy of use. 
``Substantial evidence'' has been defined by Congress to mean ``evidence 
consisting of adequate and well-controlled investigations, including 
clinical investigations, by experts qualified by scientific training and 
experience to evaluate the effectiveness of the drug involved, on the 
basis of which it could fairly and responsibly be concluded by such 
experts that the drug will have the effect it purports or is represented 
to have under the conditions prescribed, recommended, or suggested in 
the labeling or proposed labeling thereof.'' This definition is made 
applicable to a number of antibiotic drugs by section 507(h) of the act 
and it is the test of efficacy that FDA will apply in promulgating, 
amending, or repealing regulations for all antibiotics under section 
507(a) of the act as well.
    (2) The scientific essentials of an adequate and well-controlled 
clinical investigation are described in Sec. 314.126.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0001)

[50 FR 7493, Feb. 22, 1985; 50 FR 14212, Apr. 11, 1985, as amended at 50 
FR 21238, May 23, 1985; 55 FR 11580, Mar. 29, 1990; 59 FR 14365, Mar. 
28, 1994; 63 FR 5252, Feb. 2, 1998]

[[Page 173]]


    Effective Date Note: At 63 FR 5252, Feb. 2, 1998, Sec. 314.300 was 
amended in the introductory text of paragraph (b)(6) by adding a new 
sentence after the first sentence, effective Feb. 2, 1999.



                   Subpart G--Miscellaneous Provisions

    Source:  50 FR 7493, Feb. 22, 1985, unless otherwise noted. 
Redesignated at 57 FR 17983, Apr. 28, 1992.



Sec. 314.410  Imports and exports of new drugs and antibiotics.

    (a) Imports. (1) A new drug or an antibiotic may be imported into 
the United States if: (i) It is the subject of an approved application 
under this part or, in the case of an antibiotic not exempt from 
certification under part 433, it is also certified or released; or (ii) 
it complies with the regulations pertaining to investigational new drugs 
under part 312; and it complies with the general regulations pertaining 
to imports under subpart E of part 1.
    (2) A drug substance intended for use in the manufacture, 
processing, or repacking of a new drug may be imported into the United 
States if it complies with the labeling exemption in Sec. 201.122 
pertaining to shipments of drug substances in domestic commerce.
    (b) Exports. (1) A new drug or an antibiotic may be exported if it 
is the subject of an approved application under this part, and, in the 
case of an antibiotic, it is certified or released, or it complies with 
the regulations pertaining to investigational new drugs under part 312.
    (2) A new drug substance that is covered by an application approved 
under this part for use in the manufacture of an approved drug product 
may be exported by the applicant or any person listed as a supplier in 
the approved application, provided the drug substance intended for 
export meets the specifications of, and is shipped with a copy of the 
labeling required for, the approved drug product.
    (3) An antibiotic drug product or drug substance that is subject to 
certification under section 507 of the act, but which has not been 
certified or released, may be exported under section 801(e) of the act 
if it meets the following conditions:
    (i) It meets the specifications of the foreign purchaser;
    (ii) It is not in conflict with the laws of the country to which it 
is intended for export;
    (iii) It is labeled on the outside of the shipping package that it 
is intended for export; and
    (iv) It is not sold or offered for sale in the United States.



Sec. 314.420  Drug master files.

    (a) A drug master file is a submission of information to the Food 
and Drug Administration by a person (the drug master file holder) who 
intends it to be used for one of the following purposes: To permit the 
holder to incorporate the information by reference when the holder 
submits an investigational new drug application under part 312 or 
submits an application or an abbreviated application or an amendment or 
supplement to them under this part, or to permit the holder to authorize 
other persons to rely on the information to support a submission to FDA 
without the holder having to disclose the information to the person. FDA 
ordinarily neither independently reviews drug master files nor approves 
or disapproves submissions to a drug master file. Instead, the agency 
customarily reviews the information only in the context of an 
application under part 312 or this part. A drug master file may contain 
information of the kind required for any submission to the agency, 
including information about the following:
    (1) Manufacturing site, facilities, operating procedures, and 
personnel (because an FDA on-site inspection of a foreign drug 
manufacturing facility presents unique problems of planning and travel 
not presented by an inspection of a domestic manufacturing facility, 
this information is only recommended for foreign manufacturing 
establishments);
    (2) Drug substance, drug substance intermediate, and materials used 
in their preparation, or drug product;
    (3) Packaging materials;
    (4) Excipient, colorant, flavor, essence, or materials used in their 
preparation;

[[Page 174]]

    (5) FDA-accepted reference information. (A person wishing to submit 
information and supporting data in a drug master file (DMF) that is not 
covered by Types I through IV DMF's must first submit a letter of intent 
to the Drug Master File Staff, Food and Drug Administration, 12420 
Parklawn Dr., Rm. 2-14, Rockville, MD 20852. FDA will then contact the 
person to discuss the proposed submission.)
    (b) An investigational new drug application or an application, 
abbreviated application, amendment, or supplement may incorporate by 
reference all or part of the contents of any drug master file in support 
of the submission if the holder authorizes the incorporation in writing. 
Each incorporation by reference is required to describe the incorporated 
material by name, reference number, volume, and page number of the drug 
master file.
    (c) A drug master file is required to be submitted in two copies. 
The agency has prepared under Sec. 10.90(b) a guideline that provides 
information about how to prepare a well-organized drug master file. If 
the drug master file holder adds, changes, or deletes any information in 
the file, the holder shall notify in writing, each person authorized to 
reference that information. Any addition, change, or deletion of 
information in a drug master file (except the list required under 
paragraph (d) of this section) is required to be submitted in two copies 
and to describe by name, reference number, volume, and page number the 
information affected in the drug master file.
    (d) The drug master file is required to contain a complete list of 
each person currently authorized to incorporate by reference any 
information in the file, identifying by name, reference number, volume, 
and page number the information that each person is authorized to 
incorporate. If the holder restricts the authorization to particular 
drug products, the list is required to include the name of each drug 
product and the application number, if known, to which the authorization 
applies.
    (e) The public availability of data and information in a drug master 
file, including the availability of data and information in the file to 
a person authorized to reference the file, is determined under part 20 
and Sec. 314.430.

(Collection of information requirements approved by the Office of 
Management and Budget under control number 0910-0001)

[50 FR 7493, Feb. 22, 1985, as amended at 50 FR 21238, May 23, 1985; 53 
FR 33122, Aug. 30, 1988; 55 FR 28380, July 11, 1990]



Sec. 314.430  Availability for public disclosure of data and information in an application or abbreviated application.

    (a) The Food and Drug Administration will determine the public 
availability of any part of an application or abbreviated application 
under this section and part 20 of this chapter. For purposes of this 
section, the application or abbreviated application includes all data 
and information submitted with or incorporated by reference in the 
application or abbreviated application, including investigational new 
drug applications, drug master files under Sec. 314.420, supplements 
submitted under Sec. 314.70 or Sec. 314.97, reports under Sec. 314.80 or 
Sec. 314.98, and other submissions. For purposes of this section, safety 
and effectiveness data include all studies and tests of a drug on 
animals and humans and all studies and tests of the drug for identity, 
stability, purity, potency, and bioavailability.
    (b) FDA will not publicly disclose the existence of an application 
or abbreviated application before an approvable letter is sent to the 
applicant under Sec. 314.110, unless the existence of the application or 
abbreviated application has been previously publicly disclosed or 
acknowledged. The Center for Drug Evaluation and Research will maintain 
and make available for public disclosure a list of applications or 
abbreviated applications for which the agency has sent an approvable 
letter to the applicant.
    (c) If the existence of an unapproved application or abbreviated 
application has not been publicly disclosed or acknowledged, no data or 
information in the application or abbreviated application is available 
for public disclosure.
    (d)(1) If the existence of an application or abbreviated application 
has

[[Page 175]]

been publicly disclosed or acknowledged before the agency sends an 
approval letter to the applicant, no data or information contained in 
the application or abbreviated application is available for public 
disclosure before the agency sends an approval letter, but the 
Commissioner may, in his or her discretion, disclose a summary of 
selected portions of the safety and effectiveness data that are 
appropriate for public consideration of a specific pending issue; for 
example, for consideration of an open session of an FDA advisory 
committee.
    (2) Notwithstanding paragraph (d)(1) of this section, FDA will make 
available to the public upon request the information in the 
investigational new drug application that was required to be filed in 
Docket Number 95S-0158 in the Dockets Management Branch (HFA-305), Food 
and Drug Administration, 12420 Parklawn Dr., rm. 1-23, Rockville, MD 
20857, for investigations involving an exception from informed consent 
under Sec. 50.24 of this chapter. Persons wishing to request this 
information shall submit a request under the Freedom of Information Act.
    (e) After FDA sends an approval letter to the applicant, the 
following data and information in the application or abbreviated 
application are immediately available for public disclosure, unless the 
applicant shows that extraordinary circumstances exist. A list of 
approved applications and abbreviated applications, entitled ``Approved 
Drug Products with Therapeutic Equivalence Evaluations,'' is available 
from the Government Printing Office, Washington, DC 20402. This list is 
updated monthly.
    (1) [Reserved]
    (2) If the application applies to a new drug, all safety and 
effectiveness data previously disclosed to the public as set forth in 
Sec. 20.81 and a summary or summaries of the safety and effectiveness 
data and information submitted with or incorporated by reference in the 
application. The summaries do not constitute the full reports of 
investigations under section 505(b)(1) of the act (21 U.S.C. 355(b)(1)) 
on which the safety or effectiveness of the drug may be approved. The 
summaries consist of the following:
    (i) For an application approved before July 1, 1975, internal agency 
records that describe safety and effectiveness data and information, for 
example, a summary of the basis for approval or internal reviews of the 
data and information, after deletion of the following:
    (a) Names and any information that would identify patients or test 
subjects or investigators.
    (b) Any inappropriate gratuitous comments unnecessary to an 
objective analysis of the data and information.
    (ii) For an application approved on or after July 1, 1975, a Summary 
Basis of Approval (SBA) document that contains a summary of the safety 
and effectiveness data and information evaluated by FDA during the drug 
approval process. The SBA is prepared in one of the following ways:
    (a) Before approval of the application, the applicant may prepare a 
draft SBA which the Center for Drug Evaluation and Research will review 
and may revise. The draft may be submitted with the application or as an 
amendment.
    (b) The Center for Drug Evaluation and Research may prepare the SBA.
    (3) A protocol for a test or study, unless it is shown to fall 
within the exemption established for trade secrets and confidential 
commercial information in Sec. 20.61.
    (4) Adverse reaction reports, product experience reports, consumer 
complaints, and other similar data and information after deletion of the 
following:
    (i) Names and any information that would identify the person using 
the product.
    (ii) Names and any information that would identify any third party 
involved with the report, such as a physician or hospital or other 
institution.
    (5) A list of all active ingredients and any inactive ingredients 
previously disclosed to the public as set forth in Sec. 20.81.
    (6) An assay method or other analytical method, unless it serves no 
regulatory or compliance purpose and is shown to fall within the 
exemption established for trade secrets and confidential commercial 
information in Sec. 20.61.

[[Page 176]]

    (7) All correspondence and written summaries of oral discussions 
between FDA and the applicant relating to the application, under the 
provisions of part 20.
    (8) All records showing the testing of an action on a particular lot 
of a certifiable antibiotic by FDA.
    (f) All safety and effectiveness data and information which have 
been submitted in an application and which have not previously been 
disclosed to the public are available to the public, upon request, at 
the time any one of the following events occurs unless extraordinary 
circumstances are shown:
    (1) No work is being or will be undertaken to have the application 
approved.
    (2) A final determination is made that the application is not 
approvable and all legal appeals have been exhausted.
    (3) Approval of the application is withdrawn and all legal appeals 
have been exhausted.
    (4) A final determination has been made that the drug is not a new 
drug.
    (5) For applications submitted under section 505(b) of the act, the 
effective date of the approval of the first abbreviated application 
submitted under section 505(j) of the act which refers to such drug, or 
the date on which the approval of an abbreviated application under 
section 505(j) of the act which refers to such drug could be made 
effective if such an abbreviated application had been submitted.
    (6) For applications or abbreviated applications submitted under 
sections 505(j), 506, and 507 of the act, when FDA sends an approval 
letter to the applicant.
    (g) The following data and information in an application or 
abbreviated application are not available for public disclosure unless 
they have been previously disclosed to the public as set forth in 
Sec. 20.81 of this chapter or they relate to a product or ingredient 
that has been abandoned and they do not represent a trade secret or 
confidential commercial or financial information under Sec. 20.61 of 
this chapter:
    (1) Manufacturing methods or processes, including quality control 
procedures.
    (2) Production, sales distribution, and similar data and 
information, except that any compilation of that data and information 
aggregated and prepared in a way that does not reveal data or 
information which is not available for public disclosure under this 
provision is available for public disclosure.
    (3) Quantitative or semiquantitative formulas.
    (h) The compilations of information specified in Sec. 20.117 are 
available for public disclosure.
[50 FR 7493, Feb. 22, 1985, as amended at 50 FR 21238, May 23, 1985; 55 
FR 11580, Mar. 29, 1990; 57 FR 17996, Apr. 28, 1992; 61 FR 51530, Oct. 
2, 1996]



Sec. 314.440  Addresses for applications and abbreviated applications.

    (a) Applicants shall send applications, abbreviated applications, 
and other correspondence relating to matters covered by this part, 
except for products listed in paragraph (b) of this section, to the 
Center for Drug Evaluation and Research, Food and Drug Administration, 
5600 Fishers Lane, Rockville, MD 20857, and directed to the appropriate 
office identified below:
    (1) Except as provided in paragraph (a)(4) of this section, an 
application under Sec. 314.50 or Sec. 314.54 submitted for filing should 
be directed to the Document and Records Section, 12420 Parklawn Dr., 
Rockville, MD 20852. Applicants may obtain folders for binding 
applications from the Consolidated Forms and Publications Distribution 
Center, Washington Commerce Center, 3222 Hubbard Rd., Landover, MD 
20785. After FDA has filed the application, the agency will inform the 
applicant which division is responsible for the application. Amendments, 
supplements, resubmissions, requests for waivers, and other 
correspondence about an application that has been filed should be 
directed to the appropriate division.
    (2) Except as provided in paragraph (a)(4) of this section, an 
abbreviated application under Sec. 314.94, and amendments, supplements, 
and resubmissions should be directed to the Office of Generic Drugs 
(HFD-600), Center for Drug Evaluation and Research, Food and Drug 
Administration, 5600 Fishers Lane, Rockville, MD 20857. Items sent

[[Page 177]]

by parcel post or overnight courier service should be directed to the 
Office of Generic Drugs (HFD-600), Center for Drug Evaluation and 
Research, Food and Drug Administration, Metro Park North II, 7500 
Standish Place, rm. 150, Rockville, MD 20855. Correspondence not 
associated with an application should be addressed specifically to the 
intended office or division and to the person as follows: Center for 
Drug Evaluation and Research, Food and Drug Administration, Attn: 
[insert name of person], MPN II, HFD-[insert mail code of office or 
division], 5600 Fishers Lane, Rockville, MD 20857. The mail code for the 
Office of Generic Drugs is HFD-600, the mail code for the Division of 
Chemistry is HFD-630, and the mail code for the Division of 
Bioequivalence is HFD-650.
    (3) A request for an opportunity for a hearing under Sec. 314.110 or 
Sec. 314.120 on the question of whether there are grounds for denying 
approval of an application, except an application under paragraph (b) of 
this section, should be directed to the Associate Director for Policy 
(HFD-5).
    (4) The field copy of an application, an abbreviated application, 
amendments, supplements, resubmissions, requests for waivers, and other 
correspondence about an application and an abbreviated application shall 
be sent to the applicant's home FDA district office, except that a 
foreign applicant shall send the field copy to the appropriate address 
identified in paragraphs (a)(1) and (a)(2) of this section.
    (b) Applicants shall send applications and other correspondence 
relating to matters covered by this part for the drug products listed 
below to the Division of Product Certification (HFB-240), Center for 
Biologics Evaluation and Research, Food and Drug Administration, 8800 
Rockville Pike, Bethesda, MD 20892, except applicants shall send a 
request for an opportunity for a hearing under Sec. 314.110 or 
Sec. 314.120 on the question of whether there are grounds for denying 
approval of an application to the Director, Center for Biologics 
Evaluation and Research (HFB-1), at the same address.
    (1) Ingredients packaged together with containers intended for the 
collection, processing, or storage of blood and blood components.
    (2) Urokinase products.
    (3) Plasma volume expanders and hydroxyethyl starch for 
leukapheresis.
[50 FR 7493, Feb. 22, 1985, as amended at 50 FR 21238, May 23, 1985; 55 
FR 11581, Mar. 29, 1990; 57 FR 17997, Apr. 28, 1992; 58 FR 47352, Sept. 
8, 1993; 62 FR 43639, Aug. 15, 1997]



Sec. 314.445  Guidelines.

    (a) The Food and Drug Administration prepares guidelines under 
Sec. 10.90(b) to help persons comply with requirements in this part.
    (b) The Center for Drug Evaluation and Research will maintain and 
make publicly available a list of guidelines that apply to the Center's 
regulations. The list states how a person can obtain a copy of each 
guideline. A request for a copy of the list should be directed to the 
CDER Executive Secretariat Staff (HFD-8), Center for Drug Evaluation and 
Research, Food and Drug Administration, 5600 Fishers Lane, Rockville, MD 
20857.
[50 FR 7493, Feb. 22, 1985, as amended at 55 FR 11581, Mar. 29, 1990; 56 
FR 3776, Jan. 31, 1991]



    Subpart H--Accelerated Approval of New Drugs for Serious or Life-
                          Threatening Illnesses

    Source:  57 FR 58958, Dec. 11, 1992, unless otherwise noted.



Sec. 314.500  Scope.

    This subpart applies to certain new drug and antibiotic products 
that have been studied for their safety and effectiveness in treating 
serious or life-threatening illnesses and that provide meaningful 
therapeutic benefit to patients over existing treatments (e.g., ability 
to treat patients unresponsive to, or intolerant of, available therapy, 
or improved patient response over available therapy).



Sec. 314.510  Approval based on a surrogate endpoint or on an effect on a clinical endpoint other than survival or irreversible morbidity.

    FDA may grant marketing approval for a new drug product on the basis 
of adequate and well-controlled clinical

[[Page 178]]

trials establishing that the drug product has an effect on a surrogate 
endpoint that is reasonably likely, based on epidemiologic, therapeutic, 
pathophysiologic, or other evidence, to predict clinical benefit or on 
the basis of an effect on a clinical endpoint other than survival or 
irreversible morbidity. Approval under this section will be subject to 
the requirement that the applicant study the drug further, to verify and 
describe its clinical benefit, where there is uncertainty as to the 
relation of the surrogate endpoint to clinical benefit, or of the 
observed clinical benefit to ultimate outcome. Postmarketing studies 
would usually be studies already underway. When required to be 
conducted, such studies must also be adequate and well-controlled. The 
applicant shall carry out any such studies with due diligence.



Sec. 314.520  Approval with restrictions to assure safe use.

    (a) If FDA concludes that a drug product shown to be effective can 
be safely used only if distribution or use is restricted, FDA will 
require such postmarketing restrictions as are needed to assure safe use 
of the drug product, such as:
    (1) Distribution restricted to certain facilities or physicians with 
special training or experience; or
    (2) Distribution conditioned on the performance of specified medical 
procedures.
    (b) The limitations imposed will be commensurate with the specific 
safety concerns presented by the drug product.



Sec. 314.530  Withdrawal procedures.

    (a) For new drugs and antibiotics approved under Secs. 314.510 and 
314.520, FDA may withdraw approval, following a hearing as provided in 
part 15 of this chapter, as modified by this section, if:
    (1) A postmarketing clinical study fails to verify clinical benefit;
    (2) The applicant fails to perform the required postmarketing study 
with due diligence;
    (3) Use after marketing demonstrates that postmarketing restrictions 
are inadequate to assure safe use of the drug product;
    (4) The applicant fails to adhere to the postmarketing restrictions 
agreed upon;
    (5) The promotional materials are false or misleading; or
    (6) Other evidence demonstrates that the drug product is not shown 
to be safe or effective under its conditions of use.
    (b) Notice of opportunity for a hearing. The Director of the Center 
for Drug Evaluation and Research will give the applicant notice of an 
opportunity for a hearing on the Center's proposal to withdraw the 
approval of an application approved under Sec. 314.510 or Sec. 314.520. 
The notice, which will ordinarily be a letter, will state generally the 
reasons for the action and the proposed grounds for the order.
    (c) Submission of data and information. (1) If the applicant fails 
to file a written request for a hearing within 15 days of receipt of the 
notice, the applicant waives the opportunity for a hearing.
    (2) If the applicant files a timely request for a hearing, the 
agency will publish a notice of hearing in the Federal Register in 
accordance with Secs. 12.32(e) and 15.20 of this chapter.
    (3) An applicant who requests a hearing under this section must, 
within 30 days of receipt of the notice of opportunity for a hearing, 
submit the data and information upon which the applicant intends to rely 
at the hearing.
    (d) Separation of functions. Separation of functions (as specified 
in Sec. 10.55 of this chapter) will not apply at any point in withdrawal 
proceedings under this section.
    (e) Procedures for hearings. Hearings held under this section will 
be conducted in accordance with the provisions of part 15 of this 
chapter, with the following modifications:
    (1) An advisory committee duly constituted under part 14 of this 
chapter will be present at the hearing. The committee will be asked to 
review the issues involved and to provide advice and recommendations to 
the Commissioner of Food and Drugs.
    (2) The presiding officer, the advisory committee members, up to 
three representatives of the applicant, and up to three representatives 
of the Center may question any person during or at

[[Page 179]]

the conclusion of the person's presentation. No other person attending 
the hearing may question a person making a presentation. The presiding 
officer may, as a matter of discretion, permit questions to be submitted 
to the presiding officer for response by a person making a presentation.
    (f) Judicial review. The Commissioner's decision constitutes final 
agency action from which the applicant may petition for judicial review. 
Before requesting an order from a court for a stay of action pending 
review, an applicant must first submit a petition for a stay of action 
under Sec. 10.35 of this chapter.



Sec. 314.540  Postmarketing safety reporting.

    Drug products approved under this program are subject to the 
postmarketing recordkeeping and safety reporting applicable to all 
approved drug products, as provided in Secs. 314.80 and 314.81.



Sec. 314.550  Promotional materials.

    For drug products being considered for approval under this subpart, 
unless otherwise informed by the agency, applicants must submit to the 
agency for consideration during the preapproval review period copies of 
all promotional materials, including promotional labeling as well as 
advertisements, intended for dissemination or publication within 120 
days following marketing approval. After 120 days following marketing 
approval, unless otherwise informed by the agency, the applicant must 
submit promotional materials at least 30 days prior to the intended time 
of initial dissemination of the labeling or initial publication of the 
advertisement.



Sec. 314.560  Termination of requirements.

    If FDA determines after approval that the requirements established 
in Sec. 314.520, Sec. 314.530, or Sec. 314.550 are no longer necessary 
for the safe and effective use of a drug product, it will so notify the 
applicant. Ordinarily, for drug products approved under Sec. 314.510, 
these requirements will no longer apply when FDA determines that the 
required postmarketing study verifies and describes the drug product's 
clinical benefit and the drug product would be appropriate for approval 
under traditional procedures. For drug products approved under 
Sec. 314.520, the restrictions would no longer apply when FDA determines 
that safe use of the drug product can be assured through appropriate 
labeling. FDA also retains the discretion to remove specific 
postapproval requirements upon review of a petition submitted by the 
sponsor in accordance with Sec. 10.30.



PART 316--ORPHAN DRUGS--Table of Contents




                      Subpart A--General Provisions

Sec.
316.1  Scope of this part.
316.2  Purpose.
316.3  Definitions.
316.4  Address for submissions.

  Subpart B--Written Recommendations for Investigations of Orphan Drugs

316.10  Content and format of a request for written recommendations.
316.12  Providing written recommendations.
316.14  Refusal to provide written recommendations.

                Subpart C--Designation of an Orphan Drug

316.20  Content and format of a request for orphan-drug designation.
316.21  Verification of orphan-drug status.
316.22  Permanent-resident agent for foreign sponsor.
316.23  Timing of requests for orphan-drug designation; designation of 
          already approved drugs.
316.24  Granting orphan-drug designation.
316.25  Refusal to grant orphan-drug designation.
316.26  Amendment to orphan-drug designation.
316.27  Change in ownership of orphan-drug designation.
316.28  Publication of orphan-drug designations.
316.29  Revocation of orphan-drug designation.
316.30  Annual reports of holder of orphan-drug designation.

                Subpart D--Orphan-drug Exclusive Approval

316.31  Scope of orphan-drug exclusive approval.
316.34  FDA recognition of exclusive approval.
316.36  Insufficient quantities of orphan drugs.

[[Page 180]]

              Subpart E--Open Protocols for Investigations

316.40  Treatment use of a designated orphan drug.

                 Subpart F--Availability of Information

316.50  Guidelines.
316.52  Availability for public disclosure of data and information in 
          requests and applications.

    Authority:  21 U.S.C. 360aa, 360bb, 360cc, 360dd, 371.

    Source:  57 FR 62085, Dec. 29, 1992, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 316.1  Scope of this part.

    (a) This part implements sections 525, 526, 527, and 528 of the act 
and provides procedures to encourage and facilitate the development of 
drugs for rare diseases or conditions, including biological products and 
antibiotics. This part sets forth the procedures and requirements for:
    (1) Submissions to FDA of:
    (i) Requests for recommendations for investigations of drugs for 
rare diseases or conditions;
    (ii) Requests for designation of a drug for a rare disease or 
condition; and
    (iii) Requests for gaining exclusive approval for a drug product for 
a rare disease or condition.
    (2) Allowing a sponsor to provide an investigational drug product 
under a treatment protocol to patients who need the drug for treatment 
of a rare disease or condition.
    (b) This part does not apply to food, medical devices, or drugs for 
veterinary use.
    (c) References in this part to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21, unless otherwise 
noted.



Sec. 316.2  Purpose.

    The purpose of this part is to establish standards and procedures 
for determining eligibility for the benefits provided for in section 2 
of the Orphan Drug Act, including written recommendations for 
investigations of orphan drugs, a 7-year period of exclusive marketing, 
and treatment use of investigational orphan drugs. This part is also 
intended to satisfy Congress' requirements that FDA promulgate 
procedures for the implementation of sections 525(a) and 526(a) of the 
act.



Sec. 316.3  Definitions.

    (a) The definitions and interpretations contained in section 201 of 
the act apply to those terms when used in this part.
    (b) The following definitions of terms apply to this part:
    (1) Act means the Federal Food, Drug, and Cosmetic Act as amended by 
section 2 of the Orphan Drug Act (sections 525-528 (21 U.S.C. 360aa-
360dd)).
    (2) Active moiety means the molecule or ion, excluding those 
appended portions of the molecule that cause the drug to be an ester, 
salt (including a salt with hydrogen or coordination bonds), or other 
noncovalent derivative (such as a complex, chelate, or clathrate) of the 
molecule, responsible for the physiological or pharmacological action of 
the drug substance.
    (3) Clinically superior means that a drug is shown to provide a 
significant therapeutic advantage over and above that provided by an 
approved orphan drug (that is otherwise the same drug) in one or more of 
the following ways:
    (i) Greater effectiveness than an approved orphan drug (as assessed 
by effect on a clinically meaningful endpoint in adequate and well 
controlled clinical trials). Generally, this would represent the same 
kind of evidence needed to support a comparative effectiveness claim for 
two different drugs; in most cases, direct comparative clinical trials 
would be necessary; or
    (ii) Greater safety in a substantial portion of the target 
populations, for example, by the elimination of an ingredient or 
contaminant that is associated with relatively frequent adverse effects. 
In some cases, direct comparative clinical trials will be necessary; or
    (iii) In unusual cases, where neither greater safety nor greater 
effectiveness has been shown, a demonstration that the drug otherwise 
makes a major contribution to patient care.
    (4) Director means the Director of FDA's Office of Orphan Products 
Development.
    (5) FDA means the Food and Drug Administration.

[[Page 181]]

    (6) Holder means the sponsor in whose name an orphan drug is 
designated and approved.
    (7) IND means an investigational new drug application under part 312 
of this chapter.
    (8) Manufacturer means any person or agency engaged in the 
manufacture of a drug that is subject to investigation and approval 
under the act or the biologics provisions of the Public Health Service 
Act (42 U.S.C. 262-263).
    (9) Marketing application means an application for approval of a new 
drug filed under section 505(b) of the act, a request for certification 
of an antibiotic under section 507 of the act, or an application for a 
biological product/establishment license submitted under section 351 of 
the Public Health Service Act (42 U.S.C. 262).
    (10) Orphan drug means a drug intended for use in a rare disease or 
condition as defined in section 526 of the act.
    (11) Orphan-drug designation means FDA's act of granting a request 
for designation under section 526 of the act.
    (12) Orphan-drug exclusive approval or exclusive approval means 
that, effective on the date of FDA approval as stated in the approval 
letter of a marketing application for a sponsor of a designated orphan 
drug, no approval will be given to a subsequent sponsor of the same drug 
product for the same indication for 7 years, except as otherwise 
provided by law or in this part.
    (13) Same drug means:
    (i) If it is a drug composed of small molecules, a drug that 
contains the same active moiety as a previously approved drug and is 
intended for the same use as the previously approved drug, even if the 
particular ester or salt (including a salt with hydrogen or coordination 
bonds) or other noncovalent derivative such as a complex, chelate or 
clathrate has not been previously approved, except that if the 
subsequent drug can be shown to be clinically superior to the first 
drug, it will not be considered to be the same drug.
    (ii) If it is a drug composed of large molecules (macromolecules), a 
drug that contains the same principal molecular structural features (but 
not necessarily all of the same structural features) and is intended for 
the same use as a previously approved drug, except that, if the 
subsequent drug can be shown to be clinically superior, it will not be 
considered to be the same drug. This criterion will be applied as 
follows to different kinds of macromolecules:
    (A) Two protein drugs would be considered the same if the only 
differences in structure between them were due to post-translational 
events or infidelity of translation or transcription or were minor 
differences in amino acid sequence; other potentially important 
differences, such as different glycosylation patterns or different 
tertiary structures, would not cause the drugs to be considered 
different unless the differences were shown to be clinically superior.
    (B) Two polysaccharide drugs would be considered the same if they 
had identical saccharide repeating units, even if the number of units 
were to vary and even if there were postpolymerization modifications, 
unless the subsequent drug could be shown to be clinically superior.
    (C) Two polynucleotide drugs consisting of two or more distinct 
nucleotides would be considered the same if they had an identical 
sequence of purine and pyrimidine bases (or their derivatives) bound to 
an identical sugar backbone (ribose, deoxyribose, or modifications of 
these sugars), unless the subsequent drug were shown to be clinically 
superior.
    (D) Closely related, complex partly definable drugs with similar 
therapeutic intent, such as two live viral vaccines for the same 
indication, would be considered the same unless the subsequent drug was 
shown to be clinically superior.
    (14) Sponsor means the entity that assumes responsibility for a 
clinical or nonclinical investigation of a drug, including the 
responsibility for compliance with applicable provisions of the act and 
regulations. A sponsor may be an individual, partnership, corporation, 
or Government agency and may be a manufacturer, scientific institution, 
or an investigator regularly and lawfully engaged in the investigation 
of drugs. For purposes of the Orphan Drug Act, FDA considers the real 
party or parties in interest to be a sponsor.

[[Page 182]]



Sec. 316.4  Address for submissions.

    All correspondence and requests for FDA action pursuant to the 
provisions of this rule should be addressed as follows: Office of Orphan 
Products Development (HF-35), Food and Drug Administration, 5600 Fishers 
Lane, Rockville, MD 20857.



  Subpart B--Written Recommendations for Investigations of Orphan Drugs



Sec. 316.10  Content and format of a request for written recommendations.

    (a) A sponsor's request for written recommendations from FDA 
concerning the nonclinical and clinical investigations necessary for 
approval of a marketing application shall be submitted in the form and 
contain the information required in this section. FDA may require the 
sponsor to submit information in addition to that specified in paragraph 
(b) of this section if FDA determines that the sponsor's initial request 
does not contain adequate information on which to base recommendations.
    (b) A sponsor shall submit two copies of a completed, dated, and 
signed request for written recommendations that contains the following:
    (1) The sponsor's name and address.
    (2) A statement that the sponsor is requesting written 
recommendations on orphan-drug development under section 525 of the act.
    (3) The name of the sponsor's primary contact person and/or resident 
agent, and the person's title, address, and telephone number.
    (4) The generic name and trade name, if any, of the drug and a list 
of the drug product's components or description of the drug product's 
formulation, and chemical and physical properties.
    (5) The proposed dosage form and route of administration.
    (6) A description of the disease or condition for which the drug is 
proposed to be investigated and the proposed indication or indications 
for use for such disease or condition.
    (7) Current regulatory and marketing status and history of the drug 
product, including:
    (i) Whether the product is the subject of an IND or a marketing 
application (if the product is the subject of an IND or a marketing 
application, the IND or marketing application numbers should be stated 
and the investigational or approved indication or indications for use 
specified);
    (ii) Known marketing experience or investigational status outside 
the United States;
    (iii) So far as is known or can be determined, all indications 
previously or currently under investigation anywhere;
    (iv) All adverse regulatory actions taken by the United States or 
foreign authorities.
    (8) The basis for concluding that the drug is for a disease or 
condition that is rare in the United States, including the following:
    (i) The size and other known demographic characteristics of the 
patient population affected and the source of this information.
    (ii) For drugs intended for diseases or conditions affecting 200,000 
or more people in the United States, or for a vaccine, diagnostic drug, 
or preventive drug that would be given to 200,000 or more persons per 
year, a summary of the sponsor's basis for believing that the disease or 
condition described in paragraph (b)(6) of this section occurs so 
infrequently that there is no reasonable expectation that the costs of 
drug development and marketing will be recovered in future sales of the 
drug in the United States. The estimated costs and sales data should be 
submitted as provided for in Sec. 316.21(c).
    (9) A summary and analysis of available data on the pharmacologic 
effects of the drug.
    (10) A summary and analysis of available nonclinical and clinical 
data pertinent to the drug and the disease to be studied including 
copies of pertinent published reports. When a drug proposed for orphan 
drug designation is intended to treat a life-threatening or severely 
debilitating illness, especially where no satisfactory alternative 
therapy exists, the sponsor may wish voluntarily to provide this 
information. A sponsor of such a drug may be entitled

[[Page 183]]

to expeditious development, evaluation, and marketing under 21 CFR part 
312, subpart E.
    (11) An explanation of how the data summarized and analyzed under 
paragraphs (b)(9) and (b)(10) of this section support the rationale for 
use of the drug in the rare disease or condition.
    (12) A definition of the population from which subjects will be 
identified for clinical trials, if known.
    (13) A detailed outline of any protocols under which the drug has 
been or is being studied for the rare disease or condition and a summary 
and analysis of any available data from such studies.
    (14) The sponsor's proposal as to the scope of nonclinical and 
clinical investigations needed to establish the safety and effectiveness 
of the drug.
    (15) Detailed protocols for each proposed United States or foreign 
clinical investigation, if available.
    (16) Specific questions to be addressed by FDA in its 
recommendations for nonclinical laboratory studies and clinical 
investigations.
[57 FR 62085, Dec. 29, 1992; 58 FR 6167, Jan. 26, 1993]



Sec. 316.12  Providing written recommendations.

    (a) FDA will provide the sponsor with written recommendations 
concerning the nonclinical laboratory studies and clinical 
investigations necessary for approval of a marketing application if none 
of the reasons described in Sec. 316.14 for refusing to do so applies.
    (b) When a sponsor seeks written recommendations at a stage of drug 
development at which advice on any clinical investigations, or on 
particular investigations would be premature, FDA's response may be 
limited to written recommendations concerning only nonclinical 
laboratory studies, or only certain of the clinical studies (e.g., Phase 
1 studies as described in Sec. 312.21 of this chapter). Prior to 
providing written recommendations for the clinical investigations 
required to achieve marketing approval, FDA may require that the results 
of the nonclinical laboratory studies or completed early clinical 
studies be submitted to FDA for agency review.



Sec. 316.14  Refusal to provide written recommendations.

    (a) FDA may refuse to provide written recommendations concerning the 
nonclinical laboratory studies and clinical investigations necessary for 
approval of a marketing application for any of the following reasons:
    (1) The information required to be submitted by Sec. 316.10(b) has 
not been submitted, or the information submitted is incomplete.
    (2) There is insufficient information about:
    (i) The drug to identify the active moiety and its physical and 
chemical properties, if these characteristics can be determined; or
    (ii) The disease or condition to determine that the disease or 
condition is rare in the United States; or
    (iii) The reasons for believing that the drug may be useful for 
treating the rare disease or condition with that drug; or
    (iv) The regulatory and marketing history of the drug to determine 
the scope and type of investigations that have already been conducted on 
the drug for the rare disease or condition; or
    (v) The plan of study for establishing the safety and effectiveness 
of the drug for treatment of the rare disease or condition.
    (3) The specific questions for which the sponsor seeks the advice of 
the agency are unclear or are not sufficiently specific.
    (4) On the basis of the information submitted and on other 
information available to the agency, FDA determines that the disease or 
condition for which the drug is intended is not rare in the United 
States.
    (5) On the basis of the information submitted and on other 
information available to the agency, FDA determines that there is an 
inadequate basis for permitting investigational use of the drug under 
part 312 of this chapter for the rare disease or condition.
    (6) The request for information contains an untrue statement of 
material fact.
    (b) A refusal to provide written recommendations will be in writing 
and will include a statement of the reason for FDA's refusal. Where 
practicable,

[[Page 184]]

FDA will describe the information or material it requires or the 
conditions the sponsor must meet for FDA to provide recommendations.
    (c) Within 90 days after the date of a letter from FDA requesting 
additional information or material or setting forth the conditions that 
the sponsor is asked to meet, the sponsor shall either:
    (1) Provide the information or material or amend the request for 
written recommendations to meet the conditions sought by FDA; or
    (2) Withdraw the request for written recommendations. FDA will 
consider a sponsor's failure to respond within 90 days to an FDA letter 
requesting information or material or setting forth conditions to be met 
to be a withdrawal of the request for written recommendations.



                Subpart C--Designation of an Orphan Drug



Sec. 316.20  Content and format of a request for orphan-drug designation.

    (a) A sponsor that submits a request for orphan-drug designation of 
a drug for a specified rare disease or condition shall submit each 
request in the form and containing the information required in paragraph 
(b) of this section. A sponsor may request orphan-drug designation of a 
previously unapproved drug, or of a new orphan indication for an already 
marketed drug. In addition, a sponsor of a drug that is otherwise the 
same drug as an already approved orphan drug may seek and obtain orphan-
drug designation for the subsequent drug for the same rare disease or 
condition if it can present a plausible hypothesis that its drug may be 
clinically superior to the first drug. More than one sponsor may receive 
orphan-drug designation of the same drug for the same rare disease or 
condition, but each sponsor seeking orphan-drug designation must file a 
complete request for designation as provided in paragraph (b) of this 
section.
    (b) A sponsor shall submit two copies of a completed, dated, and 
signed request for designation that contains the following:
    (1) A statement that the sponsor requests orphan-drug designation 
for a rare disease or condition, which shall be identified with 
specificity.
    (2) The name and address of the sponsor; the name of the sponsor's 
primary contact person and/or resident agent including title, address, 
and telephone number; the generic and trade name, if any, of the drug or 
drug product; and the name and address of the source of the drug if it 
is not manufactured by the sponsor.
    (3) A description of the rare disease or condition for which the 
drug is being or will be investigated, the proposed indication or 
indications for use of the drug, and the reasons why such therapy is 
needed.
    (4) A description of the drug and a discussion of the scientific 
rationale for the use of the drug for the rare disease or condition, 
including all data from nonclinical laboratory studies, clinical 
investigations, and other relevant data that are available to the 
sponsor, whether positive, negative, or inconclusive. Copies of 
pertinent unpublished and published papers are also required.
    (5) Where the sponsor of a drug that is otherwise the same drug as 
an already-approved orphan drug seeks orphan-drug designation for the 
subsequent drug for the same rare disease or condition, an explanation 
of why the proposed variation may be clinically superior to the first 
drug.
    (6) Where a drug is under development for only a subset of persons 
with a particular disease or condition, a demonstration that the subset 
is medically plausible.
    (7) A summary of the regulatory status and marketing history of the 
drug in the United States and in foreign countries, e.g., IND and 
marketing application status and dispositions, what uses are under 
investigation and in what countries; for what indication is the drug 
approved in foreign countries; what adverse regulatory actions have been 
taken against the drug in any country.
    (8) Documentation, with appended authoritative references, to 
demonstrate that:
    (i) The disease or condition for which the drug is intended affects 
fewer than 200,000 people in the United States or, if the drug is a 
vaccine, diagnostic

[[Page 185]]

drug, or preventive drug, the persons to whom the drug will be 
administered in the United States are fewer than 200,000 per year as 
specified in Sec. 316.21(b), or
    (ii) For a drug intended for diseases or conditions affecting 
200,000 or more people, or for a vaccine, diagnostic drug, or preventive 
drug to be administered to 200,000 or more persons per year in the 
United States, there is no reasonable expectation that costs of research 
and development of the drug for the indication can be recovered by sales 
of the drug in the United States as specified in Sec. 316.21(c).
    (9) A statement as to whether the sponsor submitting the request is 
the real party in interest of the development and the intended or actual 
production and sales of the product.
    (c) Any of the information previously provided by the sponsor to FDA 
under subpart B of this part may be referenced by specific page or 
location if it duplicates information required elsewhere in this 
section.



Sec. 316.21  Verification of orphan-drug status.

    (a) So that FDA can determine whether a drug qualifies for orphan-
drug designation under section 526(a) of the act, the sponsor shall 
include in its request to FDA for orphan-drug designation under 
Sec. 316.20 either:
    (1) Documentation as described in paragraph (b) of this section that 
the number of people affected by the disease or condition for which the 
drug product is indicated is fewer than 200,000 persons; or
    (2) Documentation as described in paragraph (c) of this section that 
demonstrates that there is no reasonable expectation that the sales of 
the drug will be sufficient to offset the costs of developing the drug 
for the U.S. market and the costs of making the drug available in the 
United States.
    (b) For the purpose of documenting that the number of people 
affected by the disease or condition for which the drug product is 
indicated is less than 200,000 persons, ``prevalence'' is defined as the 
number of persons in the United States who have been diagnosed as having 
the disease or condition at the time of the submission of the request 
for orphan-drug designation. To document the number of persons in the 
United States who have the disease or condition for which the drug is to 
be indicated, the sponsor shall submit to FDA evidence showing:
    (1) The estimated prevalence of the disease or condition for which 
the drug is being developed, together with a list of the sources 
(including dates of information provided and literature citations) for 
the estimate;
    (2) Upon request by FDA, the estimated prevalence of any other 
disease or condition for which the drug has already been approved or for 
which the drug is currently being developed, together with an 
explanation of the bases of these estimates; and
    (3) The estimated number of people to whom the drug will be 
administered annually if the drug is a vaccine or is a drug intended for 
diagnosis or prevention of a rare disease or condition, together with an 
explanation of the bases of these estimates (including dates of 
information provided and literature citations).
    (c) When submitting documentation that there is no reasonable 
expectation that costs of research and development of the drug for the 
disease or condition can be recovered by sales of the drug in the United 
States, the sponsor shall submit to FDA:
    (1) Data on all costs that the sponsor has incurred in the course of 
developing the drug for the U.S. market. These costs shall include, but 
are not limited to, nonclinical laboratory studies, clinical studies, 
dosage form development, record and report maintenance, meetings with 
FDA, determination of patentability, preparation of designation request, 
IND/marketing application preparation, distribution of the drug under a 
``treatment'' protocol, licensing costs, liability insurance, and 
overhead and depreciation. Furthermore, the sponsor shall demonstrate 
the reasonableness of the cost data. For example, if the sponsor has 
incurred costs for clinical investigations, the sponsor shall provide 
information on the number of investigations, the years in which they 
took place, and on the scope, duration, and number of patients that were 
involved in each investigation.

[[Page 186]]

    (2) If the drug was developed wholly or in part outside the United 
States, in addition to the documentation listed in paragraph (c)(1) of 
this section:
    (i) Data on and justification for all costs that the sponsor has 
incurred outside of the United States in the course of developing the 
drug for the U.S. market. The justification, in addition to 
demonstrating the reasonableness of the cost data, must also explain the 
method that was used to determine which portion of the foreign 
development costs should be applied to the U.S. market, and what percent 
these costs are of total worldwide development costs. Any data submitted 
to foreign government authorities to support drug pricing determinations 
must be included with this information.
    (ii) Data that show which foreign development costs were recovered 
through cost recovery procedures that are allowed during drug 
development in some foreign countries. For example, if the sponsor 
charged patients for the drug during clinical investigations, the 
revenues collected by the sponsor must be reported to FDA.
    (3) In cases where the drug has already been approved for marketing 
for any indication or in cases where the drug is currently under 
investigation for one or more other indications (in addition to the 
indication for which orphan-drug designation is being sought), a clear 
explanation of and justification for the method that is used to 
apportion the development costs among the various indications.
    (4) A statement of and justification for any development costs that 
the sponsor expects to incur after the submission of the designation 
request. In cases where the extent of these future development costs are 
not clear, the sponsor should request FDA's advice and assistance in 
estimating the scope of nonclinical laboratory studies and clinical 
investigations and other data that are needed to support marketing 
approval. Based on these recommendations, a cost estimate should be 
prepared.
    (5) A statement of and justification for production and marketing 
costs that the sponsor has incurred in the past and expects to incur 
during the first 7 years that the drug is marketed.
    (6) An estimate of and justification for the expected revenues from 
sales of the drug in the United States during its first 7 years of 
marketing. The justification should assume that the total market for the 
drug is equal to the prevalence of the disease or condition that the 
drug will be used to treat. The justification should include:
    (i) An estimate of the expected market share of the drug in each of 
the first 7 years that it is marketed, together with an explanation of 
the basis for that estimate;
    (ii) A projection of and justification for the price at which the 
drug will be sold; and
    (iii) Comparisons with sales of similarly situated drugs, where 
available.
    (7) The name of each country where the drug has already been 
approved for marketing for any indication, the dates of approval, the 
indication for which the drug is approved, and the annual sales and 
number of prescriptions in each country since the first approval date.
    (8) A report of an independent certified public accountant in 
accordance with Statement on Standards for Attestation established by 
the American Institute of Certified Public Accountants on agreed upon 
procedures performed with respect to the data estimates and 
justifications submitted pursuant to this section. Cost data shall be 
determined in accordance with generally accepted accounting principles.
    (d) A sponsor that is requesting orphan-drug designation for a drug 
designed to treat a disease or condition that affects 200,000 or more 
persons shall, at FDA's request, allow FDA or FDA-designated personnel 
to examine at reasonable times and in a reasonable manner all relevant 
financial records and sales data of the sponsor and manufacturer.



Sec. 316.22  Permanent-resident agent for foreign sponsor.

    Every foreign sponsor that seeks orphan-drug designation shall name 
a permanent resident of the United States as the sponsor's agent upon 
whom service of all processes, notices, orders, decisions, requirements, 
and other communications may be made on

[[Page 187]]

behalf of the sponsor. Notifications of changes in such agents or 
changes of address of agents should preferably be provided in advance, 
but not later than 60 days after the effective date of such changes. The 
permanent-resident agent may be an individual, firm, or domestic 
corporation and may represent any number of sponsors. The name of the 
permanent-resident agent shall be provided to: Office of Orphan Products 
Development (HF-35), Food and Drug Administration, 5600 Fishers Lane, 
Rockville, MD 20857.



Sec. 316.23  Timing of requests for orphan-drug designation; designation of already approved drugs.

    (a) A sponsor may request orphan-drug designation at any time in the 
drug development process prior to the submission of a marketing 
application for the drug product for the orphan indication.
    (b) A sponsor may request orphan-drug designation of an already 
approved drug product for an unapproved use without regard to whether 
the prior marketing approval was for an orphan-drug indication.



Sec. 316.24  Granting orphan-drug designation.

    (a) FDA will grant the request for orphan-drug designation if none 
of the reasons described in Sec. 316.25 for requiring or permitting 
refusal to grant such a request applies.
    (b) When a request for orphan-drug designation is granted, FDA will 
notify the sponsor in writing and will publicize the orphan-drug 
designation in accordance with Sec. 316.28.



Sec. 316.25  Refusal to grant orphan-drug designation.

    (a) FDA will refuse to grant a request for orphan-drug designation 
if any of the following reasons apply:
    (1) The drug is not intended for a rare disease or condition 
because:
    (i) There is insufficient evidence to support the estimate that the 
drug is intended for treatment of a disease or condition in fewer than 
200,000 people in the United States, or that the drug is intended for 
use in prevention or in diagnosis in fewer than 200,000 people annually 
in the United States; or
    (ii) Where the drug is intended for prevention, diagnosis, or 
treatment of a disease or condition affecting 200,000 or more people in 
the United States, the sponsor has failed to demonstrate that there is 
no reasonable expectation that development and production costs will be 
recovered from sales of the drug for the orphan indication in the United 
States. A sponsor's failure to comply with Sec. 316.21 shall constitute 
a failure to make the demonstration required in this paragraph.
    (2) There is insufficient information about the drug, or the disease 
or condition for which it is intended, to establish a medically 
plausible basis for expecting the drug to be effective in the 
prevention, diagnosis, or treatment of that disease or condition.
    (3) A drug that is otherwise the same drug as one that already has 
orphan-drug exclusive approval for the same rare disease or condition 
and the sponsor has not submitted a medically plausible hypothesis for 
the possible clinical superiority of the subsequent drug.
    (b) FDA may refuse to grant a request for orphan-drug designation if 
the request for designation contains an untrue statement of material 
fact or omits material information.



Sec. 316.26  Amendment to orphan-drug designation.

    (a) At any time prior to approval of a marketing application for a 
designated orphan drug, the sponsor holding designation may apply for an 
amendment to the indication stated in the orphan-drug designation if the 
proposed change is due to new and unexpected findings in research on the 
drugs, information arising from FDA recommendations, or unforeseen 
developments in treatment or diagnosis of the disease or condition.
    (b) FDA will grant the amendment if it finds that the initial 
designation request was made in good faith and that the amendment is 
intended to conform the orphan-drug designation indication to the 
results of unanticipated research findings, to unforeseen developments 
in the treatment or diagnosis of the disease or condition, or to changes 
based on FDA recommendations, and that, as of the date of the submission

[[Page 188]]

of the amendment request, the amendment would not result in exceeding 
the prevalence or cost recovery thresholds in Sec. 316.21 (a)(1) or 
(a)(2) upon which the drug was originally designated.



Sec. 316.27  Change in ownership of orphan-drug designation.

    (a) A sponsor may transfer ownership of or any beneficial interest 
in the orphan-drug designation of a drug to a new sponsor. At the time 
of the transfer, the new and former owners are required to submit the 
following information to FDA:
    (1) The former owner or assignor of rights shall submit a letter or 
other document that states that all or some rights to the orphan-drug 
designation of the drug have been transferred to the new owner or 
assignee and that a complete copy of the request for orphan-drug 
designation, including any amendments to the request, supplements to the 
granted request, and correspondence relevant to the orphan-drug 
designation, has been provided to the new owner or assignee.
    (2) The new owner or assignee of rights shall submit a statement 
accepting orphan-drug designation and a letter or other document 
containing the following:
    (i) The date that the change in ownership or assignment of rights is 
effective;
    (ii) A statement that the new owner has a complete copy of the 
request for orphan-drug designation including any amendments to the 
request, supplements to the granted request, and correspondence relevant 
to the orphan-drug designation; and
    (iii) A specific description of the rights that have been assigned 
and those that have been reserved. This may be satisfied by the 
submission of either a list of rights assigned and reserved or copies of 
all relevant agreements between assignors and assignees; and
    (iv) The name and address of a new primary contact person or 
resident agent.
    (b) No sponsor may relieve itself of responsibilities under the 
Orphan Drug Act or under this part by assigning rights to another person 
without:
    (1) Assuring that the sponsor or the assignee will carry out such 
responsibilities; or
    (2) Obtaining prior permission from FDA.
[57 FR 62085, Dec. 29, 1992; 58 FR 6167, Jan. 26, 1993]



Sec. 316.28  Publication of orphan-drug designations.

    Each month FDA will update a publically available list of drugs 
designated as orphan drugs. A cumulative, updated list of all designated 
drugs will be provided annually. These will be placed on file at the FDA 
Dockets Management Branch, and will contain the following information:
    (a) The name and address of the manufacturer and sponsor;
    (b) The generic name and trade name, if any, of the drug and the 
date of the granting of orphan-drug designation;
    (c) The rare disease or condition for which orphan-drug designation 
was granted; and
    (d) The proposed indication for use of the drug.



Sec. 316.29  Revocation of orphan-drug designation.

    (a) FDA may revoke orphan-drug designation for any drug if the 
agency finds that:
    (1) The request for designation contained an untrue statement of 
material fact; or
    (2) The request for designation omitted material information 
required by this part; or
    (3) FDA subsequently finds that the drug in fact had not been 
eligible for orphan-drug designation at the time of submission of the 
request therefor.
    (b) For an approved drug, revocation of orphan-drug designation also 
suspends or withdraws the sponsor's exclusive marketing rights for the 
drug but not the approval of the drug's marketing application.
    (c) Where a drug has been designated as an orphan drug because the 
prevalence of a disease or condition (or, in the case of vaccines, 
diagnostic drugs, or preventive drugs, the target population) is under 
200,000 in the United States at the time of designation, its designation 
will not be revoked on the

[[Page 189]]

ground that the prevalence of the disease or condition (or the target 
population) becomes more than 200,000 persons.



Sec. 316.30  Annual reports of holder of orphan-drug designation.

    Within 14 months after the date on which a drug was designated as an 
orphan drug and annually thereafter until marketing approval, the 
sponsor of a designated drug shall submit a brief progress report to the 
FDA Office of Orphan Products Development on the drug that includes:
    (a) A short account of the progress of drug development including a 
review of preclinical and clinical studies initiated, ongoing, and 
completed and a short summary of the status or results of such studies.
    (b) A description of the investigational plan for the coming year, 
as well as any anticipated difficulties in development, testing, and 
marketing; and
    (c) A brief discussion of any changes that may affect the orphan-
drug status of the product. For example, for products nearing the end of 
the approval process, sponsors should discuss any disparity between the 
probable marketing indication and the designated indication as related 
to the need for an amendment to the orphan-drug designation pursuant to 
Sec. 316.26.



                Subpart D--Orphan-drug Exclusive Approval



Sec. 316.31  Scope of orphan-drug exclusive approval.

    (a) After approval of a sponsor's marketing application for a 
designated orphan-drug product for treatment of the rare disease or 
condition concerning which orphan-drug designation was granted, FDA will 
not approve another sponsor's marketing application for the same drug 
before the expiration of 7 years from the date of such approval as 
stated in the approval letter from FDA, except that such a marketing 
application can be approved sooner if, and such time as, any of the 
following occurs:
    (1) Withdrawal of exclusive approval or revocation of orphan-drug 
designation by FDA under any provision of this part; or
    (2) Withdrawal for any reason of the marketing application for the 
drug in question; or
    (3) Consent by the holder of exclusive approval to permit another 
marketing application to gain approval; or
    (4) Failure of the holder of exclusive approval to assure a 
sufficient quantity of the drug under section 527 of the act and 
Sec. 316.36.
    (b) If a sponsor's marketing application for a drug product is 
determined not to be approvable because approval is barred under section 
527 of the act until the expiration of the period of exclusive marketing 
of another drug product, FDA will so notify the sponsor in writing.



Sec. 316.34  FDA recognition of exclusive approval.

    (a) FDA will send the sponsor (or, the permanent-resident agent, if 
applicable) timely written notice recognizing exclusive approval once 
the marketing application for a designated orphan-drug product has been 
approved. The written notice will inform the sponsor of the requirements 
for maintaining orphan-drug exclusive approval for the full 7-year term 
of exclusive approval.
    (b) When a marketing application is approved for a designated orphan 
drug that qualifies for exclusive approval, FDA will publish in its 
publication entitled ``Approved Drug Products with Therapeutic 
Equivalence Evaluations'' information identifying the sponsor, the drug, 
and the date of termination of the orphan-drug exclusive approval. A 
subscription to this publication and its monthly cumulative supplements 
is available from the Superintendent of Documents, Government Printing 
Office, Washington, DC 20402-9325.



Sec. 316.36  Insufficient quantities of orphan drugs.

    (a) Under section 527 of the act, whenever the Director has reason 
to believe that the holder of exclusive approval cannot assure the 
availability of sufficient quantities of an orphan drug to meet the 
needs of patients with the disease or condition for which the drug was 
designated, the Director will so notify the holder of this possible 
insufficiency and will offer the holder one of the following options, 
which must be

[[Page 190]]

exercised by a time that the Director specifies:
    (1) Provide the Director in writing, or orally, or both, at the 
Director's discretion, views and data as to how the holder can assure 
the availability of sufficient quantities of the orphan drug within a 
reasonable time to meet the needs of patients with the disease or 
condition for which the drug was designated; or
    (2) Provide the Director in writing the holder's consent for the 
approval of other marketing applications for the same drug before the 
expiration of the 7-year period of exclusive approval.
    (b) If, within the time that the Director specifies, the holder 
fails to consent to the approval of other marketing applications and if 
the Director finds that the holder has not shown that it can assure the 
availability of sufficient quantities of the orphan drug to meet the 
needs of patients with the disease or condition for which the drug was 
designated, the Director will issue a written order withdrawing the drug 
product's exclusive approval. This order will embody the Director's 
findings and conclusions and will constitute final agency action. An 
order withdrawing the sponsor's exclusive marketing rights may issue 
whether or not there are other sponsors that can assure the availability 
of alternative sources of supply. Once withdrawn under this section, 
exclusive approval may not be reinstated for that drug.



              Subpart E--Open Protocols for Investigations



Sec. 316.40  Treatment use of a designated orphan drug.

    Prospective investigators seeking to obtain treatment use of 
designated orphan drugs may do so as provided in Sec. 312.34 of this 
chapter.



                 Subpart F--Availability of Information



Sec. 316.50  Guidelines.

    FDA's Office of Orphan Products Development will maintain and make 
publicly available a list of guidelines that apply to the regulations in 
this part. The list states how a person can obtain a copy of each 
guideline. A request for a copy of the list or for any guideline should 
be directed to the Office of Orphan Products Development (HF-35), Food 
and Drug Administration, 5600 Fishers Lane, Rockville, MD 20857.



Sec. 316.52  Availability for public disclosure of data and information in requests and applications.

    (a) FDA will not publicly disclose the existence of a request for 
orphan-drug designation under section 526 of the act prior to final FDA 
action on the request unless the existence of the request has been 
previously publicly disclosed or acknowledged.
    (b) Whether or not the existence of a pending request for 
designation has been publicly disclosed or acknowledged, no data or 
information in the request are available for public disclosure prior to 
final FDA action on the request.
    (c) Upon final FDA action on a request for designation, FDA will 
determine the public availability of data and information in the request 
in accordance with part 20 and Sec. 314.430 of this chapter and other 
applicable statutes and regulations.
    (d) In accordance with Sec. 316.28, FDA will make a cumulative list 
of all orphan drug designations available to the public and update such 
list monthly.
    (e) FDA will not publicly disclose the existence of a pending 
marketing application for a designated orphan drug for the use for which 
the drug was designated unless the existence of the application has been 
previously publicly disclosed or acknowledged.
    (f) FDA will determine the public availability of data and 
information contained in pending and approved marketing applications for 
a designated orphan drug for the use for which the drug was designated 
in accordance with part 20 and Sec. 314.430 of this chapter and other 
applicable statutes and regulations.



PART 320--BIOAVAILABILITY AND BIOEQUIVALENCE REQUIREMENTS--Table of Contents




                      Subpart A--General Provisions

Sec.
320.1  Definitions.

[[Page 191]]

      Subpart B--Procedures for Determining the Bioavailability or 
                     Bioequivalence of Drug Products

320.21  Requirements for submission of in vivo bioavailability and 
          bioequivalence data.
320.22  Criteria for waiver of evidence of in vivo bioavailability or 
          bioequivalence.
320.23  Basis for demonstrating in vivo bioavailability or 
          bioequivalence.
320.24  Types of evidence to establish bioavailability or 
          bioequivalence.
320.25  Guidelines for the conduct of an in vivo bioavailability study.
320.26  Guidelines on the design of a single-dose in vivo 
          bioavailability study.
320.27  Guidelines on the design of a multiple-dose in vivo 
          bioavailability study.
320.28  Correlation of bioavailability with an acute pharmacological 
          effect or clinical evidence.
320.29  Analytical methods for an in vivo bioavailability study.
320.30  Inquiries regarding bioavailability and bioequivalence 
          requirements and review of protocols by the Food and Drug 
          Administration.
320.31  Applicability of requirements regarding an ``Investigational New 
          Drug Application.''
320.32  Procedures for establishing or amending a bioequivalence 
          requirement.
320.33  Criteria and evidence to assess actual or potential 
          bioequivalence problems.
320.34  Requirements for batch testing and certification by the Food and 
          Drug Administration.
320.35  Requirements for in vitro testing of each batch.
320.36  Requirements for maintenance of records of bioequivalence 
          testing.
320.38  Retention of bioavailability samples.
320.63  Retention of bioequivalence samples.

    Authority:  21 U.S.C. 321, 351, 352, 355, 357, 371.



                      Subpart A--General Provisions



Sec. 320.1  Definitions.

    (a) Bioavailability means the rate and extent to which the active 
ingredient or active moiety is absorbed from a drug product and becomes 
available at the site of action. For drug products that are not intended 
to be absorbed into the bloodstream, bioavailability may be assessed by 
measurements intended to reflect the rate and extent to which the active 
ingredient or active moiety becomes available at the site of action.
    (b) Drug product means a finished dosage form, e.g., tablet, 
capsule, or solution, that contains the active drug ingredient, 
generally, but not necessarily, in association with inactive 
ingredients.
    (c) Pharmaceutical equivalents means drug products that contain 
identical amounts of the identical active drug ingredient, i.e., the 
same sat or ester of the same therapeutic moiety, in identical dosage 
forms, but not necessarily containing the same inactive ingredients, and 
that meet the identical compendial or other applicable standard of 
identity, strength, quality, and purity, including potency and, where 
applicable, content uniformity, disintegration times and/or dissolution 
rates.
    (d) Pharmaceutical alternatives means drug products that contain the 
identical therapeutic moiety, or its precursor, but not necessarily in 
the same amount or dosage form or as the same salt or ester. Each such 
drug product individually meets either the identical or its own 
respective compendial or other applicable standard of identity, 
strength, quality, and purity, including potency and, where applicable, 
content uniformity, disintegration times and/or dissolution rates.
    (e) Bioequivalence means the absence of a significant difference in 
the rate and extent to which the active ingredient or active moiety in 
pharmaceutical equivalents or pharmaceutical alternatives becomes 
available at the site of drug action when administered at the same molar 
dose under similar conditions in an appropriately designed study. Where 
there is an intentional difference in rate (e.g., in certain controlled 
release dosage forms), certain pharmaceutical equivalents or 
alternatives may be considered bioequivalent if there is no significant 
difference in the extent to which the active ingredient or moiety from 
each product becomes available at the site of drug action. This applies 
only if the difference in the rate at which the active ingredient or 
moiety becomes available at the site of drug action is intentional and 
is reflected in the proposed labeling, is not essential to the 
attainment of effective body drug concentrations on chronic use, and is 
considered medically insignificant for the drug.
    (f) Bioequivalence requirement means a requirement imposed by the 
Food and

[[Page 192]]

Drug Administration for in vitro and/or in vivo testing of specified 
drug products which must be satisfied as a condition of marketing.
[42 FR 1634, Jan. 7, 1977, as amended at 42 FR 1648, Jan. 7, 1977; 57 FR 
17997, Apr. 28, 1992]



      Subpart B--Procedures for Determining the Bioavailability or 
                     Bioequivalence of Drug Products

    Source:  42 FR 1648, Jan. 7, 1977, unless otherwise noted.



Sec. 320.21  Requirements for submission of in vivo bioavailability and bioequivalence data.

    (a) Any person submitting a full new drug application to the Food 
and Drug Administration (FDA) shall include in the application either:
    (1) Evidence demonstrating the in vivo bioavailability of the drug 
product that is the subject of the application; or
    (2) Information to permit FDA to waive the submission of evidence 
demonstrating in vivo bioavailability.
    (b) Any person submitting an abbreviated new drug application to FDA 
shall include in the application either:
    (1) Evidence demonstrating that the drug product that is the subject 
of the abbreviated new drug application is bioequivalent to the 
reference listed drug (defined in Sec. 314.3(b)); or
    (2) Information to show that the drug product is bioequivalent to 
the reference listed drug which would permit FDA to waive the submission 
of evidence demonstrating bioequivalence as provided in paragraph (f) of 
this section.
    (c) Any person submitting a supplemental application to FDA shall 
include in the supplemental application the evidence or information set 
forth in paragraphs (a) and (b) of this section if the supplemental 
application proposes any of the following changes:
    (1) A change in the manufacturing process, including a change in 
product formulation or dosage strength, beyond the variations provided 
for in the approved application.
    (2) A change in the labeling to provide for a new indication for use 
of the drug product, if clinical studies are required to support the new 
indication for use.
    (3) A change in the labeling to provide for a new dosage regimen or 
for an additional dosage regimen for a special patient population, e.g., 
infants, if clinical studies are required to support the new or 
additional dosage regimen.
    (d) FDA may approve a full new drug application, or a supplemental 
application proposing any of the changes set forth in paragraph (c) of 
this section, that does not contain evidence of in vivo bioavailability 
or information to permit waiver of the requirement for in vivo 
bioavailability data, if all of the following conditions are met.
    (1) The application was under review by FDA on July 7, 1977.
    (2) The application is otherwise approvable.
    (3) The application agrees to submit, within the time specified by 
FDA, either:
    (i) Evidence demonstrating the in vivo bioavailability of the drug 
product that is the subject of the application; or
    (ii) Information to permit FDA to waive demonstration of in vivo 
bioavailability.
    (e) Evidence demonstrating the in vivo bioavailability and 
bioequivalence of a drug product shall be obtained using one of the 
approaches for determining bioavailability set forth in Sec. 320.24.
    (f) Information to permit FDA to waive the submission of evidence 
demonstrating the in vivo bioavailability or bioequivalence shall meet 
the criteria set forth in Sec. 320.24.
    (g) Any person holding an approved full or abbreviated new drug 
application shall submit to FDA a supplemental application containing 
new evidence demonstrating the in vivo bioavailability or bioequivalence 
of the drug product that is the subject of the application if notified 
by FDA that:
    (1) There are data demonstrating that the dosage regimen in the 
labeling is based on incorrect assumptions or facts regarding the 
pharmacokinetics of the drug product and that following this dosage 
regimen could potentially result in subtherapeutic or toxic levels; or

[[Page 193]]

    (2) There are data demonstrating significant intra-batch and batch-
to-batch variability, e.g., plus or minus 25 percent, in the 
bioavailability of the drug product.
    (h) The requirements of this section regarding the submission of 
evidence demonstrating in vivo bioavailability and bioequivalence apply 
only to a full or abbreviated new drug application or a supplemental 
application for a finished dosage formulation.
[57 FR 17998, Apr. 28, 1992]



Sec. 320.22  Criteria for waiver of evidence of in vivo bioavailability or bioequivalence.

    (a) Any person submitting a full or abbreviated new drug 
application, or a supplemental application proposing any of the changes 
set forth in Sec. 320.21(c), may request FDA to waive the requirement 
for the submission of evidence demonstrating the in vivo bioavailability 
or bioequivalence of the drug product that is the subject of the 
application. An applicant shall submit a request for waiver with the 
application. Except as provided in paragraph (g) of this section, FDA 
shall waive the requirement for the submission of evidence of in vivo 
bioavailability or bioequivalence if the drug product meets any of the 
provisions of paragraphs (b), (c), (d), or (e) of this section.
    (b) For certain drug products, the in vivo bioavailability or 
bioequivalence of the drug product may be self-evident. FDA shall waive 
the requirement for the submission of evidence obtained in vivo 
demonstrating the bioavailability or bioequivalence of these drug 
products. A drug product's in vivo bioavailability or bioequivalence may 
be considered self-evident based on other data in the application if the 
product meets one of the following criteria:
    (1) The drug product:
    (i) Is a parenteral solution intended solely for administration by 
injection, or an ophthalmic or otic solution; and
    (ii) Contains the same active and inactive ingredients in the same 
concentration as a drug product that is the subject of an approved full 
new drug application.
    (2) The drug product:
    (i) Is administered by inhalation as a gas, e.g., a medicinal or an 
inhalation anesthetic; and
    (ii) Contains an active ingredient in the same dosage form as a drug 
product that is the subject of an approved full new drug application.
    (3) The drug product:
    (i) Is a solution for application to the skin, an oral solution, 
elixir, syrup, tincture, or similar other solubilized form.
    (ii) Contains an active drug ingredient in the same concentration 
and dosage form as a drug product that is the subject of an approved 
full new drug application; and
    (iii) Contains no inactive ingredient or other change in formulation 
from the drug product that is the subject of the approved full new drug 
application that may significantly affect absorption of the active drug 
ingredient or active moiety.
    (c) FDA shall waive the requirement for the submission of evidence 
demonstrating the in vivo bioavailability of a solid oral dosage form 
(other than an enteric coated or controlled release dosage form) of a 
drug product determined to be effective for at least one indication in a 
Drug Efficacy Study Implementation notice or which is identical, 
related, or similar to such a drug product under Sec. 310.6 of this 
chapter unless FDA has evaluated the drug product under the criteria set 
forth in Sec. 320.32, included the drug product in the Approved Drug 
Products with Therapeutic Equivalence Evaluations List, and rated the 
drug product as having a known or potential bioequivalence problem. A 
drug product so rated reflects a determination by FDA that an in vivo 
bioequivalence study is required.
    (d) For certain drug products, bioavailability or bioequivalence may 
be demonstrated by evidence obtained in vitro in lieu of in vivo data. 
FDA shall waive the requirement for the submission of evidence obtained 
in vivo demonstrating the bioavailability of the drug product if the 
drug product meets one of the following criteria:
    (1) [Reserved]
    (2) The drug product is in the same dosage form, but in a different 
strength, and is proportionally similar

[[Page 194]]

in its active and inactive ingredients to another drug product for which 
the same manufacturer has obtained approval and the conditions in 
paragraphs (d)(2)(i) through (d)(2)(iii) of this section are met:
    (i) The bioavailability of this other drug product has been 
demonstrated;
    (ii) Both drug products meet an appropriate in vitro test approved 
by FDA; and
    (iii) The applicant submits evidence showing that both drug products 
are proportionally similar in their active and inactive ingredients.
    (iv) This subparagraph does not apply to enteric coated or 
controlled release dosage forms.
    (3) The drug product is, on the basis of scientific evidence 
submitted in the application, shown to meet an in vitro test that has 
been correlated with in vivo data.
    (4) The drug product is a reformulated product that is identical, 
except for a different color, flavor, or preservative that could not 
affect the bioavailability of the reformulated product, to another drug 
product for which the same manufacturer has obtained approval and the 
following conditions are met:
    (i) The bioavailability of the other product has been demonstrated; 
and
    (ii) Both drug products meet an appropriate in vitro test approved 
by FDA.
    (e) FDA, for good cause, may waive a requirement for the submission 
of evidence of in vivo bioavailability if waiver is compatible with the 
protection of the public health. For full new drug applications, FDA may 
defer a requirement for the submission of evidence of in vivo 
bioavailability if deferral is compatible with the protection of the 
public health.
    (f) FDA, for good cause, may require evidence of in vivo 
bioavailability or bioequivalence for any drug product if the agency 
determines that any difference between the drug product and a listed 
drug may affect the bioavailability or bioequivalence of the drug 
product.
[57 FR 17998, Apr. 28, 1992]



Sec. 320.23  Basis for demonstrating in vivo bioavailability or bioequivalence.

    (a)(1) The in vivo bioavailability of a drug product is demonstrated 
if the product's rate and extent of absorption, as determined by 
comparison of measured parameters, e.g., concentration of the active 
drug ingredient in the blood, urinary excretion rates, or 
pharmacological effects, do not indicate a significant difference from 
the reference material's rate and extent of absorption. For drug 
products that are not intended to be absorbed into the bloodstream, 
bioavailability may be assessed by measurements intended to reflect the 
rate and extent to which the active ingredient or active moiety becomes 
available at the site of action.
    (2) Statistical techniques used shall be of sufficient sensitivity 
to detect differences in rate and extent of absorption that are not 
attributable to subject variability.
    (3) A drug product that differs from the reference material in its 
rate of absorption, but not in its extent of absorption, may be 
considered to be bioavailable if the difference in the rate of 
absorption is intentional, is appropriately reflected in the labeling, 
is not essential to the attainment of effective body drug concentrations 
on chronic use, and is considered medically insignificant for the drug 
product.
    (b) Two drug products will be considered bioequivalent drug products 
if they are pharmaceutical equivalents or pharmaceutical alternatives 
whose rate and extent of absorption do not show a significant difference 
when administered at the same molar dose of the active moiety under 
similar experimental conditions, either single dose or multiple dose. 
Some pharmaceutical equivalents or pharmaceutical alternatives may be 
equivalent in the extent of their absorption but not in their rate of 
absorption and yet may be considered bioequivalent because such 
differences in the rate of absorption are intentional and are reflected 
in the labeling, are not essential to the attainment of effective body 
drug concentrations on chronic use, and are considered medically 
insignificant for the particular drug product studied.
[57 FR 17999, Apr. 28, 1992]

[[Page 195]]



Sec. 320.24  Types of evidence to establish bioavailability or bioequivalence.

    (a) Bioavailability or bioequivalence may be determined by several 
in vivo and in vitro methods. FDA may require in vivo or in vitro 
testing, or both, to establish the bioavailability of a drug product or 
the bioequivalence of specific drug products. Information on 
bioequivalence requirements for specific products is included in the 
current edition of FDA's publication ``Approved Drug Products with 
Therapeutic Equivalence Evaluations'' and any current supplement to the 
publication. The selection of the method used to meet an in vivo or in 
vitro testing requirement depends upon the purpose of the study, the 
analytical methods available, and the nature of the drug product. 
Applicants shall conduct bioavailability and bioequivalence testing 
using the most accurate, sensitive, and reproducible approach available 
among those set forth in paragraph (b) of this section. The method used 
must be capable of demonstrating bioavailability or bioequivalence, as 
appropriate, for the product being tested.
    (b) The following in vivo and in vitro approaches, in descending 
order of accuracy, sensitivity, and reproducibility, are acceptable for 
determining the bioavailability or bioequivalence of a drug product.
    (1)(i) An in vivo test in humans in which the concentration of the 
active ingredient or active moiety, and, when appropriate, its active 
metabolite(s), in whole blood, plasma, serum, or other appropriate 
biological fluid is measured as a function of time. This approach is 
particularly applicable to dosage forms intended to deliver the active 
moiety to the bloodstream for systemic distribution within the body; or
    (ii) An in vitro test that has been correlated with and is 
predictive of human in vivo bioavailability data; or
    (iii) An in vivo test in animals that has been correlated with and 
is predictive of human bioavailability data.
    (2) An in vivo test in humans in which the urinary excretion of the 
active moiety, and, when appropriate, its active metabolite(s), are 
measured as a function of time. The intervals at which measurements are 
taken should ordinarily be as short as possible so that the measure of 
the rate of elimination is as accurate as possible. Depending on the 
nature of the drug product, this approach may be applicable to the 
category of dosage forms described in paragraph (b)(1)(i) of this 
section. This method is not appropriate where urinary excretion is not a 
significant mechanism of elimination.
    (3) An in vivo test in humans in which an appropriate acute 
pharmacological effect of the active moiety, and, when appropriate, its 
active metabolite(s), are measured as a function of time if such effect 
can be measured with sufficient accuracy, sensitivity, and 
reproducibility. This approach is applicable to the category of dosage 
forms described in paragraph (b)(1)(i) of this section only when 
appropriate methods are not available for measurement of the 
concentration of the moiety, and, when appropriate, its active 
metabolite(s), in biological fluids or excretory products but a method 
is available for the measurement of an appropriate acute pharmacological 
effect. This approach may be particularly applicable to dosage forms 
that are not intended to deliver the active moiety to the bloodstream 
for systemic distribution.
    (4) Well-controlled clinical trials in humans that establish the 
safety and effectiveness of the drug product, for purposes of 
establishing bioavailability, or appropriately designed comparative 
clinical trials, for purposes of demonstrating bioequivalence. This 
approach is the least accurate, sensitive, and reproducible of the 
general approaches for determining bioavailability or bioequivalence. 
For dosage forms intended to deliver the active moiety to the 
bloodstream for systemic distribution, this approach may be considered 
acceptable only when analytical methods cannot be developed to permit 
use of one of the approaches outlined in paragraphs (b)(1)(i) and (b)(2) 
of this section, when the approaches described in paragraphs (b)(1)(ii), 
(b)(1)(iii), and (b)(3) of this section are not available. This approach 
may also be considered sufficiently accurate for determining the 
bioavailability or bioequivalence of dosage forms intended to deliver 
the

[[Page 196]]

active moiety locally, e.g., topical preparations for the skin, eye, and 
mucous membranes; oral dosage forms not intended to be absorbed, e.g., 
an antacid or radiopaque medium; and bronchodilators administered by 
inhalation if the onset and duration of pharmacological activity are 
defined.
    (5) A currently available in vitro test acceptable to FDA (unusually 
a dissolution rate test) that ensures human in vivo bioavailability.
    (6) Any other approach deemed adequate by FDA to establish 
bioavailability or bioequivalence.
    (c) FDA may, notwithstanding prior requirements for establishing 
bioavailability or bioequivalence, require in vivo testing in humans of 
a product at any time if the agency has evidence that the product:
    (1) May not produce therapeutic effects comparable to a 
pharmaceutical equivalent or alternative with which it is intended to be 
used interchangeably;
    (2) May not be bioequivalent to a pharmaceutical equivalent or 
alternative with which it is intended to be used interchangeably; or
    (3) Has greater than anticipated potential toxicity related to 
pharmacokinetic or other characteristics.
[57 FR 17999, Apr. 28, 1992; 57 FR 29354, July 1, 1992]



Sec. 320.25  Guidelines for the conduct of an in vivo bioavailability study.

    (a) Guiding principles. (1) The basic principle in an in vivo 
bioavailability study is that no unnecessary human research should be 
done.
    (2) An in vivo bioavailability study shall not be conducted in 
humans if an appropriate animal model exists and correlation of results 
in animals and humans has been demonstrated. If an appropriate animal 
model does not exist, however, an in vivo bioavailability study shall 
ordinarily be done in normal adults under standardized conditions.
    (3) In some situations, an in vivo bioavailability study in humans 
may preferably and more properly be done in suitable patients. 
Critically ill patients shall not be included in an in vivo 
bioavailability study unless the attending physician determines that 
there is a potential benefit to the patient.
    (b) Basic design. The basic design of an in vivo bioavailability 
study is determined by the following:
    (1) The scientific questions to be answered.
    (2) The nature of the reference material and the dosage form to be 
tested.
    (3) The availability of analytical methods.
    (4) Benefit-risk considerations in regard to testing in humans.
    (c) Comparison to a reference material. In vivo bioavailability 
testing of a drug product shall be in comparison to an appropriate 
reference material unless some other approach is more appropriate for 
valid scientific reasons.
    (d) Previously unmarketed active drug ingredients or therapeutic 
moieties. (1) The purpose of an in vivo bioavailability study involving 
a drug product containing an active drug ingredient or therapeutic 
moiety that has not been approved for marketing is to determine:
    (i) The bioavailability of the formulation proposed for marketing; 
and
    (ii) The essential pharmacokinetic characteristics of the active 
drug ingredient or therapeutic moiety, such as the rate of absorption, 
the extent of absorption, the half-life of the therapeutic moiety in 
vivo, and the rate of excretion and/or metabolism. Dose proportionality 
of the active drug ingredient or the therapeutic moiety needs to be 
established after single-dose administration and in certain instances 
after multiple-dose administration. This characterization is a necessary 
part of the investigation of the drug to support drug labeling.
    (2) The reference material in such a bioavailability study should be 
a solution or suspension containing the same quantity of the active drug 
ingredient or therapeutic moiety as the formulation proposed for 
marketing.
    (3) The reference material should be administered by the same route 
as the formulation proposed for marketing unless an alternative or 
additional route is necessary to answer the scientific question under 
study. For example, in the case of an active drug ingredient or 
therapeutic moiety that is poorly absorbed after oral administration, it 
may be necessary to compare

[[Page 197]]

the oral dosage form proposed for marketing with the active drug 
ingredient or therapeutic moiety administered in solution both orally 
and intravenously.
    (e) New formulations of active drug ingredients or therapeutic 
moieties approved for marketing. (1) The purpose of an in vivo 
bioavailability study involving a drug product that is a new 
formulation, a new dosage form, or a new salt or ester of an active drug 
ingredient or therapeutic moiety that has been approved for marketing is 
to:
    (i) Determine the bioavailability of the new formulation, new dosage 
form, or new salt or ester relative to an appropriate reference 
material; and
    (ii) Define the pharmacokinetic parameters of the new formulation, 
new dosage form, or new salt or ester to establish dosage 
recommendation.
    (2) The selection of the reference material(s) in such a 
bioavailability study depends upon the scientific questions to be 
answered, the data needed to establish comparability to a currently 
marketed drug product, and the data needed to establish dosage 
recommendations.
    (3) The reference material should be taken from a current batch of a 
drug product that is the subject of an approved new drug application and 
that contains the same active drug ingredient or therapeutic moiety, if 
the new formulation, new dosage form, or new salt or ester is intended 
to be comparable to or to meet any comparative labeling claims made in 
relation to the drug product that is the subject of an approved new drug 
application.
    (f) Controlled release formulations. (1) The purpose of an in vivo 
bioavailability study involving a drug product for which a controlled 
release claim is made is to determine if all of the following conditions 
are met:
    (i) The drug product meets the controlled release claims made for 
it.
    (ii) The bioavailability profile established for the drug product 
rules out the occurrence of any dose dumping.
    (iii) The drug product's steady-state performance is equivalent to a 
currently marketed noncontrolled release or controlled release drug 
product that contains the same active drug ingredient or therapeutic 
moiety and that is subject to an approved full new drug application.
    (iv) The drug product's formulation provides consistent 
pharmacokinetic performance between individual dosage units.
    (2) The reference material(s) for such a bioavailability study shall 
be chosen to permit an appropriate scientific evaluation of the 
controlled release claims made for the drug product. The reference 
material shall be one of the following or any combination thereof:
    (i) A solution or suspension of the active drug ingredient or 
therapeutic moiety.
    (ii) A currently marketed noncontrolled release drug product 
containing the same active drug ingredient or therapeutic moiety and 
administered according to the dosage recommendations in the labeling of 
the noncontrolled release drug product.
    (iii) A currently marketed controlled release drug product subject 
to an approved full new drug application containing the same active drug 
ingredient or therapeutic moiety and administered according to the 
dosage recommendations in the labeling proposed for the controlled 
release drug product.
    (iv) A reference material other than one set forth in paragraph 
(f)(2) (i), (ii) or (iii) of this section that is appropriate for valid 
scientific reasons.
    (g) Combination drug products. (1) Generally, the purpose of an in 
vivo bioavailability study involving a combination drug product is to 
determine if the rate and extent of absorption of each active drug 
ingredient or therapeutic moiety in the combination drug product is 
equivalent to the rate and extent of absorption of each active drug 
ingredient or therapeutic moiety administered concurrently in separate 
single-ingredient preparations.
    (2) The reference material in such a bioavailability study should be 
two or more currently marketed, single-ingredient drug products each of 
which contains one of the active drug ingredients or therapeutic 
moieties in the combination drug product. The Food and Drug 
Administration may, for valid scientific reasons, specify that the 
reference material shall be a combination drug product that is the 
subject of an approved new drug application.

[[Page 198]]

    (3) The Food and Drug Administration may permit a bioavailability 
study involving a combination drug product to determine the rate and 
extent of absorption of selected, but not all, active drug ingredients 
or therapeutic moieties in the combination drug product. The Food and 
Drug Administration may permit this determination if the 
pharmacokinetics and the interactions of the active drug ingredients or 
therapeutic moieties in the combination drug product are well known and 
the therapeutic activity of the combination drug product is generally 
recognized to reside in only one of the active drug ingredients or 
therapeutic moieties, e.g., ampicillin in an ampicillin-probenecid 
combination drug product.
    (h) Use of a placebo as the reference material. Where appropriate or 
where necessary to demonstrate the sensitivity of the test, the 
reference material in a bioavailability study may be a placebo if:
    (1) The study measures the therapeutic or acute pharmacological 
effect of the active drug ingredient or therapeutic moiety; or
    (2) The study is a clinical trial to establish the safety and 
effectiveness of the drug product.
    (i) Standards for test drug product and reference material. (1) Both 
the drug product to be tested and the reference material, if it is 
another drug product, shall be shown to meet all compendial or other 
applicable standards of identity, strength, quality, and purity, 
including potency and, where applicable, content uniformity, 
disintegration times, and dissolution rates.
    (2) Samples of the drug product to be tested shall be manufactured 
using the same equipment and under the same conditions as those used for 
full-scale production.



Sec. 320.26  Guidelines on the design of a single-dose in vivo bioavailability study.

    (a) Basic principles. (1) An in vivo bioavailability study should be 
a single-dose comparison of the drug product to be tested and the 
appropriate reference material conducted in normal adults.
    (2) The test product and the reference material should be 
administered to subjects in the fasting state, unless some other 
approach is more appropriate for valid scientific reasons.
    (b) Study design. (1) A single-dose study should be crossover in 
design, unless a parallel design or other design is more appropriate for 
valid scientific reasons, and should provide for a drug elimination 
period.
    (2) Unless some other approach is appropriate for valid scientific 
reasons, the drug elimination period should be either:
    (i) At least three times the half-life of the active drug ingredient 
or therapeutic moiety, or its metabolite(s), measured in the blood or 
urine; or
    (ii) At least three times the half-life of decay of the acute 
pharmacological effect.
    (c) Collection of blood samples. (1) When comparison of the test 
product and the reference material is to be based on blood concentration 
time curves, unless some other approach is more appropriate for valid 
scientific reasons, blood samples should be taken with sufficient 
frequency to permit an estimate of both:
    (i) The peak concentration in the blood of the active drug 
ingredient or therapeutic moiety, or its metabolite(s), measured; and
    (ii) The total area under the curve for a time period at least three 
times the half-life of the active drug ingredient or therapeutic moiety, 
or its metabolite(s), measured.
    (2) In a study comparing oral dosage forms, the sampling times 
should be identical.
    (3) In a study comparing an intravenous dosage form and an oral 
dosage form, the sampling times should be those needed to describe both:
    (i) The distribution and elimination phase of the intravenous dosage 
form; and
    (ii) The absorption and elimination phase of the oral dosage form.
    (4) In a study comparing drug delivery systems other than oral or 
intravenous dosage forms with an appropriate reference standard, the 
sampling times should be based on valid scientific reasons.
    (d) Collection of urine samples. When comparison of the test product 
and the

[[Page 199]]

reference material is to be based on cumulative urinary excretion-time 
curves, unless some other approach is more appropriate for valid 
scientific reasons, samples of the urine should be collected with 
sufficient frequency to permit an estimate of the rate and extent of 
urinary excretion of the active drug ingredient or therapeutic moiety, 
or its metabolite(s), measured.
    (e) Measurement of an acute pharmacological effect. (1) When 
comparison of the test product and the reference material is to be based 
on acute pharmacological effect-time curves, measurements of this effect 
should be made with sufficient frequency to permit a reasonable estimate 
of the total area under the curve for a time period at least three times 
the half-life of decay of the pharmacological effect, unless some other 
approach is more appropriate for valid scientific reasons.
    (2) The use of an acute pharmacological effect to determine 
bioavailability may further require demonstration of dose-related 
response. In such a case, bioavailability may be determined by 
comparison of the dose-response curves as well as the total area under 
the acute pharmacological effect-time curves for any given dose.



Sec. 320.27  Guidelines on the design of a multiple-dose in vivo bioavailability study.

    (a) Basic principles. (1) In selected circumstances it may be 
necessary for the test product and the reference material to be compared 
after repeated administration to determine steady-state levels of the 
active drug ingredient or therapeutic moiety in the body.
    (2) The test product and the reference material should be 
administered to subjects in the fasting or nonfasting state, depending 
upon the conditions reflected in the proposed labeling of the test 
product.
    (3) A multiple-dose study may be required to determine the 
bioavailability of a drug product in the following circumstances:
    (i) There is a difference in the rate of absorption but not in the 
extent of absorption.
    (ii) There is excessive variability in bioavailability from subject 
to subject.
    (iii) The concentration of the active drug ingredient or therapeutic 
moiety, or its metabolite(s), in the blood resulting from a single dose 
is too low for accurate determination by the analytical method.
    (iv) The drug product is a controlled release dosage form.
    (b) Study design. (1) A multiple-dose study should be crossover in 
design, unless a parallel design or other design is more appropriate for 
valid scientific reasons, and should provide for a drug elimination 
period if steady-state conditions are not achieved.
    (2) A multiple-dose study is not required to be of crossover design 
if the study is to establish dose proportionality under a multiple-dose 
regimen or to establish the pharmacokinetic profile of a new drug 
product, a new drug delivery system, or a controlled release dosage 
form.
    (3) If a drug elimination period is required, unless some other 
approach is more appropriate for valid scientific reasons, the drug 
elimination period should be either:
    (i) At least five times the half-life of the active drug ingredient 
or therapeutic moiety, or its metabolite(s), measured in the blood or 
urine; or
    (ii) At least five times the half-life of decay of the acute 
pharmacological effect.
    (c) Achievement of steady-state conditions. Whenever a multiple-dose 
study is conducted, unless some other approach is more appropriate for 
valid scientific reasons, sufficient doses of the test product and 
reference material should be administered in accordance with the 
labeling to achieve steady-state conditions.
    (d) Collection of blood or urine samples. (1) Whenever comparison of 
the test product and the reference material is to be based on blood 
concentration-time curves at steady-state, sufficient samples of blood 
should be taken to define adequately the maximum (Cmax) and minimum 
(Cmin) blood concentrations on 2 or more consecutive days to establish 
that steady-state conditions are achieved.
    (2) Whenever comparison of the test product and the reference 
material is to be based on cumulative urinary excretion-time curves at 
steady-state,

[[Page 200]]

sufficient samples of urine should be taken to define the rate and 
extent of urinary excretion on 2 or more consecutive days to establish 
that steady-state conditions are achieved.
    (3) A more complete characterization of the blood concentration or 
urinary excretion rate during the absorption and elimination phases of a 
single dose administered at steady-state is encouraged to permit 
estimation of the total area under concentration-time curves or 
cumulative urinary excretion-time curves and to obtain pharmacokinetic 
information, e.g., half-life or blood clearance, that is essential in 
preparing adequate labeling for the drug product.
    (e) Steady-state parameters. (1) In certain instances, e.g., in a 
study involving a new drug entity, blood clearances at steady-state 
obtained in a multiple-dose study should be compared to blood clearances 
obtained in a single-dose study to support adequate dosage 
recommendations.
    (2) In a linear system, the area under the blood concentration-time 
curve during a dosing interval in a multiple-dose steady-state study is 
directly proportional to the fraction of the dose absorbed and is equal 
to the corresponding ``zero to infinity'' area under the curve for a 
single-dose study. Therefore, when steady-state conditions are achieved, 
a comparison of blood concentrations during a dosing interval may be 
used to define the fraction of the active drug ingredient or therapeutic 
moiety absorbed.
    (3) Other methods based on valid scientific reasons should be used 
to determine the bioavailability of a drug product having dose-dependent 
kinetics (non-linear system).
    (f) Measurement of an acute pharmacological effect. When comparison 
of the test product and the reference material is to be based on acute 
pharmacological effect-time curves, measurements of this effect should 
be made with sufficient frequency to demonstrate a maximum effect and a 
lack of significant difference between the test product and the 
reference material.



Sec. 320.28  Correlation of bioavailability with an acute pharmacological effect or clinical evidence.

    Correlation of in vivo bioavailability data with an acute 
pharmacological effect or clinical evidence of safety and effectiveness 
may be required if needed to establish the clinical significance of a 
special claim, e.g., in the case of a controlled release preparation.



Sec. 320.29  Analytical methods for an in vivo bioavailability study.

    (a) The analytical method used in an in vivo bioavailability study 
to measure the concentration of the active drug ingredient or 
therapeutic moiety, or its metabolite(s), in body fluids or excretory 
products, or the method used to measure an acute pharmacological effect 
shall be demonstrated to be accurate and of sufficient sensitivity to 
measure, with appropriate precision, the actual concentration of the 
active drug ingredient or therapeutic moiety, or its metabolite(s), 
achieved in the body.
    (b) When the analytical method is not sensitive enough to measure 
accurately the concentration of the active drug ingredient or 
therapeutic moiety, or its metabolite(s), in body fluids or excretory 
products produced by a single dose of the test product, two or more 
single doses may be given together to produce higher concentration if 
the requirements of Sec. 320.31 are met.



Sec. 320.30  Inquiries regarding bioavailability and bioequivalence requirements and review of protocols by the Food and Drug Administration.

    (a) The Commissioner of Food and Drugs strongly recommends that, to 
avoid the conduct of an improper study and unnecessary human research, 
any person planning to conduct a bioavailability or bioequivalence study 
submit the proposed protocol for the study to FDA for review prior to 
the initiation of the study.
    (b) FDA may review a proposed protocol for a bioavailability or 
bioequivalence study and will offer advice with respect to whether the 
following conditions are met:

[[Page 201]]

    (1) The design of the proposed bioavailability or bioequivalence 
study is appropriate.
    (2) The reference material to be used in the bioavailability or 
bioequivalence study is appropriate.
    (3) The proposed chemical and statistical analytical methods are 
adequate.
    (c)(1) General inquiries relating to in vivo bioavailability 
requirements and methodology shall be submitted to the Food and Drug 
Administration, Center for Drug Evaluation and Research, Division of 
Biopharmaceutics (HFD-420), 5600 Fishers Lane, Rockville, MD 20857.
    (2) General inquiries relating to bioequivalence requirements and 
methodology shall be submitted to the Food and Drug Administration, 
Center for Drug Evaluation and Research, Division of Bioequivalence 
(HFD-650), 5600 Fishers Lane, Rockville, MD 20857.
[57 FR 18000, Apr. 28, 1992]



Sec. 320.31  Applicability of requirements regarding an ``Investigational New Drug Application.''

    (a) Any person planning to conduct an in vivo bioavailability or 
bioequivalence study in humans shall submit an ``Investigational New 
Drug Application'' (IND) if:
    (1) The test product contains a new chemical entity as defined in 
Sec. 314.108(a) of this chapter; or
    (2) The study involves a radioactively labeled drug product; or
    (3) The study involves a cytotoxic drug product.
    (b) Any person planning to conduct a bioavailability study in humans 
using a drug product that contains an already approved, non-new chemical 
entity shall submit an IND if the study is one of the following:
    (1) A single-dose study in normal subjects or patients where either 
the maximum single or total daily dose exceeds that specified in the 
labeling of the drug product that is the subject of an approved new drug 
application or abbreviated new drug application.
    (2) A multiple-dose study in normal subjects or patients where 
either the single or total daily dose exceeds that specified in the 
labeling of the drug product that is the subject of an approved new drug 
application or abbreviated new drug application.
    (3) A multiple-dose study on a controlled release product on which 
no single-dose study has been completed.
    (c) The provisions of parts 50, 56, and 312 of this chapter are 
applicable to any bioavailability or bioequivalence study in humans 
conducted under an IND.
    (d) A bioavailability or bioequivalence study in humans other than 
one described in paragraphs (a) through (c) of this section is exempt 
from the requirements of part 312 of this chapter if the following 
conditions are satisfied:
    (1) If the study is one described under Sec. 320.38(b) or 
Sec. 320.63, the person conducting the study, including any contract 
research organization, shall retain reserve samples of any test article 
and reference standard used in the study and release the reserve samples 
to FDA upon request, in accordance with, and for the period specified 
in, Sec. 320.38; and
    (2) An in vivo bioavailability or bioequivalence study in humans 
shall be conducted in compliance with the requirements for institutional 
review set forth in part 56 of this chapter, and informed consent set 
forth in part 50 of this chapter.
[57 FR 18000, Apr. 28, 1992, as amended at 58 FR 25927, Apr. 28, 1993]



Sec. 320.32  Procedures for establishing or amending a bioequivalence requirement.

    (a) The Food and Drug Administration, on its own initiative or in 
response to a petition by an interested person, may propose and 
promulgate a regulation to establish a bioequivalence requirement for a 
product not subject to section 505(j) of the act if it finds there is 
well-documented evidence that specific pharmaceutical equivalents or 
pharmaceutical alternatives intended to be used interchangeably for the 
same therapeutic effect:
    (1) Are not bioequivalent drug products; or
    (2) May not be bioequivalent drug products based on the criteria set 
forth in Sec. 320.33; or
    (3) May not be bioequivalent drug products because they are members 
of a class of drug products that have close

[[Page 202]]

structural similarity and similar physicochemical or pharmacokinetic 
properties to other drug products in the same class that FDA finds are 
not bioequivalent drug products.
    (b) FDA shall include in a proposed rule to establish a 
bioequivalence requirement the evidence and criteria set forth in 
Sec. 320.33 that are to be considered in determining whether to issue 
the proposal. If the rulemaking is proposed in response to a petition, 
FDA shall include in the proposal a summary and analysis of the relevant 
information that was submitted in the petition as well as other 
available information to support the establishment of a bioequivalence 
requirement.
    (c) FDA, on its own initiative or in response to a petition by an 
interested person, may propose and promulgate an amendment to a 
bioequivalence requirement established under this subpart.
[57 FR 18000, Apr. 28, 1992]



Sec. 320.33  Criteria and evidence to assess actual or potential bioequivalence problems.

    The Commissioner of Food and Drugs shall consider the following 
factors, when supported by well-documented evidence, to identify 
specific pharmaceutical equivalents and pharmaceutical alternatives that 
are not or may not be bioequivalent drug products.
    (a) Evidence from well-controlled clinical trials or controlled 
observations in patients that such drug products do not give comparable 
therapeutic effects.
    (b) Evidence from well-controlled bioequivalence studies that such 
products are not bioequivalent drug products.
    (c) Evidence that the drug products exhibit a narrow therapeutic 
ratio, e.g., there is less than a 2-fold difference in median lethal 
dose (LD50) and median effective dose (ED50) 
values, or have less than a 2-fold difference in the minimum toxic 
concentrations and minimum effective concentrations in the blood, and 
safe and effective use of the drug products requires careful dosage 
titration and patient monitoring.
    (d) Competent medical determination that a lack of bioequivalence 
would have a serious adverse effect in the treatment or prevention of a 
serious disease or condition.
    (e) Physicochemical evidence that:
    (1) The active drug ingredient has a low solubility in water, e.g., 
less than 5 milligrams per 1 milliliter, or, if dissolution in the 
stomach is critical to absorption, the volume of gastric fluids required 
to dissolve the recommended dose far exceeds the volume of fluids 
present in the stomach (taken to be 100 milliliters for adults and 
prorated for infants and children).
    (2) The dissolution rate of one or more such products is slow, e.g., 
less than 50 percent in 30 minutes when tested using either a general 
method specified in an official compendium or a paddle method at 50 
revolutions per minute in 900 milliliters of distilled or deionized 
water at 37 deg. C, or differs significantly from that of an appropriate 
reference material such as an identical drug product that is the subject 
of an approved full new drug application.
    (3) The particle size and/or surface area of the active drug 
ingredient is critical in determining its bioavailability.
    (4) Certain physical structural characteristics of the active drug 
ingredient, e.g., polymorphic forms, conforms, solvates, complexes, and 
crystal modifications, dissolve poorly and this poor dissolution may 
affect absorption.
    (5) Such drug products have a high ratio of excipients to active 
ingredients, e.g., greater than 5 to 1.
    (6) Specific inactive ingredients, e.g., hydrophilic or hydrophobic 
excipients and lubricants, either may be required for absorption of the 
active drug ingredient or therapeutic moiety or, alternatively, if 
present, may interfere with such absorption.
    (f) Pharmacokinetic evidence that:
    (1) The active drug ingredient, therapeutic moiety, or its precursor 
is absorbed in large part in a particular segment of the 
gastrointestinal tract or is absorbed from a localized site.
    (2) The degree of absorption of the active drug ingredient, 
therapeutic moiety, or its precursor is poor, e.g., less than 50 
percent, ordinarily in comparison to an intravenous dose, even when

[[Page 203]]

it is administered in pure form, e.g., in solution.
    (3) There is rapid metabolism of the therapeutic moiety in the 
intestinal wall or liver during the process of absorption (first-class 
metabolism) so the therapeutic effect and/or toxicity of such drug 
product is determined by the rate as well as the degree of absorption.
    (4) The therapeutic moiety is rapidly metabolized or excreted so 
that rapid dissolution and absorption are required for effectiveness.
    (5) The active drug ingredient or therapeutic moiety is unstable in 
specific portions of the gastrointestinal tract and requires special 
coatings or formulations, e.g., buffers, enteric coatings, and film 
coatings, to assure adequate absorption.
    (6) The drug product is subject to dose dependent kinetics in or 
near the therapeutic range, and the rate and extent of absorption are 
important to bioequivalence.
[42 FR 1635, Jan. 7, 1977. Redesignated and amended at 57 FR 18001, Apr. 
28, 1992]



Sec. 320.34  Requirements for batch testing and certification by the Food and Drug Administration.

    (a) If the Commissioner determines that individual batch testing by 
the Food and Drug Administration is necessary to assure that all batches 
of the same drug product meet an appropriate in vitro test, he shall 
include in the bioequivalence requirement a requirement for 
manufacturers to submit samples of each batch to the Food and Drug 
Administration and to withhold distribution of the batch until notified 
by the Food and Drug Administration that the batch may be introduced 
into interstate commerce.
    (b) The Commissioner will ordinarily terminate a requirement for a 
manufacturer to submit samples for batch testing on a finding that the 
manufacturer has produced four consecutive batches that were tested by 
the Food and Drug Administration and found to meet the bioequivalence 
requirement, unless the public health requires that batch testing be 
extended to additional batches.
[42 FR 1635, Jan. 7, 1977. Redesignated at 57 FR 18001, Apr. 28, 1992]



Sec. 320.35  Requirements for in vitro testing of each batch.

    If a bioequivalence requirement specifies a currently available in 
vitro test or an in vitro bioequivalence standard comparing the drug 
product to a reference standard, the manufacturer shall conduct the test 
on a sample of each batch of the drug product to assure batch-to-batch 
uniformity.
[42 FR 1635, Jan. 7, 1977. Redesignated at 57 FR 18001, Apr. 28, 1992]



Sec. 320.36  Requirements for maintenance of records of bioequivalence testing.

    (a) All records of in vivo or in vitro tests conducted on any 
marketed batch of a drug product to assure that the product meets a 
bioequivalence requirement shall be maintained by the manufacturer for 
at least 2 years after the expiration date of the batch and submitted to 
the Food and Drug Administration on request.
    (b) Any person who contracts with another party to conduct a 
bioequivalence study from which the data are intended to be submitted to 
FDA as part of an application submitted under part 314 of this chapter 
shall obtain from the person conducting the study sufficient accurate 
financial information to allow the submission of complete and accurate 
financial certifications or disclosure statements required under part 54 
of this chapter and shall maintain that information and all records 
relating to the compensation given for that study and all other 
financial interest information required under part 54 of this chapter 
for 2 years after the date of approval of the application. The person 
maintaining these records shall, upon request for any properly 
authorized officer or employee of the Food and Drug Administration, at 
reasonable time, permit such officer or employee to have access to and 
copy and verify these records.
[42 FR 1635, Jan. 7, 1977. Redesignated at 57 FR 18001, Apr. 28, 1992, 
as amended at 63 FR 5252, Feb. 2, 1998]

    Effective Date Note: At 63 FR 5252, Feb. 2, 1998, Sec. 320.36 was 
amended by designating the existing text as paragraph (a) and by adding 
new paragraph (b), effective Feb. 2, 1999.

[[Page 204]]



Sec. 320.38  Retention of bioavailability samples.

    (a) The applicant of an application or supplemental application 
submitted under section 505 or 507 of the Federal Food, Drug, and 
Cosmetic Act, or, if bioavailability testing was performed under 
contract, the contract research organization shall retain an 
appropriately identified reserve sample of the drug product for which 
the applicant is seeking approval (test article) and of the reference 
standard used to perform an in vivo bioavailability study in accordance 
with and for the studies described in paragraph (b) of this section that 
is representative of each sample of the test article and reference 
standard provided by the applicant for the testing.
    (b) Reserve samples shall be retained for the following test 
articles and reference standards and for the studies described:
    (1) If the formulation of the test article is the same as the 
formulation(s) used in the clinical studies demonstrating substantial 
evidence of safety and effectiveness for the test article's claimed 
indications, a reserve sample of the test article used to conduct an in 
vivo bioavailability study comparing the test article to a reference 
oral solution, suspension, or injection.
    (2) If the formulation of the test article differs from the 
formulation(s) used in the clinical studies demonstrating substantial 
evidence of safety and effectiveness for the test article's claimed 
indications, a reserve sample of the test article and of the reference 
standard used to conduct an in vivo bioequivalence study comparing the 
test article to the formulation(s) (reference standard) used in the 
clinical studies.
    (3) For a new formulation, new dosage form, or a new salt or ester 
of an active drug ingredient or therapeutic moiety that has been 
approved for marketing, a reserve sample of the test article and of the 
reference standard used to conduct an in vivo bioequivalence study 
comparing the test article to a marketed product (reference standard) 
that contains the same active drug ingredient or therapeutic moiety.
    (c) Each reserve sample shall consist of a sufficient quantity to 
permit FDA to perform five times all of the release tests required in 
the application or supplemental application.
    (d) Each reserve sample shall be adequately identified so that the 
reserve sample can be positively identified as having come from the same 
sample as used in the specific bioavailability study.
    (e) Each reserve sample shall be stored under conditions consistent 
with product labeling and in an area segregated from the area where 
testing is conducted and with access limited to authorized personnel. 
Each reserve sample shall be retained for a period of at least 5 years 
following the date on which the application or supplemental application 
is approved, or, if such application or supplemental application is not 
approved, at least 5 years following the date of completion of the 
bioavailability study in which the sample from which the reserve sample 
was obtained was used.
    (f) Authorized FDA personnel will ordinarily collect reserve samples 
directly from the applicant or contract research organization at the 
storage site during a preapproval inspection. If authorized FDA 
personnel are unable to collect samples, FDA may require the applicant 
or contract research organization to submit the reserve samples to the 
place identified in the agency's request. If FDA has not collected or 
requested delivery of a reserve sample, or if FDA has not collected or 
requested delivery of any portion of a reserve sample, the applicant or 
contract research organization shall retain the sample or remaining 
sample for the 5-year period specified in paragraph (e) of this section.
    (g) Upon release of the reserve samples to FDA, the applicant or 
contract research organization shall provide a written assurance that, 
to the best knowledge and belief of the individual executing the 
assurance, the reserve samples came from the same samples as used in the 
specific bioavailability or bioequivalence study identified by the 
agency. The assurance shall be executed by an individual authorized to 
act for the applicant or contract research organization in releasing the 
reserve samples to FDA.

[[Page 205]]

    (h) A contract research organization may contract with an 
appropriate, independent third party to provide storage of reserve 
samples provided that the sponsor of the study has been notified in 
writing of the name and address of the facility at which the reserve 
samples will be stored.
    (i) If a contract research organization conducting a bioavailability 
or bioequivalence study that requires reserve sample retention under 
this section or Sec. 320.63 goes out of business, it shall transfer its 
reserve samples to an appropriate, independent third party, and shall 
notify in writing the sponsor of the study of the transfer and provide 
the study sponsor with the name and address of the facility to which the 
reserve samples have been transferred.
[58 FR 25927, Apr. 28, 1993]



Sec. 320.63  Retention of bioequivalence samples.

    The applicant of an abbreviated application or a supplemental 
application submitted under section 505 or 507 of the Federal Food, 
Drug, and Cosmetic Act, or, if bioequivalence testing was performed 
under contract, the contract research organization shall retain reserve 
samples of any test article and reference standard used in conducting an 
in vivo or in vitro bioequivalence study required for approval of the 
abbreviated application or supplemental application. The applicant or 
contract research organization shall retain the reserve samples in 
accordance with, and for the period specified in, Sec. 320.38 and shall 
release the reserve samples to FDA upon request in accordance with 
Sec. 320.38.
[58 FR 25928, Apr. 28, 1993]



PART 328--OVER-THE-COUNTER DRUG PRODUCTS INTENDED FOR ORAL INGESTION THAT CONTAIN ALCOHOL--Table of Contents




                      Subpart A--General Provisions

Sec.
328.1  Scope.
328.3  Definitions.

                         Subpart B--Ingredients

328.10  Alcohol.

                           Subpart C--Labeling

328.50  Principal display panel of all OTC drug products intended for 
          oral ingestion that contain alcohol.

    Authority:  Secs. 201, 301, 501, 502, 503, 505, 701 of the Federal 
Food, Drug, and Cosmetic Act (21 U.S.C. 321, 331, 351, 352, 353, 355, 
371).

    Source:  60 FR 13595, Mar. 13, 1995, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 328.1  Scope.

    Reference in this part to regulatory sections of the Code of Federal 
Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 328.3  Definitions.

    As used in this part:
    (a) Alcohol means the substance known as ethanol, ethyl alcohol, or 
Alcohol, USP.
    (b) Inactive ingredient means any component of a product other than 
an active ingredient as defined in Sec. 210.3(b)(7) of this chapter.



                         Subpart B--Ingredients



Sec. 328.10  Alcohol.

    (a) Any over-the-counter (OTC) drug product intended for oral 
ingestion shall not contain alcohol as an inactive ingredient in 
concentrations that exceed those established in this part, unless a 
specific exemption, as provided in paragraph (e) or (f) of this section, 
has been approved.
    (b) For any OTC drug product intended for oral ingestion and labeled 
for use by adults and children 12 years of age and over, the amount of 
alcohol in the product shall not exceed 10 percent.
    (c) For any OTC drug product intended for oral ingestion and labeled 
for use by children 6 to under 12 years of age, the amount of alcohol in 
the product shall not exceed 5 percent.
    (d) For any OTC drug product intended for oral ingestion and labeled 
for use by children under 6 years of age, the amount of alcohol in the 
product shall not exceed 0.5 percent.
    (e) The Food and Drug Administration will grant an exemption from

[[Page 206]]

paragraphs (b), (c), and (d) of this section where appropriate, upon 
petition under the provisions of Sec. 10.30 of this chapter. Appropriate 
cause, such as a specific solubility or manufacturing problem, must be 
adequately documented in the petition. Decisions with respect to 
requests for exemption shall be maintained in a permanent file for 
public review by the Dockets Management Branch (HFA-305), Food and Drug 
Administration, rm. 1-23, 12420 Parklawn Dr., Rockville, MD 20857.
    (f) Ipecac syrup is exempt from the provisions of paragraph (d) of 
this section.
    (g) The following drugs are temporarily exempt from the provisions 
of paragraphs (b), (c), and (d) of this section:
    (1) Aromatic Cascara Fluidextract.
    (2) Cascara Sagrada Fluidextract.
    (3) Orally ingested homeopathic drug products.
[60 FR 13595, Mar. 13, 1995, as amended at 61 FR 58630, Nov. 18, 1996]



                           Subpart C--Labeling



Sec. 328.50  Principal display panel of all OTC drug products intended for oral ingestion that contain alcohol.

    (a) The amount (percentage) of alcohol present in a product shall be 
stated in terms of percent volume of absolute alcohol at 60  deg. F 
(15.56  deg. C) in accordance with Sec. 201.10(d)(2) of this chapter.
    (b) A statement expressing the amount (percentage) of alcohol 
present in a product shall appear prominently and conspicuously on the 
``principal display panel,'' as defined in Sec. 201.60 of this chapter. 
For products whose principal display panel is on the immediate container 
label and that are not marketed in another retail package (e.g., an 
outer box), the statement of the percentage of alcohol present in the 
product shall appear prominently and conspicuously on the ``principal 
display panel'' of the immediate container label.
    (c) For products whose principal display panel is on the retail 
package and the retail package is not the immediate container, the 
statement of the percentage of alcohol present in the product shall also 
appear on the immediate container label; it may appear anywhere on that 
label in accord with section 502(e) of the Federal Food, Drug, and 
Cosmetic Act.
    (d) The statement expressing the amount (percentage) of alcohol 
present in the product shall be in a size reasonably related to the most 
prominent printed matter on the panel or label on which it appears, and 
shall be in lines generally parallel to the base on which the package 
rests as it is designed to be displayed.
    (e) For a product to state in its labeling that it is ``alcohol 
free,'' it must contain no alcohol (0 percent).
    (f) For any OTC drug product intended for oral ingestion containing 
over 5 percent alcohol and labeled for use by adults and children 12 
years of age and over, the labeling shall contain the following 
statement in the directions section: ``Consult a physician for use in 
children under 12 years of age.''
    (g) For any OTC drug product intended for oral ingestion containing 
over 0.5 percent alcohol and labeled for use by children ages 6 to under 
12 years of age, the labeling shall contain the following statement in 
the directions section: ``Consult a physician for use in children under 
6 years of age.''
    (h) When the direction regarding age in paragraph (e) or (f) of this 
section differs from an age-limiting direction contained in any OTC drug 
monograph in this chapter, the direction containing the more stringent 
age limitation shall be used.



PART 329--HABIT-FORMING DRUGS--Table of Contents




           Subpart A--Derivatives Designated as Habit Forming

Sec.
329.1  Habit-forming drugs which are chemical derivatives of substances 
          specified in section 502(d) of the Federal Food, Drug, and 
          Cosmetic Act.

                           Subpart B--Labeling

329.10  Labeling requirements for habit-forming drugs.

                          Subpart C--Exemptions

329.20  Exemption of certain habit-forming drugs from prescription 
          requirements.

    Authority:  21 U.S.C. 352, 353, 355, 371.

[[Page 207]]


    Source:  39 FR 11736, Mar. 29, 1974, unless otherwise noted.



           Subpart A--Derivatives Designated as Habit Forming



Sec. 329.1  Habit-forming drugs which are chemical derivatives of substances specified in section 502(d) of the Federal Food, Drug, and Cosmetic Act.

    Each of the following chemical derivatives of a substance named in 
section 502(d) of the Federal Food, Drug, and Cosmetic Act is hereby 
designated as habit forming:

                                                                        
------------------------------------------------------------------------
                                 Common or official  Some trade or other
    Chemical description of       name of chemical    names of chemical 
          derivative             derivative or its    derivative or its 
                                       salts              salts \1\     
------------------------------------------------------------------------
                    PARENT SUBSTANCE--BARBITURIC ACID                   
                                                                        
------------------------------------------------------------------------
5-Allyl-5-sec-butylbarbituric   Talbutal...........  Lotusate.          
 acid \2\.                                                              
5-Allyl-5-                      ...................  Cyclopal.          
 cyclopentenylbarbituric acid.                       Cyclopen.          
5-Allyl-5-isobutylbarbituric    Allylbarbituric      Sandoptal.         
 acid.                           acid.                                  
                                Allylisobutylbarbit                     
                                 uric acid..                            
5-Allyl-5-isopropylbarbituric   Aprobarbital.......  Alurate.           
 acid.                                                                  
                                Allylisopropylbarbi  Numal.             
                                 turic acid.                            
                                Allylisopropylmalon                     
                                 ylurea..                               
5-Allyl-5-isopropyl-1-          ...................  Narconumal.        
 methylbarbituric acid.                                                 
5-Allyl-5-(1-                   Secobarbital sodium  Seconal Sodium.    
 methylbutyl)barbituric acid.                                           
                                Soluble              Evronal Sodium.    
                                 secobarbital.                          
5-Allyl-5-(1-methylbutyl)-2-    Sodium thiamylal...  Surital Sodium.    
 thiobarbituric acid.                                                   
5-Allyl-1-methyl-5-(1-methyl-2- Sodium methohexital  Brevital Sodium.   
 pentynyl) barbituric acid.                                             
5-(2-Bromoallyl)-5-isoprophyl-  ...................  Eunarcon.          
 1-methylbarbituric acid.                                               
5-(2-Bromoallyl)-5-(1-          -           Sigmodal.          
 methylbutyl)-barbituric acid.   Bromoallyl sec-     Rectidon.          
                                 amylbarbituric      R239.              
                                 acid.                                  
5-sec-Butyl-5-(2-bromoallyl)-   Butallylonal.......  Pernoston.         
 barbituric acid.                                    Pernocton.         
5-(1-Cyclohepten-1-yl)-5-       Heptabarbital......  Medomin.           
 ethylbarbituric acid.                                                  
5,5-Diallylbarbituric acid....  Diallyl barbituric   Dial.              
                                 acid.               Allobarbital.      
                                                     Allobarbitone.     
                                                     Curral.            
                                                     Diadol.            
5,5-Diethylbarbituric acid....  Barbital...........  Deba.              
                                Barbitone..........  Dormonal.          
                                Diethylbarbituric    Hypnogene.         
                                 acid.                                  
                                Diethylmalonylurea.  Malonal.           
                                                     Medinal.           
                                                     Sedeval.           
                                                     Veronal.           
                                                     Uronal.            
                                                     Vesperal.          
5,5-Diethyl-1-methylbarbituric  Metharbital........  Gemonil.           
 acid.                                                                  
1,5-Dimethyl-5-(1-              Hexobarbital sodium  Cyclonal Sodium.   
 cyclohexenyl)-barbituric acid.                      Dorico Soluble.    
                                                     Evipal Sodium.     
                                                     Evipan Sodium.     
                                                     Hexanastab.        
                                                     Hexobarbitone      
                                                      Sodium.           
                                                     Methenexyl Sodium. 
5,5-Dipropylbarbituric acid...  Dipropylbarbituric   Proponal.          
                                 acid.                                  
5-Ethyl-5-butylbarbituric acid  Butethal...........  Etoval.            
                                Butobarbital.......  Neonal             
                                                      Butobarbital.     
                                                     Soneryl.           
5-Ethyl-5-sec-butylbarbituric   Butabarbital sodium  Butisol Sodium.    
 acid.                                                                  

[[Page 208]]

                                                                        
5-Ethyl-5-(1-cyclohexenyl)-     Cyclobarbital......  Cyclobarbitone.    
 barbituric acid.                                    Namuron.           
                                                     Palinum.           
                                                     Phanodorm.         
                                                     Phanodorn.         
                                                     Tetrahydro         
                                                      phenobarbital.    
5-Ethyl-5-cyclopentenyl-        ...................  Pentenal.          
 barbituric acid.                                                       
5-Ethyl-5-hexylbarbituric acid  Hexethal sodium....  Hebaral.           
                                                     Ortal Sodium.      
5-Ethyl-5-isoamylbarbituric     Amobarbital........  Amytal.            
 acid.                                                                  
5-Ethyl-5-isopropylbarbituric   Probarbital........  Ipral.             
 acid.                                                                  
5-Ethyl-5-(1-methylbutyl)-      Pentobarbital        844.               
 barbituric acid.                sodium.                                
                                Soluble              Embutal.           
                                 pentobarbital.      Nembutal.          
                                                     Napethal.          
                                                     Pentyl.            
5-Ethyl-5-(1-methylbutyl)-2-    Thiopental sodium..  Intraval Sodium.   
 thiobarbituric acid.                                                   
                                Thiopentone sodium.  Nesdonal Sodium.   
                                                     Pentothal Sodium.  
                                                     Thiothal Sodium.   
5-Ethyl-5-(1-methyl-1-butenyl)- Vinbarbital........  Delvinal Sodium.   
 barbituric acid.                                                       
5-Ethyl-5-phenylbarbituric      Phenobarbital......  Barbenyl.          
 acid.                                                                  
                                Phenobarbitone.....  Barbiphenyl.       
                                Phenylethylmalonylu  Dormiral.          
                                 rea.                Euneryl.           
                                                     Gardenal.          
                                                     Luminal.           
                                                     Nunol.             
                                                     Neurobarb.         
                                                     Phenonyl.          
                                                     Somonal.           
5-Ethyl-5-phenyl-1-             Mephobarbital......  Mebaral.           
 methylbarbituric acid.                              Phemitone.         
                                                     Prominal.          
5-Ethyl-5-(1 piperidyl)-        ...................  Eldoral.           
 barbituric acid.                                                       
5-Isopropyl-5-(2-bromoallyl)-   Propallylonal......  Noctal.            
 barbituric acid.                                    Nostal.            
5-(1-Methylbutyl)-5-[2-         Methitural (sodium   Methioturiate.     
 (methylthio)ethyl]-2-           salt).              Neraval.           
 thiobarbituric acid.                                Thiogenal.         
5-Methyl-5-phenylbarbituric     Phenylmethylbarbitu  Rutonal.           
 acid.                           ric acid.                              
All lithium, sodium,                                                    
 potassium, magnesium,                                                  
 calcium, strontium, and                                                
 ammonium salts of the                                                  
 foregoing chemical                                                     
 derivatives of barbituric                                              
 acid.                                                                  
                                                                        
------------------------------------------------------------------------
                 PARENT SUBSTANCE--CANNABIS (MARIHUANA)                 
                                                                        
------------------------------------------------------------------------
                                Extract of cannabis                     
                                Fluid extract of                        
                                 cannabis.                              
                                Tincture of                             
                                 cannabis.                              
                                                                        
------------------------------------------------------------------------
                        PARENT SUBSTANCE--BROMAL                        
                                                                        
------------------------------------------------------------------------
Tribromoacetaldehyde hydrate..  Bromal hydrate.....                     
Tribromomethane...............  Bromoform..........                     
2-(Tribromomethyl)-2-propanol.  Tribromo-tert-butyl  Acetone-Bromoform. 
                                 alcohol.            Brometone.         
                                                                        
------------------------------------------------------------------------
                       PARENT SUBSTANCE--CARBROMAL                      
                                                                        
------------------------------------------------------------------------
a-Bromo-a-ethylbutyryl-         Acetylcarbromal....  A basin.           
 acetylurea.                                         Acetyl Adalin.     
                                                     N-Acetyl-N-        
                                                      bromodiethylacetyl
                                                      urea.             
                                                     N-Acetyl-N '-a-    
                                                      bromo-a-          
                                                      ethylbutyryl      
                                                      carbamide.        

[[Page 209]]

                                                                        
a-Bromoisovalerylurea.........  Bromisovalum.......  Bromisoval.        
                                                     a-Bromo-- 
                                                      dimethyl-         
                                                      propanoylurea.    
                                                     Bromural.          
                                                     Bromvaletone.      
                                                     Brovalurea.        
                                                     B. V. U.           
                                                     Dormigene.         
                                                     Isobromyl.         
                                                     2-                 
                                                      Monobromoisovalery
                                                      lurea.            
                                                     Pivadorm.          
                                                     Uvaleral.          
a-Bromo-a,a-diethylacetamide..  Diethylbromo         Neuronal.          
                                 acetamide.                             
a-Allylisovaleryl-urea........  ...................  Allyl-isopropyl-   
                                                      acetyl-carbamide. 
                                                     (2-Isopropyl-4-    
                                                      pentenoyl)-urea.  
                                                     Sedormid.          
                                                                        
------------------------------------------------------------------------
                        PARENT SUBSTANCE--CHLORAL                       
                                                                        
------------------------------------------------------------------------
Trichloroacetaldehyde hydrate.  Chloral............  2,2,2-Trichloro-1,1-
                                                      ethanediol.       
                                Chloral hydrate....  Trichloroethylidene
                                                      glycol.           
Trichloroethylideneimine......  Chloralimide.......                     
N-(-Trichloro-a-       Chloralformamide...  Chloralamide.      
 hydroxyethyl)-formamide.                            Chloramide.        
a-(-trichloro-a-       a-Chloralose.......  A-D-               
 hydroxyethyl)-D-glucoside.                           Glucochloralose.  
                                                     Anhydro-           
                                                      Glucochloral.     
                                                     Glucochloral.      
                                                     Chloralosone.      
2-(Trichloromethyl)-2-propanol  Chlorbutanol.......  Acetone chloroform.
                                Chlorbutol.........  Chloretone.        
                                Chlorobutanol......  Methaform.         
                                                     Sedaform.          
                                                     1,1,1-trichloro-2- 
                                                      methyl 2-propanol.
                                                     ,
                                                      ,-       
                                                      trichloro-tert-   
                                                      butylalcohol.     
                                                                        
------------------------------------------------------------------------
                        PARENT SUBSTANCE--COCAINE                       
                                                                        
------------------------------------------------------------------------
All salts of cocaine obtained   Cocaine                                 
 by combining cocaine with any   hydrochloride.                         
 acid.                          Cocainium chloride.                     
                                                                        
------------------------------------------------------------------------
                        PARENT SUBSTANCE--CODEINE                       
                                                                        
------------------------------------------------------------------------
Codeine methylbromide.........  Eucodin............                     
Dihydrocodeinone..............  ...................  Dicodid.           
Dihydrohydroxycodeinone.......  Eucodal............  Oxycodone          
                                                      hydrochloride.    
                                                     14-                
                                                      hydroxydihydrocode
                                                      inone.            
All salts of the foregoing                                              
 chemical derivatives of                                                
 codeine obtained by combining                                          
 any such derivative of                                                 
 codeine with any acid.                                                 
                                                                        
------------------------------------------------------------------------
                        PARENT SUBSTANCE--HEROIN                        
                                                                        
------------------------------------------------------------------------
All salts of heroin obtained                                            
 by combining heroin with any                                           
 acid.                                                                  
                                                                        
------------------------------------------------------------------------
                       PARENT SUBSTANCE--MORPHINE                       
                                                                        
------------------------------------------------------------------------
Dihydromorphine...............  Paramorphan........                     
Dihydromorphinone.............  Dihydromorphinone    Dilaudid.          
                                 hydrochloride.      Dimorphone.        
                                Dihydromorphinonium  Hydromorphone      
                                 chloride.            hydrochloride.    
Ethylmorphine.................  Ethylmorphine        Dionin.            
                                 hydrochloride.                         
                                Ethylmorphinium                         
                                 chloride..                             
All salts of the foregoing                                              
 chemical derivatives of                                                
 morphine and all salts of                                              
 morphine obtained by                                                   
 combining any such derivative                                          
 or morphine with any acid.                                             
                                                                        
------------------------------------------------------------------------
                         PARENT SUBSTANCE--OPIUM                        
                                                                        
------------------------------------------------------------------------
                                Extract of opium...                     

[[Page 210]]

                                                                        
                                Fluidextract of                         
                                 opium.                                 
                                Camphorated opium                       
                                 tincture.                              
                                Deodorized opium                        
                                 tincture.                              
                                Laudanum...........                     
                                Opium tincture.....                     
                                Paregoric..........                     
                                Tincture of opium..                     
                                                                        
------------------------------------------------------------------------
                      PARENT SUBSTANCE--PARALDEHYDE                     
                                                                        
------------------------------------------------------------------------
Metaldehyde...................                                          
                                                                        
------------------------------------------------------------------------
                     PARENT SUBSTANCE--SULFONMETHANE                    
                                                                        
------------------------------------------------------------------------
2,2-Diethylsulfonylbutane.....  Sulfonethylmethane.  Diethylsulfonmethyl
                                                      ethyl-methane.    
                                                     Ethylsulfonal.     
                                                     2,2-bis-           
                                                      (Ethylsulfonyl)-  
                                                      butane.           
                                                     Methylsufonal.     
                                                     Sulfonethlylmethanu
                                                      m.                
                                                     Trional.           
3,3-Diethylsulfonylpentane....  Sulfondiethylmethan                     
                                 e.                                     
------------------------------------------------------------------------
\1\ This list of trade or other names is not a complete list of the many
  proprietary names under which the designated habit-forming chemical   
  derivatives are distributed.                                          
\2\ The name ``butalbital'' is obsolete for this compound;              
  ``butalbital'' is the nonproprietary name assigned by the United      
  States Adopted Name Council and the World Health Organization for 5-  
  allyl-5-isobutylbarbituric acid.                                      



                           Subpart B--Labeling



Sec. 329.10  Labeling requirements for habit-forming drugs.

    (a)(1) The name of a substance or derivative required to be borne on 
the label of a drug by section 502(d) of the act shall be the common or 
usual name of such substance or derivative, unless it is designated 
solely by a name recognized in an official compendium and such 
designation complies with the provisions of section 502(c).
    (2) A statement on the label of a drug of the name of a constituent, 
which constituent is a chemical derivative of a substance named in 
section 502(d) of the act, shall show the substance from which such 
constituent is derived and that such constituent is a derivative 
thereof.
    (b) If the drug is in tablet, capsule, ampul, or other unit form, 
the statement of the quantity or proportion of such substance or 
derivative contained therein shall express the weight or measure of such 
substance or derivative in each such unit. If the drug is not in such 
unit form the statement shall express the weight or measure of such 
substance or derivative in a specified unit of weight or measure of the 
drug. Such statement shall be in terms which are informative to the 
ordinary consumer and user of the drug.
    (c) The names and quantities or proportions of all such substances 
and derivatives, and the statement ``Warning--May be habit forming'', 
shall immediately follow (without intervening written, printed, or 
graphic matter) the name by which such drug is titled in the part or 
panel of the label thereof which is presented or displayed under 
customary conditions of purchase.
    (d) A drug shall not be considered to be misbranded by reason of 
failure of its label to bear the statement ``Warning--May be habit 
forming'':
    (1) If such drug is not suitable for internal use, and is 
distributed and sold exclusively for such external use as involves no 
possibility of habit formation; or
    (2) If the only substance or derivative subject to section 502(d) of 
the act contained in such drug is chlorobutanol, which is present solely 
as a preservative and in a quantity not more than 0.5 percent by weight, 
and such drug is for parenteral use only; or
    (3) If the only substance or derivative subject to section 502(d) of 
the act contained in such drug is chlorobutanol which is present as an 
analgesic or as an analgesic and a preservative in a quantity not more 
than 3.0 percent,

[[Page 211]]

and such drug contains one or more other active ingredients and is for 
parenteral use only.

    Cross Reference: For the Spanish-language version of the required 
labeling statement, see Sec. 201.16(b) of this chapter.
[39 FR 11736, Mar. 29, 1974, as amended at 40 FR 13496, Mar. 27, 1975]



                          Subpart C--Exemptions



Sec. 329.20  Exemption of certain habit-forming drugs from prescription requirements.

    The prescription-dispensing requirements of section 503(b)(1)(A) of 
the act are not necessary for the protection of the public health with 
respect to the following drugs subject to section 502(d):
    (a) The following exempt narcotic preparations:
    (1) Pharmaceutical preparations containing not more than 100 
milligrams of opium per 100 milliliters or per 100 grams.
    (2) Pharmaceutical preparations containing not more than 16.2 
milligrams (\1/4\ grain) morphine, or any of its salts, per 29.5729 
cubic centimeters (1 fluid ounce) or per 28.3 grams (1 avoirdupois 
ounce);
    (3) Pharmaceutical preparations containing not more than 64.8 
milligrams (1 grain) codeine, or any of its salts, per 29.5729 cubic 
centimeters (1 fluid ounce) or per 28.3 grams (1 avoirdupois ounce);
    (4) Pharmaceutical preparations containing not more than 32.4 
milligrams (\1/2\ grain) dihydrocodeine, or any of its salts, per 
29.5729 cubic centimeters (1 fluid ounce) or per 28.3 grams (1 
avoirdupois ounce);
    (5) Pharmaceutical preparations containing not more than 16.2 
milligrams (\1/4\ grain) ethylmorphine, or any of its salts, per 29.5729 
cubic centimeters (1 fluid ounce) or per 28.3 grams (1 avoirdupois 
ounce);

Provided, That the preparations described in this paragraph contain one 
or more nonnarcotic active medicinal ingredients in sufficient 
proportion to confer upon the preparation valuable medicinal qualities 
other than those possessed by the narcotic drug alone.
    (b) Drugs containing chlorobutanol, intended for external use only.
    (c) Epinephrine solution, 1 percent, preserved with chlorobutanol 
and intended for use solely as a spray.
    (d) Combination drugs listed in part 329 as exempted from section 
511 of the act.
[39 FR 11736, Mar. 29, 1974, as amended at 55 FR 11581, Mar. 29, 1990]



PART 330--OVER-THE-COUNTER (OTC) HUMAN DRUGS WHICH ARE GENERALLY RECOGNIZED AS SAFE AND EFFECTIVE AND NOT MISBRANDED--Table of Contents




                      Subpart A--General Provisions

Sec.
330.1  General conditions for general recognition as safe, effective and 
          not misbranded.
330.2  Pregnancy-nursing warning.
330.3  Imprinting of solid oral dosage form drug products.
330.5  Drug categories.

                  Subpart B--Administrative Procedures

330.10  Procedures for classifying OTC drugs as generally recognized as 
          safe and effective and not misbranded, and for establishing 
          monographs.
330.11  NDA deviations from applicable monograph.
330.12  Status of over-the-counter (OTC) drugs previously reviewed under 
          the Drug Efficacy Study (DESI).
330.13  Conditions for marketing ingredients recommended for over-the-
          counter (OTC) use under the OTC drug review.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  39 FR 11741, Mar. 29, 1974, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 330.1  General conditions for general recognition as safe, effective and not misbranded.

    An over-the-counter (OTC) drug listed in this subchapter is 
generally recognized as safe and effective and is not misbranded if it 
meets each of the conditions contained in this part and each of the 
conditions contained in any applicable monograph. Any product which 
fails to conform to each of the conditions contained in this part and

[[Page 212]]

in an applicable monograph is liable to regulatory action.
    (a) The product is manufactured in compliance with current good 
manufacturing practices, as established by parts 210 and 211 of this 
chapter.
    (b) The establishment(s) in which the drug product is manufactured 
is registered, and the drug product is listed, in compliance with part 
207 of this chapter. It is requested but not required that the number 
assigned to the product pursuant to part 207 of this chapter appear on 
all drug labels and in all drug labeling. If this number is used, it 
shall be placed in the manner set forth in part 207 of this chapter.
    (c)(1) The product is labeled in compliance with chapter V of the 
act and subchapter C et seq. of this chapter. For purposes of 
Sec. 201.61(b) of this chapter, the statement of identity of the product 
shall be the term or phrase used in the applicable monograph established 
in this part.
    (2)(i) The label and labeling of the product contain in a prominent 
and conspicuous location the labeling describing the ``Indications'' 
that have been established in an applicable final monograph. At the 
option of the manufacturer, this labeling may be designated ``APPROVED 
USES,'' or be given a similar designation as permitted by this 
paragraph, each time it appears in the labeling, e.g., on the outer 
carton, inner bottle label, and on any package insert or display 
material. If the ``APPROVED USES'' or a similar designation is used, the 
labeling involved shall appear within a boxed area. Other applicable 
labeling established under this subchapter and subchapter C of this 
chapter may be included in the boxed area. If such other labeling is 
included, the boxed area shall be designated ``APPROVED INFORMATION'' 
rather than ``APPROVED USES.'' The ``indications'' information appearing 
in the boxed area shall be stated in the exact language of the 
monograph. Other information within the boxed area also shall be stated 
in exact language where exact language has been established and 
identified by quotation marks in an applicable monograph or by 
regulation (e.g., Sec. 201.63 of this chapter). A statement that the 
information in the box was ``published by the Food and Drug 
Administration'' shall appear within the boxed area, or reasonably close 
by. In lieu of such statement, the designation of the boxed area may be 
modified to read: ``FDA APPROVED USES'' or ``FDA APPROVED INFORMATION,'' 
as appropriate, or ``USES (or ``INFORMATION'') APPROVED BY THE FOOD AND 
DRUG ADMINISTRATION,'' or other similar wording.
    (ii) At the option of the manufacturer, as an alternative to the 
requirements of paragraph (c)(2)(i) of this section, the label and 
labeling of the product may contain in a prominent and conspicuous 
location other truthful and nonmisleading statements describing only 
those indications for use that have been established in an applicable 
monograph, subject to the provisions of section 502 of the act relating 
to misbranding and the prohibition in section 301(d) of the act against 
the introduction or delivery for introduction into interstate commerce 
of unapproved new drugs in violation of section 505(a) of the act. Such 
labeling shall not be boxed and shall not contain the statements 
provided in paragraph (c)(2)(i) of this section relating to ``APPROVED 
USES,'' or ``APPROVED INFORMATION,'' or contain a statement that the 
labeling has been published by the Food and Drug Administration.
    (iii) At the option of the manufacturer, the label and labeling may 
meet the boxed-area requirements of paragraph (c)(2)(i) of this section 
and, in addition, other truthful and nonmisleading statements describing 
only those indications for use that have been established in an 
applicable monograph may appear elsewhere in the labeling, that is, 
outside the boxed area, subject to the provisions of section 502 of the 
act relating to misbranding and the prohibition in section 301(d) of the 
act against the introduction or delivery for introduction into 
interstate commerce of unapproved new drugs in violation of section 
505(a) of the act.
    (iv) At the option of the manufacturer, more than one of the 
alternatives described in paragraphs (c)(2)(i), (ii), and (iii) may be 
used in separate labeling, e.g., container label, outer carton, package 
insert, display material, for a particular OTC drug

[[Page 213]]

product provided each labeling complies with all applicable statutory 
and regulatory labeling requirements in all respects.
    (v) The term ``prominent and conspicuous location'' as used in 
paragraphs (c)(2) (i) and (ii) of this section means that the labeling 
within the boxed or nonboxed area shall be presented and displayed in 
such a manner as to render it likely to be read as understood by the 
ordinary individual under customary conditions at both time of purchase 
and use.
    (vi) Regardless of the alternative selected by the manufacturer to 
describe indications, paragraphs (c)(2)(i), (ii), and (iii) of this 
section require other labeling established under this subchapter and 
subchapter C of this chapter to be stated in the exact language where 
exact language has been established and identified by quotation marks in 
an applicable monograph or by regulation (e.g., Sec. 201.63 of this 
chapter).
    (d) The advertising for the product prescribes, recommends, or 
suggests its use only under the conditions stated in the labeling.
    (e) The product contains only suitable inactive ingredients which 
are safe in the amounts administered and do not interfere with the 
effectiveness of the preparation or with suitable tests or assays to 
determine if the product meets its professed standards of identity, 
strength, quality, and purity. Color additives may be used only in 
accordance with section 721 of the act and subchapter A of this chapter.
    (f) The product container and container components meet the 
requirements of Sec. 211.94 of this chapter.
    (g) The labeling for all drugs contains the general warning: ``Keep 
this and all drugs out of the reach of children.'' The labeling of drugs 
used for oral administation shall also state: ``In case of accidental 
overdose, seek professional assistance or contact a poison control 
center immediately.'' The labeling for drugs administered rectally or 
used topically shall state: ``In case of accidental ingestion, seek 
professional assistance or contact a Poison Control Center 
immediately.'' The Food and Drug Administration will grant an exemption 
from these general warnings where appropriate upon petition, which shall 
be maintained in a permanent file for public review by the Dockets 
Management Branch, Food and Drug Administration, rm. 1-23, 12420 
Parklawn Dr., Rockville, MD 20857.
    (h) Where no maximum daily dosage limit for an active ingredient is 
established in this part, it is used in a product at a level that does 
not exceed the amount reasonably required to achieve its intended 
effect.
    (i) The following terms may be used interchangeably in any of the 
labeling established in parts 331 through 358 of this chapter:
    (1) ``Ask'' or ``consult''.
    (2) ``Assistance'' or ``help''.
    (3) ``Clean'' or ``cleanse''.
    (4) ``Continue'' or ``persist''.
    (5) ``Continues'' or ``persists''.
    (6) ``Doctor'' or ``physician''.
    (7) ``Indication'' or ``use''.
    (8) ``Indications'' or ``uses''.
    (9) ``Lung'' or ``pulmonary''.
    (j) It is recommended that the labeling of the product contain the 
quantitative amount of each active ingredient, expressed in terms of the 
dosage unit stated in the directions for use (e.g., tablet, 
teaspoonful).
[39 FR 11741, Mar. 29, 1974, as amended at 40 FR 11718, Mar. 13, 1975; 
40 FR 13496, Mar. 27, 1975; 42 FR 15674, Mar. 22, 1977; 46 FR 8459, Jan. 
27, 1981; 50 FR 8996, Mar. 6, 1985; 51 FR 16266, May 1, 1986; 55 FR 
11581, Mar. 29, 1990; 59 FR 4000, Jan. 28, 1994; 59 FR 14365, Mar. 28, 
1994]



Sec. 330.2  Pregnancy-nursing warning.

    A pregnancy-nursing warning for OTC drugs is set forth under 
Sec. 201.63 of this chapter.
[47 FR 54758, Dec. 3, 1982]



Sec. 330.3  Imprinting of solid oral dosage form drug products.

    A requirement to imprint an identification code on solid oral dosage 
form drug products is set forth under part 206 of this chapter.
[58 FR 47959, Sept. 13, 1993]



Sec. 330.5  Drug categories.

    Monographs promulgated pursuant to the provisions of this part shall 
be established in this part 330 and following

[[Page 214]]

parts and shall cover the following designated categories:
    (a) Antacids.
    (b) Laxatives.
    (c) Antidiarrheal products.
    (d) Emetics.
    (e) Antiemetics.
    (f) Antiperspirants.
    (g) Sunburn prevention and treatment products.
    (h) Vitamin-mineral products.
    (i) Antimicrobial products.
    (j) Dandruff products.
    (k) Oral hygiene aids.
    (l) Hemorrhoidal products.
    (m) Hematinics.
    (n) Bronchodilator and antiasthmatic products.
    (o) Analgesics.
    (p) Sedatives and sleep aids.
    (q) Stimulants.
    (r) Antitussives.
    (s) Allergy treatment products.
    (t) Cold remedies.
    (u) Antirheumatic products.
    (v) Ophthalmic products.
    (w) Contraceptive products.
    (x) Miscellaneous dermatologic products.
    (y) Dentifrices and dental products such as analgesics, antiseptics, 
etc.
    (z) Miscellaneous (all other OTC drugs not falling within one of the 
above therapeutic categories).



                  Subpart B--Administrative Procedures



Sec. 330.10  Procedures for classifying OTC drugs as generally recognized as safe and effective and not misbranded, and for establishing monographs.

    For purposes of classifying over-the-counter (OTC) drugs as drugs 
generally recognized among qualified experts as safe and effective for 
use and as not misbranded drugs, the following regulations shall apply:
    (a) Procedure for establishing OTC drug monographs--(1) Advisory 
review panels. The Commissioner shall appoint advisory review panels of 
qualified experts to evaluate the safety and effectiveness of OTC drugs, 
to review OTC drug labeling, and to advise him on the promulgation of 
monographs establishing conditions under which OTC drugs are generally 
recognized as safe and effective and not misbranded. A single advisory 
review panel shall be established for each designated category of OTC 
drugs and every OTC drug category will be considered by a panel. The 
members of a panel shall be qualified experts (appointed by the 
Commissioner) and may include persons from lists submitted by 
organizations representing professional, consumer, and industry 
interests. The Commissioner shall designate the chairman of each panel. 
Summary minutes of all meetings shall be made.
    (2) Request for data and views. The Commissioner will publish a 
notice in the Federal Register requesting interested persons to submit, 
for review and evaluation by an advisory review panel, published and 
unpublished data and information pertinent to a designated category of 
OTC drugs. Data and information submitted pursuant to a published 
notice, and falling within the confidentiality provisions of 18 U.S.C. 
1905, 5 U.S.C. 552(b), or 21 U.S.C. 331(j), shall be handled by the 
advisory review panel and the Food and Drug Administration as 
confidential until publication of a proposed monograph and the full 
report(s) of the panel. Thirty days thereafter such data and information 
shall be made publicly available and may be viewed at the office of the 
Dockets Management Branch of the Food and Drug Administration, except to 
the extent that the person submitting it demonstrates that it still 
falls within the confidentiality provisions of one or more of those 
statutes. To be considered, eight copies of the data and/or views on any 
marketed drug within the class must be submitted, preferably bound, 
indexed, and on standard sized paper (approximately 8\1/2\ x 11 inches). 
When requested, abbreviated submissions should be sent. All submissions 
must be in the following format:

                       OTC Drug Review Information

    I. Label(s) and all labeling (preferably mounted and filed with the 
other data--facsimile labeling is acceptable in lieu of actual container 
labeling).
    II. A statement setting forth the quantities of active ingredients 
of the drug.
    III. Animal safety data.
    A. Individual active components.
    1. Controlled studies.

[[Page 215]]

    2. Partially controlled or uncontrolled studies.
    B. Combinations of the individual active components.
    1. Controlled studies.
    2. Partially controlled or uncontrolled studies.
    C. Finished drug product.
    1. Controlled studies.
    2. Partially controlled or uncontrolled studies.
    IV. Human safety data.
    A. Individual active components.
    1. Controlled studies.
    2. Partially controlled or uncontrolled studies.
    3. Documented case reports.
    4. Pertinent marketing experiences that may influence a 
determination as to the safety of each individual active component.
    5. Pertinent medical and scientific literature.
    B. Combinations of the individual active components.
    1. Controlled studies.
    2. Partially controlled or uncontrolled studies.
    3. Documented case reports.
    4. Pertinent marketing experiences that may influence a 
determination as to the safety of combinations of the individual active 
components.
    5. Pertinent medical and scientific literature.
    C. Finished drug product.
    1. Controlled studies.
    2. Partially controlled or uncontrolled studies.
    3. Documented case reports.
    4. Pertinent marketing experiences that may influence a 
determination as to the safety of the finished drug product.
    5. Pertinent medical and scientific literature.
    V. Efficacy data.
    A. Individual active components.
    1. Controlled studies.
    2. Partially controlled or uncontrolled studies.
    3. Documented case reports.
    4. Pertinent marketing experiences that may influence a 
determination on the efficacy of each individual active component.
    5. Pertinent medical and scientific literature.
    B. Combinations of the individual active components.
    1. Controlled studies.
    2. Partially controlled or uncontrolled studies.
    3. Documented case reports.
    4. Pertinent marketing experiences that may influence a 
determination on the efficacy of combinations of the individual active 
components.
    5. Pertinent medical and scientific literature.
    C. Finished drug product.
    1. Controlled studies.
    2. Partially controlled or uncontrolled studies.
    3. Documented case reports.
    4. Pertinent marketing experiences that may influence a 
determination on the efficacy of the finished drug product.
    5. Pertinent medical and scientific literature.
    VI. A summary of the data and views setting forth the medical 
rationale and purpose (or lack thereof) for the drug and its ingredients 
and the scientific basis (or lack thereof) for the conclusion that the 
drug and its ingredients have been proven safe and effective for the 
intended use. If there is an absence of controlled studies in the 
material submitted, an explanation as to why such studies are not 
considered necessary must be included.

    (3) Deliberations of an advisory review panel. An advisory review 
panel will meet as often and for as long as is appropriate to review the 
data submitted to it and to prepare a report containing its conclusions 
and recommendations to the Commissioner with respect to the safety and 
effectiveness of the drugs in a designated category of OTC drugs. A 
panel may consult any individual or group. Any interested person may 
request an opportunity to present oral views to the panel; such request 
may be granted or denied by the panel. Such requests for oral 
presentations should be in written form including a summarization of the 
data to be presented to the panel. Any interested person may present 
written data and views which shall be considered by the panel. This 
information shall be presented to the panel in the format set forth in 
paragraph (a)(2) of this section and within the time period established 
for the drug category in the notice for review by a panel.
    (4) Standards for safety, effectiveness, and labeling. The advisory 
review panel, in reviewing the data submitted to it and preparing its 
conclusions and recommendations, and the Commissioner, in reviewing the 
conclusions and recommendations of the panel and the published proposed, 
tentative, and the final monographs, shall apply the following standards 
to determine general recognition that a category of OTC drugs is safe 
and effective and not misbranded:

[[Page 216]]

    (i) Safety means a low incidence of adverse reactions or significant 
side effects under adequate directions for use and warnings against 
unsafe use as well as low potential for harm which may result from abuse 
under conditions of widespread availability. Proof of safety shall 
consist of adequate tests by methods reasonably applicable to show the 
drug is safe under the prescribed, recommended, or suggested conditions 
of use. This proof shall include results of significant human experience 
during marketing. General recognition of safety shall ordinarily be 
based upon published studies which may be corroborated by unpublished 
studies and other data.
    (ii) Effectiveness means a reasonable expectation that, in a 
significant proportion of the target population, the pharmacological 
effect of the drug, when used under adequate directions for use and 
warnings against unsafe use, will provide clinically significant relief 
of the type claimed. Proof of effectiveness shall consist of controlled 
clinical investigations as defined in Sec. 314.126(b) of this chapter, 
unless this requirement is waived on the basis of a showing that it is 
not reasonably applicable to the drug or essential to the validity of 
the investigation and that an alternative method of investigation is 
adequate to substantiate effectiveness. Investigations may be 
corroborated by partially controlled or uncontrolled studies, documented 
clinical studies by qualified experts, and reports of significant human 
experience during marketing. Isolated case reports, random experience, 
and reports lacking the details which permit scientific evaluation will 
not be considered. General recognition of effectiveness shall ordinarily 
be based upon published studies which may be corroborated by unpublished 
studies and other data.
    (iii) The benefit-to-risk ratio of a drug shall be considered in 
determining safety and effectiveness.
    (iv) An OTC drug may combine two or more safe and effective active 
ingredients and may be generally recognized as safe and effective when 
each active ingredient makes a contribution to the claimed effect(s); 
when combining of the active ingredients does not decrease the safety or 
effectiveness of any of the individual active ingredients; and when the 
combination, when used under adequate directions for use and warnings 
against unsafe use, provides rational concurrent therapy for a 
significant proportion of the target population.
    (v) Labeling shall be clear and truthful in all respects and may not 
be false or misleading in any particular. It shall state the intended 
uses and results of the product; adequate directions for proper use; and 
warnings against unsafe use, side effects, and adverse reactions in such 
terms as to render them likely to be read and understood by the ordinary 
individual, including individuals of low comprehension, under customary 
conditions of purchase and use.
    (vi) A drug shall be permitted for OTC sale and use by the laity 
unless, because of its toxicity or other potential for harmful effect or 
because of the method or collateral measures necessary to its use, it 
may safely be sold and used only under the supervision of a practitioner 
licensed by law to administer such drugs.
    (5) Advisory review panel report to the Commissioner. An advisory 
review panel shall submit to the Commissioner a report containing its 
conclusions and recommendations with respect to the conditions under 
which OTC drugs falling within the category covered by the panel are 
generally recognized as safe and effective and not misbranded. Included 
within this report shall be:
    (i) A recommended monograph or monographs covering the category of 
OTC drugs and establishing conditions under which the drugs involved are 
generally recognized as safe and effective and not misbranded (Category 
I). This monograph may include any conditions relating to active 
ingredients, labeling indications, warnings and adequate directions for 
use, prescription or OTC status, and any other conditions necessary and 
appropriate for the safety and effectiveness of drugs covered by the 
monograph.
    (ii) A statement of all active ingredients, labeling claims or other 
statements, or other conditions reviewed and excluded from the monograph 
on the basis of the panel's determination that they would result in the 
drug's

[[Page 217]]

not being generally recognized as safe and effective or would result in 
misbranding (Category II).
    (iii) A statement of all active ingredients, labeling claims or 
other statements, or other conditions reviewed and excluded from the 
monograph on the basis of the panel's determination that the available 
data are insufficient to classify such condition under either paragraph 
(a)(5) (i) or (ii) of this section and for which further testing is 
therefore required (Category III). The report may recommend the type of 
further testing required and the time period within which it might 
reasonably be concluded.
    (6) Proposed monograph. After reviewing the conclusions and 
recommendations of the advisory review panel, the Commissioner shall 
publish in the Federal Register a proposed order containing:
    (i) A monograph or monographs establishing conditions under which a 
category of OTC drugs is generally recognized as safe and effective and 
not misbranded (Category I).
    (ii) A statement of the conditions excluded from the monograph on 
the basis of the Commissioner's determination that they would result in 
the drug's not being generally recognized as safe and effective or would 
result in misbranding (Category II).
    (iii) A statement of the conditions excluded from the monograph on 
the basis of the Commissioner's determination that the available data 
are insufficient to classify such conditions under either paragraph 
(a)(6)(i) or (ii) of this section (Category III).
    (iv) The full report(s) of the panel to the Commissioner. The 
proposed order shall specify a reasonable period of time within which 
conditions falling within paragraph (a)(6)(iii) of this section may be 
continued in marketed products while the data necessary to support them 
are being obtained for evaluation by the Food and Drug Administration. 
The summary minutes of the panel meetings shall be made available to 
interested persons upon request. Any interested person may, within 90 
days after publication of the proposed order in the Federal Register, 
file with the Dockets Management Branch of the Food and Drug 
Administration written comments in quintuplicate. Comments may be 
accompanied by a memorandum or brief in support thereof. All comments 
may be reviewed at the office of the Dockets Management Branch during 
regular working hours, Monday through Friday. Within 30 days after the 
final day for submission of comments, reply comments may be filed with 
the Dockets Management Branch; these comments shall be utilized to reply 
to comments made by other interested persons and not to reiterate a 
position. The Commissioner may satisfy this requirement by publishing in 
the Federal Register a proposed order summarizing the full report of the 
advisory review panel, containing its conclusions and recommendations, 
to obtain full public comment before undertaking his own evaluation and 
decision on the matters involved.
    (7) Tentative final monograph. (i) After reviewing all comments, 
reply comments, and any new data and information, the Commissioner shall 
publish in the Federal Register a tentative order containing a monograph 
establishing conditions under which a category of OTC drugs is generally 
recognized as safe and effective and not misbranded. Within 60 days, any 
interested person may file with the Dockets Management Branch, Food and 
Drug Administration, written comments or written objections specifying 
with particularity the omissions or additions requested. These 
objections are to be supported by a brief statement of the grounds 
therefor. A request for an oral hearing may accompany such objections.
    (ii) The Commissioner may publish in the Federal Register a separate 
tentative order containing a statement of those active ingredients 
reviewed and proposed to be excluded from the monograph on the basis of 
the Commissioner's determination that they would result in a drug 
product not being generally recognized as safe and effective or would 
result in misbranding, and for which no substantive comments in 
opposition to the panel report or new data and information were received 
by the Food and Drug Administration pursuant to paragraph (a)(6)(iv) of 
this section. Within 60 days, any interested

[[Page 218]]

person may file with the Dockets Management Branch, Food and Drug 
Administration, written objections specifying with particularity the 
provision of the tentative order to which objection is made. These 
objections are to be supported by a brief statement of the grounds 
therefor. A request for an oral hearing may accompany such objections.
    (iii) Within 12 months after publishing a tentative order pursuant 
to paragraph (a)(7)(i) of this section, any interested person may file 
with the Dockets Management Branch, Food and Drug Administration, new 
data and information to support a condition excluded from the monograph 
in the tentative order.
    (iv) Within 60 days after the final day for submission of new data 
and information, comments on the new data and information may be filed 
with the Dockets Management Branch, Food and Drug Administration.
    (v) New data and information submitted after the time specified in 
this paragraph but prior to the establishment of a final monograph will 
be considered as a petition to amend the monograph and will be 
considered by the Commissioner only after a final monograph has been 
published in the Federal Register unless the Commisisoner finds that 
good cause has been shown that warrants earlier consideration.
    (8) Oral hearing before the Commissioner. After reviewing objections 
filed in response to the tentative final monograph, the Commissioner, if 
he finds reasonable grounds in support thereof, shall by notice in the 
Federal Register schedule an oral hearing. The notice scheduling an oral 
hearing shall specify the length of the hearing and how the time shall 
be divided among the parties requesting the hearing. The hearing shall 
be conducted by the Commissioner and may not be delegated.
    (9) Final monograph. After reviewing the objections, the entire 
administrative record including all new data and information and 
comments, and considering the arguments made at any oral hearing, the 
Commissioner shall publish in the Federal Register a final order 
containing a monograph establishing conditions under which a category of 
OTC drugs is generally recognized as safe and effective and not 
misbranded. The monograph shall become effective as specified in the 
order.
    (10) Administrative record. (i) All data and information to be 
considered in any proceeding pursuant to this section shall be submitted 
in response to the request for data and views pursuant to paragraph 
(a)(2) of this section or accepted by the panel during its deliberations 
pursuant to paragraph (a)(3) of this section or submitted to the Dockets 
Management Branch as part of the comments during the 90-day period and 
30-day rebuttal comment period permitted pursuant to paragraph (a)(6) of 
this section or submitted to the Dockets Management Branch during the 
12-month period or as part of the comments during the 60-day period 
permitted pursuant to paragraph (a)(7) of this section.
    (ii) The Commissioner shall make all decisions and issue all orders 
pursuant to this section solely on the basis of the administrative 
record, and shall not consider data or information not included as part 
of the administrative record.
    (iii) The administrative record shall consist solely of the 
following material: All notices and orders published in the Federal 
Register, all data and views submitted in response to the request 
published pursuant to paragraph (a)(2) of this section or accepted by 
the panel during its deliberations pursuant to paragraph (a)(3) of this 
section, all minutes of panel meetings, the panel report(s), all 
comments and rebuttal comments submitted on the proposed monograph and 
all new data and information submitted pursuant to paragraph (a)(6) of 
this section, all objections submitted on the tentative final monograph 
and all new data and information and comments submitted pursuant to 
paragraph (a)(7) of this section, the complete record of any oral public 
hearing conducted pursuant to paragraph (a)(8) of this section, all 
other comments requested at any time by the Commissioner, all data and 
information for which the Commissioner has reopened the administrative 
record, and all other material that the

[[Page 219]]

Commissioner includes in the administrative record as part of the basis 
for the Commissioner's decision.
    (11) Court appeal. The monograph contained in the final order 
constitutes final agency action from which appeal lies to the courts. 
The Food and Drug Administration will request consolidation of all 
appeals in a single court. Upon court appeal, the Commissioner may, at 
his discretion, stay the effective date for part or all of the monograph 
pending appeal and final court adjudication.
    (12) Amendment of monographs. (i) The Commissioner may propose on 
the Commissioner's own initiative to amend or repeal any monograph 
established pursuant to this section. Any interested person may petition 
the Commissioner for such proposal pursuant to Sec. 10.30 of this 
chapter. The Commissioner may deny the petition if the Commissioner 
finds a lack of safety or effectiveness employing the standards in 
paragraph (a)(4) of this section (in which case the appeal provisions of 
paragraph (a)(11) of this section shall apply), or the Commissioner may 
publish a proposed amendment or repeal in the Federal Register if the 
Commissioner finds general recognition of safety and effectiveness 
employing the standards in paragraph (a)(4) of this section. Any 
interested person may, within 60 days after publication of the proposed 
order in the Federal Register, file with the Dockets Management Branch, 
Food and Drug Administration, written comments in quadruplicate. 
Comments may be accompanied by a memorandum or brief in support thereof. 
All comments may be reviewed in the Dockets Management Branch between 
the hours of 9 a.m. and 4 p.m., Monday through Friday. After reviewing 
the comments, the Commissioner shall publish a final order amending the 
monograph established under the provisions of paragraph (a)(9) of this 
section or withdraw the proposal if comments opposing the amendment are 
persuasive. A new drug application may be submitted in lieu of, or in 
addition to, a petition under this paragraph.
    (ii) A new drug application may be submitted in lieu of a petition 
to amend the OTC drug monograh only if the drug product with the 
condition that is the subject of the new drug application has not been 
marketed on an interim basis (such as under the provisions of paragraph 
(a)(6)(iii) of this section), all clinical testing has been conducted 
pursuant to a new drug application plan, and no marketing of the product 
with the condition for which approval is sought is undertaken prior to 
approval of the new drug application. The Food and Drug Administration 
shall handle a new drug application as a petition for amendment of a 
monograph, and shall review it on that basis, if the provisions of this 
paragraph preclude approval of a new drug application but permit the 
granting of such a petition.
    (b) Regulatory action. Any product which fails to conform to an 
applicable monograph after its effective date is liable to regulatory 
action.
    (c) Information and data submitted under this section shall include, 
with respect to each nonclinical laboratory study contained in the 
application, either a statement that the study was conducted in 
compliance with the good laboratory practice regulations set forth in 
part 58 of this chapter, or, if the study was not conducted in 
compliance with such regulations, a brief statement of the reason for 
the noncompliance.
    (d) [Reserved]
    (e) Institutional review and informed consent. Information and data 
submitted under this section after July 27, 1981, shall include 
statements regarding each clinical investigation involving human 
subjects, from which the information and data are derived, that it 
either was conducted in compliance with the requirements for 
institutional review set forth in part 56 of this chapter, or was not 
subject to such requirements in accordance with Secs. 56.104 or 56.105, 
and that it was conducted in compliance with the requirements for 
informed consent set forth in part 50 of this chapter.
    (f) Financial certification or disclosure statement. Any clinical 
data submitted under this section must be accompanied by financial 
certifications or

[[Page 220]]

disclosure statements or both as required by part 54 of this chapter.
[39 FR 11741, Mar. 29, 1974, as amended at 39 FR 39556, Nov. 8, 1974; 42 
FR 19141, Apr. 12, 1977; 42 FR 54800, Oct. 11, 1977; 46 FR 8460, 8955, 
Jan. 27, 1981; 46 FR 14340, Feb. 27, 1981; 46 FR 21360, Apr. 10, 1981; 
46 FR 47738, Sept. 29, 1981; 50 FR 7516, Feb. 22, 1985; 55 FR 11581, 
Mar. 29, 1990; 63 FR 5253, Feb. 2, 1998]

    Effective Date Note: At 63 FR 5253, Feb. 2, 1998, Sec. 330.10 was 
amended by adding new paragraph (f), effective Feb. 2, 1999.



Sec. 330.11  NDA deviations from applicable monograph.

    A new drug application requesting approval of an OTC drug deviating 
in any respect from a monograph that has become final shall be in the 
form required by Sec. 314.50 of this chapter, but shall include a 
statement that the product meets all conditions of the applicable 
monograph except for the deviation for which approval is requested and 
may omit all information except that pertinent to the deviation.
[39 FR 11741, Mar. 29, 1974, as amended at 55 FR 11581, Mar. 29, 1990]



Sec. 330.12  Status of over-the-counter (OTC) drugs previously reviewed under the Drug Efficacy Study (DESI).

    (a) There were 420 OTC drugs reviewed in the Drug Efficacy Study (a 
review of drugs introduced to the market through new drug procedures 
between 1938 and 1962). A careful review has been made of the reports on 
these drugs to determine those drugs for which implementation may be 
deferred without significant risk to the public health, pending review 
by appropriate OTC drug advisory review panels and promulgation of a 
monograph.
    (b) On and after April 20, 1972, a number of notices were published 
in the Federal Register concerning previously unpublished OTC drugs 
reviewed by the National Academy of Sciences-National Research Council 
Drug Efficacy Study Group. Only the evaluations and comments of the 
panels were published, with no conclusions of the Commissioner of Food 
and Drugs. Those publications were for the purpose of giving interested 
persons the benefit of the Academy's opinions. For those products, and 
also for OTC drug products previously published with the Commissioner's 
conclusions (except for the products listed in paragraphs (b) (1) and 
(2) of this section, all requests for data, revised labeling, requests 
for new drug applications, abbreviated new drug applications, updating 
supplements, data to support less than effective claims, if any, etc., 
are deferred, and such OTC drug products are instead subject to the OTC 
drug review in their appropriate classes pursuant to the procedures 
established in this subpart.
    (1) The requirements of the following DESI announcements are not 
deferred (the reference document may also pertain to prescription 
drugs):
    (i) Certain Surgical Sutures (DESI 4725), published in the Federal 
Register of November 11, l971 (36 FR 21612).
    (ii) Absorbable Dusting Powder (DESI 6264), published in the Federal 
Register of May 25, 1971 (36 FR 9475).
    (iii) Certain Insulin Preparations (DESI 4286), published in the 
Federal Register of April 9, 1971 (36 FR 6842).
    (iv) Sulfo-Van Ointment (DESI 2230), published in the Federal 
Register of October 8, 1970 (35 FR 15860).
    (v) Antiperspirants and Deodorants Containing Neomycin Sulfate (DESI 
11048) for which an order revoking provisions for certification or 
release was published in the Federal Register of December 5, 1972 (37 FR 
25820) and has been stayed by the filing of objections.
    (vi) Thorexin Cough Medicine (DESI 11160) for which a notice of 
opportunity for hearing was published in the Federal Register of 
February 2, 1973 (38 FR 3210).
    (vii) Antibiotic susceptibility discs (DESI 90235) for which an 
order providing for certain discs to be certified and removing 
provisions for certification of other discs was published in the Federal 
Register of September 30, 1972 (37 FR 20525) and has been stayed by the 
filing of objections notice of which was published in the Federal 
Register of March 15, 1973 (38 FR 7007).
    (2) Deferral of requirements is not appropriate when an announcement 
has been published and has been followed by a final order classifying a 
drug either as lacking substantial evidence of effectiveness or as not 
shown to be safe. These products will be removed

[[Page 221]]

from the market, if they have not already been removed. Regulatory 
action will also be undertaken against identical, similar and related 
products (21 CFR 310.6). Deferral of requirements is not appropriate for 
the following (the referenced document may also pertain to prescription 
drugs):
    (i) Certain Sulfonamide-Decongestant Nasal Preparation (DESI 4850), 
for which notice of withdrawal of approval of new drug applications was 
published in the Federal Register of October 24, 1970 (35 FR 16605, 
16606).
    (ii) Eskay's Theranates, containing strychnine, sodium, and calcium 
glycerophosphates, thiamine hydrochloride, alcohol, and phosphoric acid 
(DESI 2220), for which notice of withdrawal of approval of the new drug 
application was published in the Federal Register of February 18, 1971 
(36 FR 3152).
    (iii) The following topical drugs (DESI 1726), for which notice of 
withdrawal of new drug applications was published in the Federal 
Register of August 28, 1971 (36 FR 17368):
    (a) Rhulitol Solution, containing tannic acid, chlorobutanol, 
phenol, camphor, alum, and isopropyl alcohol.
    (b) Zirnox Topical Lotion, containing phenyitoloxamine citrate and 
zirconium oxide.
    (iv) Menacyl Tablets, containing aspirin, menadione, and ascorbic 
acid (DESI 6363), for which notice of withdrawal of approval of the new 
drug application was published in the Federal Register of July 23, 1970 
(35 FR 11827).
    (v) Curad Medicated Adhesive Bandage containing sulfathiazole (DESI 
4964), for which notice of withdrawal of approval of the new drug 
application was published in the Federal Register of December 31, 1969 
(34 FR 20441).
    (vi) Drugs Containing Rutin, Quercetin, Hesperidin, or any 
Bioflavonoids (DESI 5960), for which notice of withdrawal of approval of 
new drug applications was published in the Federal Register of July 3, 
1970 (35 FR 10872, 10873) and October 17, 1970 (35 FR 16332). A further 
notice of opportunity for hearing with respect to the drugs covered by 
the October 17, 1970 Federal Register notice will be published at a 
later date.
    (vii) Antibiotics in Combination with Other Drugs for Nasal Use 
(DESI 7561), for which an order revoking provision for certification was 
published in the Federal Register of August 6, 1971 (36 FR 14469) and 
confirmed in the Federal Register of October 28, 1971 (36 FR 20686).
    (viii) Antibiotic Troches (DESI 8328), for which an order revoking 
provision for certification was published in the Federal Register of 
July 14, 1971 (36 FR 13089) and confirmed in the Federal Register of 
October 9, 1971 (36 FR 19695).
    (ix) Certain Drugs Containing Oxyphenisatin or Oxyphenisatin Acetate 
(DESI 10732), for which notices of withdrawal of approval of new drug 
applications were published in the Federal Register of February 1, 1972 
(37 FR 2460), and March 9, 1973 (38 FR 6419).
    (x) Curad Medicated Adhesive Bandage containing tyrothricin-
nitrofurazone (DESI 6898), for which an order revoking provision for 
certification was published March 14, 1972 (37 FR 5294), and confirmed 
in the Federal Register of July 6, 1972 (37 FR 13254).
    (xi) Candette Cough Gel (DESI 11562), for which notice of withdrawal 
of approval of the new drug application was published in the Federal 
Register of November 19, 1972 (37 FR 25249).
    (xii) Certain OTC Multiple-Vitamin Preparations for Oral Use 
containing excessive amounts of vitamin D and/or vitamin A (DESI 97), 
for which notice of withdrawal of approval of the new drug applications 
was published in the Federal Register of November 29, 1972 (37 FR 
25249).
    (xiii) Certain Sulfonamide-Containing Preparations for Topical 
Ophthalmic or Otic Use (DESI 368, for which a notice of withdrawal of 
approval was published in the Federal Register of February 2, 1973 (38 
FR 3208).
    (xiv) Those parts of the publication entitled ``Certain Mouthwash 
and Gargle Preparations'' (DESI 2855) pertaining to Tyrolaris Mouthwash, 
containing tyrothricin, panthenol, and alcohol, for which an order 
revoking provision for certification was published in the Federal 
Register of February 2, 1967 (32 FR 1172) prior to the drug efficacy 
study implementation.

[[Page 222]]

    (c) Manufacturers and distributors should take notice that the 
information on OTC drugs provided by the Drug Efficacy Study review is 
valuable information as to the deficiencies in the data available to 
support indications for use. They are encouraged to perform studies to 
obtain adequate evidence of effectiveness for the review of OTC drugs 
which is already in progress. In the interim it is in the public 
interest that manufacturers and distributors of all OTC drugs effect 
changes in their formulations and/or labeling to bring the products into 
conformity with current medical knowledge and experience.
    (d) Manufacturers and distributors of OTC drugs may be reluctant to 
make appropriate formulation and/or labeling changes for fear of losing 
the protection of the so-called ``grandfather'' provisions of the 1938 
Federal Food, Drug, and Cosmetic Act (sec. 201(p)(1)) and the 1962 
amendments to the act (sec. 107(c) of those amendments). To encourage 
and facilitate prompt changes, the Food and Drug Administration will not 
take legal action against any OTC drug, other than those not deferred, 
based on a charge that the product is a new drug and not grandfathered 
under the act as a result of the changes if the changes in formulation 
and/or labeling are of the following kind:
    (1) The addition to the labeling of warning, contraindications, side 
effects, and/or precaution information.
    (2) The deletion from the labeling of false, misleading, or 
unsupported indications for use or claims of effectiveness.
    (3) Changes in the components or composition of the drug that will 
give increased assurance that the drug will have its intended effect, 
yet not raise or contribute any added safety questions.
    (4) Changes in the components or composition of the drug which may 
reasonably be concluded to improve the safety of the drug, without 
diminishing its effectiveness.
    (e) The forbearance from legal action for lack of grandfather 
protection is an interim procedure designed to encourage appropriate 
change in formulation and/or labeling during the time period required to 
review the various classes of OTC drugs. At such time as an applicable 
OTC drug monograph becomes effective, the interim procedure will 
automatically be terminated and any appropriate regulatory action will 
be initiated.



Sec. 330.13  Conditions for marketing ingredients recommended for over-the-counter (OTC) use under the OTC drug review.

    (a) Before the publication in the Federal Register of an applicable 
proposed monograph, an OTC drug product that contains: (1) An active 
ingredient limited, on or after May 11, 1972, to prescription use for 
the indication and route of administration under consideration by an OTC 
advisory review panel, and not thereafter exempted from such limitation 
pursuant to Sec. 310.200 of this chapter, or
    (2) An active ingredient at a dosage level higher than that 
available in an OTC drug product on December 4, 1975, shall be regarded 
as a new drug within the meaning of section 201(p) of the act for which 
an approved new drug application is required.
    (b)(1) An OTC drug product that contains: (i) An active ingredient 
limited, on or after May 11, 1972, to prescription use for the 
indication and route of administration under consideration by an OTC 
advisory review panel, and not thereafter exempted from such limitation 
pursuant to Sec. 310.200 of this chapter, or
    (ii) An active ingredient at a dosage level higher than that 
available in an OTC drug product on December 4, 1975, which ingredient 
and/or dosage level is classified by the panel in category I (conditions 
subject to Sec. 330.10(a)(6)(i)) shall be regarded as a new drug within 
the meaning of section 201(p) of the act for which an approved new drug 
application is required if marketed for OTC use prior to the date of 
publication in the Federal Register of a proposed monograph.
    (2) An OTC drug product covered by paragraph (b)(1) of this section 
which is marketed after the date of publication in the Federal Register 
of a proposed monograph but prior to the effective date of a final 
monograph shall be subject to the risk that the Commissioner

[[Page 223]]

may not accept the panel's recommendation and may instead adopt a 
different position that may require relabeling, recall, or other 
regulatory action. The Commissioner may state such position at any time 
by notice in the Federal Register, either separately or as part of 
another document; appropriate regulatory action will commence 
immediately and will not await publication of a final monograph. 
Marketing of such a product with a formulation or labeling not in accord 
with a proposed monograph or tentative final monograph also may result 
in regulatory action against the product, the marketer, or both.
    (c) An OTC drug product that contains: (1) An active ingredient 
limited, on or after May 11, 1972, to prescription use for the 
indication and route of administration under consideration by an OTC 
advisory review panel, and not thereafter exempted from such limitation 
pursuant to Sec. 310.200 of this chapter, or
    (2) An active ingredient at a dosage level higher than that 
available in any OTC drug product on December 4, 1975, which ingredient 
and/or dosage level is classified by the panel in category II 
(conditions subject to Sec. 330.10(a)(6)(ii)), may be marketed only 
after:
    (i) The Center for Drug Evaluation and Research or the Commissioner 
tentatively determines that the ingredient is generally recognized as 
safe and effective, and the Commissioner states by notice in the Federal 
Register (separately or as part of another document) that marketing 
under specified conditions will be permitted;
    (ii) The ingredient is determined by the Commissioner to be 
generally recognized as safe and effective and is included in the 
appropriate published OTC drug final monograph; or
    (iii) A new drug application for the product has been approved.
    (d) An OTC drug product that contains: (1) An active ingredient 
limited, on or after May 11, 1972, to prescription use for the 
indication and route of administration under consideration by an OTC 
advisory review panel, and not thereafter exempted from such limitation 
pursuant to Sec. 310.200 of this chapter, or
    (2) An active ingredient at a dosage level higher than that 
available in any OTC drug product on December 4, 1975, which ingredient 
and/or dosage level is classified by the panel in category III 
(conditions subject to Sec. 330.10(a)(6)(iii)), may be marketed only 
after:
    (i) The Center for Drug Evaluation and Research or the Commissioner 
tentatively determines that the ingredient is generally recognized as 
safe and effective, and the Commissioner states by notice in the Federal 
Register (separately or as part of another document) that marketing 
under specified conditions will be permitted;
    (ii) The ingredient is determined by the Commissioner to be 
generally recognized as safe and effective and is included in the 
appropriate published OTC drug final monograph; or
    (iii) A new drug application for the product has been approved.
[41 FR 32582, Aug. 4, 1976, as amended at 47 FR 17739, Apr. 23, 1982; 50 
FR 8996, Mar. 6, 1985; 55 FR 11581, Mar. 29, 1990]



PART 331--ANTACID PRODUCTS FOR OVER-THE-COUNTER (OTC) HUMAN USE--Table of Contents




                      Subpart A--General Provisions

Sec.
331.1  Scope.

                      Subpart B--Active Ingredients

331.10  Antacid active ingredients.
331.11  Listing of specific active ingredients.
331.15  Combination with nonantacid active ingredients.

                      Subpart C--Testing Procedures

331.20  Determination of percent contribution of active ingredients.
331.21  Test Modifications.

                           Subpart D--Labeling

331.30  Labeling of antacid products.
331.80  Professional labeling.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  39 FR 19874, June 4, 1974, unless otherwise noted.

[[Page 224]]



                      Subpart A--General Provisions



Sec. 331.1  Scope.

    An over-the-counter antacid product in a form suitable for oral 
administration is generally recognized as safe and effective and is not 
misbranded if it meets each of the following conditions and each of the 
general conditions established in Sec. 330.1 of this chapter.



                      Subpart B--Active Ingredients



Sec. 331.10  Antacid active ingredients.

     (a) The active antacid ingredients of the product consist of one or 
more of the ingredients permitted in Sec. 331.11 within any maximum 
daily dosage limit established, each ingredient is included at a level 
that contributes at least 25 percent of the total acid neutralizing 
capacity of the product, and the finished product contains at least 5 
meq of acid neutralizing capacity as measured by the procedure provided 
in the United States Pharmacopeia 23/National Formulary 18. The method 
established in Sec. 331.20 shall be used to determine the percent 
contribution of each antacid active ingredient.
    (b) This section does not apply to an antacid ingredient 
specifically added as a corrective to prevent a laxative or constipating 
effect.
[39 FR 19874, June 4, 1974, as amended at 61 FR 4822, Feb. 8, 1996]



Sec. 331.11  Listing of specific active ingredients.

    (a) Aluminum-containing active ingredients:
    (1) Basic aluminum carbonate gel.
    (2) Aluminum hydroxide (or as aluminum hydroxide-hexitol stabilized 
polymer, aluminum hydroxide-magnesium carbonate codried gel, aluminum 
hydroxide-magnesium trisilicate codried gel, aluminum-hydroxide sucrose 
powder hydrated).
    (3) Dihydroxyaluminum aminoacetate and dihydroxyaluminum aminoacetic 
acid.
    (4) Aluminum phosphate gel when used as part of an antacid 
combination product and contributing at least 25 percent of the total 
acid neutralizing capacity; maximum daily dosage limit is 8 grams.
    (5) Dihydroxyaluminum sodium carbonate.
    (b) Bicarbonate-containing active ingredients: Bicarbonate ion; 
maximum daily dosage limit 200 mEq. for persons up to 60 years old and 
100 mEq. for persons 60 years or older.
    (c) Bismuth-containing active ingredients:
    (1) Bismuth aluminate.
    (2) Bismuth carbonate.
    (3) Bismuth subcarbonate.
    (4) Bismuth subgallate.
    (5) Bismuth subnitrate.
    (d) Calcium-containing active ingredients: Calcium, as carbonate or 
phosphate; maximum daily dosage limit 160 mEq. calcium (e.g., 8 grams 
calcium carbonate).
    (e) Citrate-containing active ingredients: Citrate ion, as citric 
acid or salt; maximum daily dosage limit 8 grams.
    (f) Glycine (aminoacetic acid).
    (g) Magnesium-containing active ingredients:
    (1) Hydrate magnesium aluminate activated sulfate.
    (2) Magaldrate.
    (3) Magnesium aluminosilicates.
    (4) Magnesium carbonate.
    (5) Magnesium glycinate.
    (6) Magnesium hydroxide.
    (7) Magnesium oxide.
    (8) Magnesium trisilicate.
    (h) Milk solids, dried.
    (i) Phosphate-containing active ingredients:
    (1) Aluminum phosphate; maximum daily dosage limit 8 grams.
    (2) Mono or dibasic calcium salt; maximum daily dosage limit 2 
grams.
    (3) Tricalcium phosphate; maximum daily dosage limit 24 grams.
    (j) Potassium-containing active ingredients:
    (1) Potassium bicarbonate (or carbonate when used as a component of 
an effervescent preparation); maximum daily dosage limit 200 mEq. of 
bicarbonate ion for persons up to 60 years old and 100 mEq. of 
bicarbonate ion for persons 60 years or older.
    (2) Sodium potassium tartrate.
    (k) Sodium-containing active ingredients:
    (1) Sodium bicarbonate (or carbonate when used as a component of an 
effervescent preparation); maximum daily dosage limit 200 mEq. of sodium 
for

[[Page 225]]

persons up to 60 years old and 100 mEq. of sodium for persons 60 years 
or older, and 200 mEq. of bicarbonate ion for persons up to 60 years old 
and 100 mEq. of bicarbonate ion for persons 60 years or older. That part 
of the warning required by Sec. 330.1(g), which states, ``Keep this and 
all drugs out of the reach of children'' is not required on a product 
which contains only sodium bicarbonate powder and which is intended 
primarily for other than drug uses.
    (2) Sodium potassium tartrate.
    (l) Silicates:
    (1) Magnesium aluminosilicates.
    (2) Magnesium trisilicate.
    (m) Tartrate-containing active ingredients. Tartaric acid or its 
salts; maximum daily dosage limit 200 mEq. (15 grams) of tartrate.
[39 FR 19874, June 4, 1974, as amended at 51 FR 27763, Aug. 1, 1986; 55 
FR 19859, May 11, 1990]



Sec. 331.15  Combination with nonantacid active ingredients.

    (a) An antacid may contain any generally recognized as safe and 
effective nonantacid laxative ingredient to correct for constipation 
caused by the antacid. No labeling claim of the laxative effect may be 
used for such a product.
    (b) An antacid may contain any generally recognized as safe and 
effective analgesic ingredient(s), if it is indicated for use solely for 
the concurrent symptoms involved, e.g., headache and acid indigestion, 
and is marketed in a form intended for ingestion as a solution.
    (c) An antacid may contain any generally recognized as safe and 
effective antiflatulent ingredient if it is indicated for use solely for 
the concurrent symptoms of gas associated with heartburn, sour stomach 
or acid indigestion.



                      Subpart C--Testing Procedures



Sec. 331.20  Determination of percent contribution of active ingredients.

    To determine the percent contribution of an antacid active 
ingredient, place an accurately weighed amount of the antacid active 
ingredient equal to the amount present in a unit dose of the product 
into a 250-milliliter (mL) beaker. If wetting is desired, add not more 
than 5 mL of alcohol (neutralized to an apparent pH of 3.5), and mix to 
wet the sample thoroughly. Add 70 mL of water, and mix on a magnetic 
stirrer at 30030 r.p.m. for 1 minute. Analyze the acid 
neutralizing capacity of the sample according to the procedure provided 
in the United States Pharmacopeia 23/National Formulary 18 and calculate 
the percent contribution of the antacid active ingredient in the total 
product as follows:
    Percent contribution = (Total mEq. Antacid Active Ingredient x100)/
(Total mEq. Antacid Product).
[61 FR 4823, Feb. 8, 1996]



Sec. 331.21  Test modifications.

    The formulation or mode of administration of certain products may 
require a modification of the United States Pharmacopeia 23/National 
Formulary 18 acid neutralizing capacity test. Any proposed modification 
and the data to support it shall be submitted as a petition under the 
rules established in Sec. 10.30 of this chapter. All information 
submitted will be subject to the disclosure rules in part 20 of this 
chapter.
[61 FR 4823, Feb. 8, 1996]



                           Subpart D--Labeling



Sec. 331.30  Labeling of antacid products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as an 
``antacid.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the following: ``For the relief of'' (optional, 
any or all of the following:) ``heartburn,'' ``sour stomach,'' and/or 
``acid indigestion'' (which may be followed by the optional statement:) 
``and upset stomach associated with'' (optional, as appropriate) ``this 
symptom'' or ``these symptoms.'' Other truthful and nonmisleading 
statements, describing only the indications for use that have been 
established and listed in this paragraph (b), may also be used, as 
provided in Sec. 330.1(c)(2) of this chapter, subject to the provisions 
of section 502 of the act relating to misbranding and the prohibition in 
section 301(d) of the act against the introduction or delivery for 
introduction

[[Page 226]]

into interstate commerce of unapproved new drugs in violation of section 
505(a) of the act.
    (c) Warnings. The labeling of the product contains the following 
warnings, under the heading ``Warnings'', which may be combined but not 
rearranged to eliminate duplicative words or phrases if the resulting 
warning is clear and understandable:
    (1) ``Do not take more than (maximum recommended daily dosage, 
broken down by age groups if appropriate, expressed in units such as 
tablets or teaspoonfuls) in a 24-hour period, or use the maximum dosage 
of this product for more than 2 weeks, except under the advice and 
supervision of a physician.''
    (2) For products which cause constipation in 5 percent or more of 
persons who take the maximum recommended dosage: ``May cause 
constipation.''
    (3) For products which cause laxation in 5 percent or more of 
persons who take the maximum recommended dosage: ``May have laxative 
effect.''
    (4) For products containing more than 50 mEq. of magnesium in the 
recommended daily dosage: ``Do not use this product except under the 
advice and supervision of a physician if you have kidney disease.''
    (5) For products containing more than 25 mEq. potassium in the 
maximum recommended daily dose: ``Do not use this product except under 
the advice and supervision of a physician if you have kidney disease.''
    (6) For products containing more than 5 gm per day lactose in a 
maximum daily dosage: ``Do not use this product except under advice and 
supervision of a physician if you are allergic to milk or milk 
products.''
    (d) Drug interaction precaution. The labeling of the product 
contains the following statements under the heading ``Drug Interaction 
Precaution'': ``Antacids may interact with certain prescription drugs. 
If you are presently taking a prescription drug, do not take this 
product without checking with your physician or other health 
professional.''
    (e) Directions for use. The labeling of the product contains the 
recommended dosage, under the heading ``Directions'', per time interval 
(e.g., every 4 hours) or time period (e.g., 4 times a day) broken down 
by age groups if appropriate, followed by ``or as directed by a 
physician.''
    (f) Exemption from the general accidental overdose warning. The 
labeling for antacid drug products containing the active ingredients 
identified in Sec. 331.11(a), (b), and (d) through (m); permitted 
combinations of these ingredients provided for in Sec. 331.10; and any 
of these ingredients or combinations of these ingredients in combination 
with simethicone (identified in Sec. 332.10 of this chapter and provided 
for in Sec. 331.15(c)), are exempt from the requirement in Sec. 330.1(g) 
of this chapter that the labeling bear the general warning statement 
``In case of accidental overdose, seek professional assistance or 
contact a poison control center immediately.'' With the exception of 
sodium bicarbonate powder products identified in Sec. 331.11(k)(1), the 
labeling must continue to bear the first part of the general warning in 
Sec. 330.1(g) of this chapter, which states, ``Keep this and all drugs 
out of the reach of children.''
    (g) [Reserved]
    (h) The word ``doctor'' may be substituted for the word 
``physician'' in any of the labeling statements in this section.
[39 FR 19874, June 4, 1974, as amended at 47 FR 38484, Aug. 31, 1982; 51 
FR 16266, May 1, 1986; 51 FR 27763, Aug. 1, 1986; 52 FR 7830, Mar. 13, 
1987; 55 FR 11581, Mar. 29, 1990; 58 FR 45208, Aug. 26, 1993; 59 FR 
60556, Nov. 25, 1994; 61 FR 17806, Apr. 22, 1996]



Sec. 331.80  Professional labeling.

    (a) The labeling of the product provided to health professionals 
(but not to the general public):
    (1) Shall contain the neutralizing capacity of the product as 
calculated using the procedure set forth in United States Pharmacopeia 
23/National Formulary 18 expressed in terms of the dosage recommended 
per minimum time interval or, if the labeling recommends more than one 
dosage, in terms of the minimum dosage recommended per minimum time 
interval.
    (2) May contain an indication for the symptomatic relief of 
hyperacidity associated with the diagnosis of peptic

[[Page 227]]

ulcer, gastritis, peptic esophagitis, gastric hyperacidity, and hiatal 
hernia.
    (3) For products containing basic aluminum carbonate gel identified 
in Sec. 331.11(a)(1)--Indication. ``For the treatment, control, or 
management of hyperphosphatemia, or for use with a low phosphate diet to 
prevent formation of phosphate urinary stones, through the reduction of 
phosphates in the serum and urine.''
    (4) For products containing aluminum identified in Sec. 331.11(a)--
Warnings. (i) Prolonged use of aluminum-containing antacids in patients 
with renal failure may result in or worsen dialysis osteomalacia. 
Elevated tissue aluminum levels contribute to the development of the 
dialysis encephalopathy and osteomalacia syndromes. Small amounts of 
aluminum are absorbed from the gastrointestinal tract and renal 
excretion of aluminum is impaired in renal failure. Aluminum is not well 
removed by dialysis because it is bound to albumin and transferrin, 
which do not cross dialysis membranes. As a result, aluminum is 
deposited in bone, and dialysis osteomalacia may develop when large 
amounts of aluminum are ingested orally by patients with impaired renal 
function.
    (ii) Aluminum forms insoluble complexes with phosphate in the 
gastrointestinal tract, thus decreasing phosphate absorption. Prolonged 
use of aluminum-containing antacids by normophosphatemic patients may 
result in hypophosphatemia if phosphate intake is not adequate. In its 
more severe forms, hypophosphatemia can lead to anorexia, malaise, 
muscle weakness, and osteomalacia.
    (b) Professional labeling for an antacid-antiflatulent combination 
may contain the information allowed for health professionals for 
antacids and antiflatulents.
[39 FR 19874, June 4, 1974. Redesignated and amended at 55 FR 19859, May 
11, 1990]



PART 332--ANTIFLATULENT PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                      Subpart A--General Provisions

Sec.
332.1  Scope.
332.3  Definitions.

                      Subpart B--Active Ingredients

332.10  Antiflatulent active ingredients.
332.15  Combination with non-antiflatulent active ingredients.

                           Subpart C--Labeling

332.30  Labeling of antiflatulent products.
332.31  Professional labeling.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  39 FR 19877, June 4, 1974, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 332.1  Scope.

    An over-the-counter antiflatulent product in a form suitable for 
oral administration is generally recognized as safe and effective and is 
not misbranded if it meets each of the following conditions and each of 
the general conditions established in Sec. 330.1 of this chapter.



Sec. 332.3  Definitions.

    As used in this part:
    Antigas. A term that may be used interchangeably with the term 
antiflatulent. Neither term should be considered as describing the 
mechanism of action of the active ingredient contained in the product.
[61 FR 8838, Mar. 5, 1996]



                      Subpart B--Active Ingredients



Sec. 332.10  Antiflatulent active ingredients.

    Simethicone; maximum daily dose 500 mg. There is no dosage 
limitation at this time for professional labeling.



Sec. 332.15  Combination with non-antiflatulent active ingredients.

    An antiflatulent may contain any generally recognized as safe and 
effective antacid ingredient(s) if it is indicated for use solely for 
the concurrent symptoms of gas associated with heartburn, sour stomach 
or acid indigestion.

[[Page 228]]



                           Subpart C--Labeling



Sec. 332.30  Labeling of antiflatulent drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as an 
``antiflatulent,'' ``antigas,'' or ``antiflatulent (antigas).''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' one or more of the phrases listed in this 
paragraph (b), as appropriate. Other truthful and nonmisleading 
statements, describing only the indications for use that have been 
established and listed in this paragraph (b), may also be used, as 
provided in Sec. 330.1(c)(2) of this chapter, subject to the provisions 
of section 502 of the Federal Food, Drug, and Cosmetic Act (the act) 
relating to misbranding and the prohibition in section 301(d) of the act 
against the introduction or delivery for introduction into interstate 
commerce of unapproved new drugs in violation of section 505(a) of the 
act.
    (1) (Select one of the following: ``Alleviates or Relieves'') ``the 
symptoms referred to as gas.''
    (2) (Select one of the following: ``Alleviates'' or ``Relieves'') 
(select one or more of the following: ``bloating,'' ``pressure,'' 
``fullness,'' or ``stuffed feeling'') ``commonly referred to as gas.''
    (c) Exemption from the general accidental overdose warning. The 
labeling for antiflatulent drug products containing simethicone 
identified in Sec. 332.10 and antacid/antiflatulent combination drug 
products provided for in Sec. 332.15, containing the active ingredients 
identified in Sec. 331.11(a), (b), and (d) through (m) of this chapter 
are exempt from the requirement in Sec. 330.1(g) of this chapter that 
the labeling bear the general warning statement ``In case of accidental 
overdose, seek professional assistance or contact a poison control 
center immediately.'' The labeling must continue to bear the first part 
of the general warning in Sec. 330.1(g) of this chapter, which states, 
``Keep this and all drugs out of the reach of children.''
[39 FR 19877, June 4, 1974, as amended at 40 FR 11719, Mar. 13, 1975; 51 
FR 16266, May 1, 1986; 51 FR 27763, Aug. 1, 1986; 52 FR 7830, Mar. 13, 
1987; 61 FR 8838, Mar. 5, 1996]



Sec. 332.31  Professional labeling.

    (a) The labeling of the product provided to health professionals 
(but not to the general public) may contain as additional indications 
postoperative gas pain or for use in endoscopic examination.
    (b) Professional labeling for an antiflatulent-antacid combination 
may contain information allowed for health professionals for antacids 
and antiflatulents.



PART 333--TOPICAL ANTIMICROBIAL DRUG PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                          Subpart A  [Reserved]

              Subpart B--First Aid Antibiotic Drug Products

Sec.
333.101  Scope.
333.103  Definitions.
333.110  First aid antibiotic active ingredients.
333.120  Permitted combinations of active ingredients.
333.150  Labeling of first aid antibiotic drug products.
333.160  Labeling of permitted combinations of active ingredients.

               Subpart C--Topical Antifungal Drug Products

333.201  Scope.
333.203  Definitions.
333.210  Antifungal active ingredients.
333.250  Labeling of antifungal drug products.
333.280  Professional labeling.

                  Subpart D--Topical Acne Drug Products

333.301  Scope.
333.303  Definitions.
333.310  Acne active ingredients.
333.320  Permitted combinations of active ingredients.
333.350  Labeling of acne drug products.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  52 FR 47322, Dec. 11, 1987, unless otherwise noted.

[[Page 229]]



                          Subpart A--[Reserved]



              Subpart B--First Aid Antibiotic Drug Products



Sec. 333.101  Scope.

    (a) An over-the-counter first aid antibiotic drug product in a form 
suitable for topical administration is generally recognized as safe and 
effective and is not misbranded if it meets each of the conditions in 
this subpart and each of the general conditions established in 
Sec. 330.1.
    (b) References in this subpart to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 333.103  Definitions.

    As used in this subpart:
    (a) Antibiotic drug. In accordance with section 507(a) of the 
Federal Food, Drug, and Cosmetic Act (21 U.S.C. 357(a)), ``any drug 
intended for use by man containing any quantity of any chemical 
substance which is produced by a microorganism and which has the 
capacity to inhibit or destroy microorganisms in dilute solution 
(including the chemically synthesized equivalent of any such 
substance).''
    (b) First aid antibiotic. An antibiotic-containing drug product 
applied topically to the skin to help prevent infection in minor cuts, 
scrapes, and burns.



Sec. 333.110  First aid antibiotic active ingredients.

    The product consists of any of the following active ingredients 
within the specified concentration established for each ingredient and 
in the specified dosage form:
    (a) Bacitracin ointment containing, in each gram, 500 units of 
bacitracin in a suitable ointment base: Provided, That it meets the 
tests and methods of assay in Sec. 448.510a(b).
    (b) Bacitracin zinc ointment containing, in each gram, 500 units of 
bacitracin zinc in a suitable ointment base: Provided, That it meets the 
tests and methods of assay in Sec. 448.513f(b).
    (c) Chlortetracycline hydrochloride ointment containing, in each 
gram, 30 milligrams of chlortetracycline hydrochloride in a suitable 
ointment base: Provided, That it meets the tests and methods of assay in 
Sec. 446.510(b).
    (d) Neomycin sulfate ointment containing, in each gram, 3.5 
milligrams of neomycin in a suitable water soluble or oleaginous 
ointment base: Provided, That it meets the tests and methods of assay in 
Sec. 444.542a(b).
    (e) Neomycin sulfate cream containing, in each gram, 3.5 milligrams 
of neomycin in a suitable cream base: Provided, That it meets the tests 
and methods of assay in Sec. 444.542b(b).
    (f) Tetracycline hydrochloride ointment containing, in each gram, 30 
milligrams of tetracycline hydrochloride in a suitable ointment base: 
Provided, That it meets the tests and methods of assay in 
Sec. 446.581d(b).
[52 FR 47322, Dec. 11, 1987, as amended at 53 FR 18838, May 25, 1988]



Sec. 333.120  Permitted combinations of active ingredients.

    The following combinations are permitted provided each active 
ingredient is present within the established concentration and in the 
specified dosage form, and the product is labeled in accordance with 
Sec. 333.160.
    (a) Combinations of antibiotic active ingredients. (1) Bacitracin-
neomycin sulfate ointment containing, in each gram, 500 units of 
bacitracin and 3.5 milligrams of neomycin in a suitable ointment base: 
Provided, That it meets the tests and methods of assay in 
Sec. 448.510d(b).
    (2) Bacitracin-neomycin sulfate-polymyxin B sulfate ointment 
containing, in each gram, in a suitable ointment base the following:
    (i) 500 units of bacitracin, 3.5 milligrams of neomycin, and 5,000 
units of polymyxin B; or
    (ii) 400 units of bacitracin, 3.5 milligrams of neomycin, and 5,000 
units of polymyxin B;

Provided, That it meets the tests and methods of assay in 
Sec. 448.510e(b).
    (3) Bacitracin-polymyxin B sulfate topical aerosol containing, in 
each gram, 500 units of bacitracin and 5,000 units of polymyxin B in a 
suitable vehicle, packaged in a pressurized container with suitable 
inert gases: Provided, That it meets the tests and methods of assay in 
Sec. 448.510f(b).

[[Page 230]]

    (4) Bacitracin zinc-neomycin sulfate ointment containing, in each 
gram, 500 units of bacitracin and 3.5 milligrams of neomycin in a 
suitable ointment base: Provided, That it meets the tests and methods of 
assay in Sec. 448.513b(b).
    (5) Bacitracin zinc-neomycin sulfate-polymyxin B sulfate ointment 
containing, in each gram, in a suitable ointment base the following:
    (i) 400 units of bacitracin, 3 milligrams of neomycin, and 8,000 
units of polymyxin B; or
    (ii) 400 units of bacitracin, 3.5 milligrams of neomycin, and 5,000 
units of polymyxin B; or
    (iii) 500 units of bacitracin, 3.5 milligrams of neomycin, and 5,000 
units of polymyxin B; or
    (iv) 500 units of bacitracin, 3.5 milligrams of neomycin, and 10,000 
units of polymyxin B;

Provided, That it meets the tests and methods of assay in 
Sec. 448.513c(b).
    (6) Bacitracin zinc-polymyxin B sulfate ointment containing, in each 
gram, 500 units of bacitracin and 10,000 units of polymyxin B in a 
suitable ointment base: Provided, That it meets the tests and methods 
assay in Sec. 448.513a(b).
    (7) Bacitracin zinc-polymyxin B sulfate topical aerosol containing, 
in each gram, 120 units of bacitracin and 2,350 units of polymyxin B in 
a suitable vehicle, packaged in a pressurized container with suitable 
inert gases: Provided, That is meets the tests and methods of assay in 
Sec. 448.513e(b) of this chapter.
    (8) Bacitracin zinc-polymyxin B sulfate topical powder containing, 
in each gram, 500 units of bacitracin and 10,000 units of polymyxin B in 
a suitable base: Provided, That it meets the tests and methods of assay 
in Sec. 448.513d(b).
    (9) Neomycin sulfate-polymyxin B sulfate ointment containing, in 
each gram, 3.5 milligrams of neomycin and 5,000 units of polymyxin B in 
a suitable water miscible base: Provided, That it meets the tests and 
methods of assay in Sec. 444.542e(b).
    (10) Neomycin sulfate-polymyxin B sulfate cream containing, in each 
gram, 3.5 milligrams of neomycin and 10,000 units of polymyxin B in a 
suitable vehicle: Provided, That it meets the tests, methods of assay, 
and potency in Sec. 444.5421(b).
    (11) Oxytetracycline hydrochloride-polymyxin B sulfate ointment 
containing, in each gram, 30 milligrams of oxytetracycline and 10,000 
units of polymyxin B in a suitable ointment base: Provided, That it 
meets the tests and methods assay in Sec. 446.567b(b).
    (12) Oxytetracycline hydrochloride-polymyxin B sulfate topical 
powder containing, in each gram, 30 milligrams of oxytetracycline and 
10,000 units of polymyxin B with a suitable filler: Provided, That it 
meets the tests and methods assay in Sec. 446.567c(b).
    (b) Combinations of first aid antibiotic active ingredients and 
local anesthetic active ingredients.
    (1) Bacitracin ointment containing, in each gram, 500 units of 
bacitracin and any single generally recognized as safe and effective 
amine or ``caine''-type local anesthetic active ingredient in a suitable 
ointment base: Provided, That it meets the tests and methods of assay in 
Sec. 448.510a(b).
    (2) Bacitracin-neomycin sulfate-polymyxin B sulfate ointment 
containing, in each gram, in a suitable ointment base the following:
    (i) 500 units of bacitracin, 3.5 milligrams of neomycin, 5,000 units 
of polymyxin B, and any single generally recognized as safe and 
effective amine or ``caine''-type local anesthetic active ingredient; or
    (ii) 400 units of bacitracin, 3.5 milligrams of neomycin, 5,000 
units of polymyxin B, and any single generally recognized as safe and 
effective amine or ``caine''-type local anesthetic active ingredient.

Provided, That it meets the tests and methods of assay in 
Sec. 448.510e(b).
    (3) Bacitracin-polymyxin B sulfate topical aerosol containing, in 
each gram, 500 units of bacitracin and 5,000 units of polymyxin B and 
any single generally recognized as safe and effective amine or 
``caine''-type local anesthetic active ingredient in a suitable vehicle, 
packaged in a pressurized container with suitable inert gases: Provided, 
That it meets the tests and methods of assay in Sec. 448.510f(b) of this 
chapter.
    (4) Bacitracin zinc-neomycin sulfate-polymyxin B sulfate ointment 
containing, in each gram, in a suitable ointment base the following:

[[Page 231]]

    (i) 400 units of bacitracin, 3 milligrams of neomycin, 8,000 units 
of polymyxin B, and any single generally recognized as safe and 
effective amine or ``caine''-type local anesthetic active ingredient; or
    (ii) 400 units of bacitracin, 3.5 milligrams of neomycin, 5,000 
units of polymyxin B, and any single generally recognized as safe and 
effective amine or ``caine''-type local anesthetic active ingredient; or
    (iii) 500 units of bacitracin, 3.5 milligrams of neomycin, 5,000 
units of polymyxin B, and any single generally recognized as safe and 
effective amine or ``caine''-type local anesthetic active ingredient; or
    (iv) 500 units of bacitracin, 3.5 milligrams of neomycin, 10,000 
units of polymyxin B, and any single generally recognized as safe and 
effective amine or ``caine''-type local anesthetic active ingredieint;

Provided, That it meets the tests and methods of assay in 
Sec. 448.513c(b) of this chapter.
    (5) Bacitracin zinc-polymyxin B sulfate ointment containing, in each 
gram, 500 units of bacitracin, 10,000 units of polymyxin B, and any 
single generally recognized as safe and effective amine or ``caine''-
type local anesthetic active ingredient in a suitable ointment base: 
Provided, That it meets the tests and methods of assay in 
Sec. 448.513a(b) of this chapter.
    (6) Neomycin sulfate-polymyxin B sulfate cream containing, in each 
gram, 3.5 milligrams of neomycin, 10,000 units of polymyxin B, and any 
single generally recognized as safe and effective amine or ``caine''-
type local anesthetic active ingredient in a suitable vehicle: Provided, 
That it meets the tests and methods of assay in Sec. 444.542l(b) of this 
chapter.
[52 FR 47322, Dec. 11, 1987; 52 FR 48792, Dec. 24, 1987, as amended at 
53 FR 18838, May 25, 1988; 55 FR 9722, Mar. 15, 1990; 55 FR 40381, Oct. 
3, 1990; 55 FR 50172, Dec. 5, 1990]



Sec. 333.150  Labeling of first aid antibiotic drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as a 
``first aid antibiotic.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the following: ``First aid to help'' [select 
one of the following: ``prevent,'' (``decrease'' (``the risk of'' or 
``the chance of'')), (``reduce'' (``the risk of'' or ``the chance 
of'')), ``guard against,'' or ``protect against''] [select one of the 
following: ``infection,'' ``bacterial contamination,'' or ``skin 
infection''] ``in minor cuts, scrapes, and burns.'' Other truthful and 
nonmisleading statements describing only the indications for use that 
have been established and listed in this paragraph (b), may also be 
used, as provided in Sec. 330.1(c)(2), subject to the provisions of 
section 502 of the act relating to misbranding and the prohibition in 
section 301(d) of the act against the introduction or delivery for 
introduction into interstate commerce of unapproved new drugs in 
violation of section 505(a) of the act.
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) ``For external use only. Do not use in the eyes or apply over 
large areas of the body. In case of deep or puncture wounds, animal 
bites, or serious burns, consult a doctor.''
    (2) For products containing chlortetracycline hydrochloride or 
tetracycline hydrochloride.``Stop use and consult a doctor if the 
condition persists or gets worse. Do not use longer than 1 week unless 
directed by doctor.''
    (3) For any product containing bacitracin, bacitracin zinc, 
neomycin, neomycin sulfate, polymyxin B, and/or polymyxin B sulfate. 
``Stop use and consult a doctor if the condition persists or gets worse, 
or if a rash or other allergic reaction develops. Do not use if you are 
allergic to any of the ingredients. Do not use longer than 1 week unless 
directed by a doctor.''
    (d) Directions. The labeling of the product contains the following 
statements under the heading ``Directions'': (1) For ointment and cream 
products. ``Clean the affected area. Apply a small amount of this 
product (an amount equal to the surface area of the tip of a finger) on 
the area 1 to 3 times daily. May be covered with a sterile bandage.''

[[Page 232]]

    (2) For powder products. ``Clean the affected area. Apply a light 
dusting of the powder on the area 1 to 3 times daily. May be covered 
with a sterile bandage.''
    (3) For aerosol products. ``Clean the affected area. Spray a small 
amount of this product on the area 1 to 3 times daily. May be covered 
with a sterile bandage.''
    (e) The word ``doctor'' may be substituted for the word 
``physician'' in any of the labeling statements in this subpart.
[52 FR 47332, Dec. 11, 1987, as amended at 61 FR 58472, Nov. 15, 1996]



Sec. 333.160  Labeling of permitted combinations of active ingredients.

    Statements of identity, indications, warnings, and directions for 
use, respectively, applicable to each ingredient in the product may be 
combined to eliminate duplicative words or phrases so that the resulting 
information is clear and understandable.
    (a) Statement of identity. For a combination drug product that has 
an established name, the labeling of the product states the established 
name of the combination drug product, followed by the statement of 
identity for each ingredient in the combination, as established in the 
statement of identity sections of the applicable OTC drug monographs. 
For a combination drug product that does not have an established name, 
the labeling of the product states the statement of identity for each 
ingredient in the combination, as established in the statement of 
identity sections of the applicable OTC drug monographs.
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the indication(s) for each ingredient in the 
combination, as established in the ``Indications'' sections of the 
applicable OTC drug monographs, unless otherwise stated in this 
paragraph. Other truthful and nonmisleading statements, describing only 
the indications for use that have been established and listed in this 
paragraph (b), may also be used, as provided in Sec. 330.1(c)(2), 
subject to the provisions of section 502 of the act relating to 
misbranding and the prohibition in section 301(d) of the act against the 
introduction or delivery for introduction into interstate commerce of 
unapproved new drugs in violation of section 505(a) of the act.
    (1) For permitted combinations identified in Sec. 333.120(a). The 
indications in Sec. 333.150 should be used.
    (2) For permitted combinations identified in Sec. 333.120(b). In 
addition to the required indication identified in Sec. 333.150, the 
labeling of the product may state, under the heading ``Indications,'' 
the following additional indication: ``First aid for the temporary 
relief of'' (select one of the following: ``pain,'' ``discomfort,'' 
``pain or discomfort'' or ``pain and itching'') ``in minor cuts, 
scrapes, and burns.''
    (c) Warnings. The labeling of the product states, under the heading 
``Warnings,'' the warning(s) for each ingredient in the combination, as 
established in the warnings sections of the applicable OTC drug 
monographs.
    (d) Directions. The labeling of the product states, under the 
heading ``Directions,'' directions that conform to the directions 
established for each ingredient in the directions sections of the 
applicable OTC drug monographs. When the time intervals or age 
limitations for administrations of the individual ingredients differ, 
the directions for the combination product may not exceed any maximum 
dosage limits established for the individual ingredients in the 
applicable OTC drug monograph.



               Subpart C--Topical Antifungal Drug Products

    Source:  58 FR 49898, Sept. 23, 1993, unless otherwise noted.



Sec. 333.201  Scope.

    (a) An over-the-counter antifungal drug product in a form suitable 
for topical administration is generally recognized as safe and effective 
and is not misbranded if it meets each of the conditions in this subpart 
and each general condition established in Sec. 330.1 of this chapter.
    (b) Reference in this subpart to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.

[[Page 233]]



Sec. 333.203  Definitions.

    As used in this subpart:
    (a) Antifungal. A drug which inhibits the growth and reproduction of 
fungal cells and decreases the number of fungi present.
    (b) Athlete's foot. An infection of the feet caused by certain 
dermatophytic fungi.
    (c) Dermatophyte. A fungus that invades and lives upon the skin or 
in the hair or nails.
    (d) Fungus. Any of a large division of plants, including 
dermatophytes, yeasts, and molds, characterized by a simple cell 
structure and the absence of chlorophyll.
    (e) Jock itch. A chronic and recurrent infection caused by certain 
dermatophytic fungi; affects the upper, inner thighs and sometimes 
extends to the groin and the pubic area; the condition most frequently 
occurs in men, but may also occur in women.
    (f) Ringworm. A skin infection caused by certain dermatophytic 
fungi.



Sec. 333.210  Antifungal active ingredients.

    The active ingredient of the product consists of any one of the 
following within the specified concentration established for each 
ingredient:
    (a) Clioquinol 3 percent.
    (b) Haloprogin 1 percent.
    (c) Miconazole nitrate 2 percent.
    (d) Povidone-iodine 10 percent.
    (e) Tolnaftate 1 percent.
    (f) Undecylenic acid, calcium undecylenate, copper undecylenate, and 
zinc undecylenate may be used individually or in any ratio that provides 
a total undecylenate concentration of 10 to 25 percent.



Sec. 333.250  Labeling of antifungal drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as an 
``antifungal.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the phrase listed in paragraph (b)(1)(i) of 
this section and may contain the additional phrase listed in paragraph 
(b)(1)(ii) of this section. Other truthful and nonmisleading statements, 
describing only the indications for use that have been established in 
paragraph (b) of this section, may also be used, as provided in 
Sec. 330.1(c)(2) of this chapter, subject to the provisions of section 
502 of the Federal Food, Drug, and Cosmetic Act (the act) relating to 
misbranding and the prohibition in section 301(d) of the act against the 
introduction or delivery for introduction into interstate commerce of 
unapproved new drugs in violation of section 505(a) of the act.
    (1) For products containing any ingredient identified in 
Sec. 333.210 labeled for the treatment of athlete's foot, jock itch, and 
ringworm. (i) (Select one of the following: ``Treats,'' ``For the 
treatment of,'' ``For effective treatment of,'' ``Cures,'' ``For the 
cure of,'' ``Clears up,'' or ``Proven clinically effective in the 
treatment of'') (select one condition from any one or more of the 
following groups of conditions:
    (A) ``Athlete's foot,'' athlete's foot (dermatophytosis),'' 
``athlete's foot (tinea pedis),'' or ``tinea pedis (athlete's foot)'';
    (B) ``Jock itch,'' ``jock itch (tinea cruris),'' or ``tinea cruris 
(jock itch)''; or
    (C) ``Ringworm,'' ``ringworm (tinea corporis),'' or ``tinea corporis 
(ringworm).'')
    (ii) In addition to the information identified in paragraph 
(b)(1)(i) of this section, the labeling of the product may contain the 
following statement: (Select one of the following: ``Relieves,'' ``For 
relief of,'' ``For effective relief of,'' or ``Soothes,'') (select one 
or more of the following: ``Itching,'' ``scaling,'' ``cracking,'' 
``burning,'' ``redness,'' ``soreness,'' ``irritation,'' ``discomfort,'' 
``chafing associated with jock itch,'' ``itchy, scaly skin between the 
toes,'' or ``itching, burning feet'').
    (2) For products containing the ingredient identified in 
Sec. 333.210(e) labeled for the prevention of athlete's foot. (i) 
(Select one of the following: ``Clinically proven to prevent,'' 
``Prevents,'' ``Proven effective in the prevention of,'' ``Helps 
prevent'', ``For the prevention of,'' ``For the prophylaxis (prevention) 
of,'' ``Guards against,'' or ``Prevents the recurrence of'') (select one 
of the following: ``Athlete's foot,'' ``athlete's foot 
(dermatophytosis),'' ``athlete's foot

[[Page 234]]

(tinea pedis),'' or ``tinea pedis (athlete's foot)'') ``with daily 
use.''
    (ii) In addition to the information identified in paragraph 
(b)(2)(i) of this section, the labeling of the product may contain the 
following statement: ``Clears up athlete's foot infection and with daily 
use helps keep it from coming back.''
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) For products containing any ingredient identified in 
Sec. 330.210. (i) ``Do not use on children under 2 years of age unless 
directed by a doctor.''
    (ii) ``For external use only.''
    (iii) ``Avoid contact with the eyes.''
    (2) For products labeled according to paragraph (b)(1) of this 
section for the treatment of athlete's foot and ringworm. ``If 
irritation occurs or if there is no improvement within 4 weeks, 
discontinue use and consult a doctor.''
    (3) For products labeled according to paragraph (b)(1) of this 
section for the treatment of jock itch. ``If irritation occurs or if 
there is no improvement within 2 weeks, discontinue use and consult a 
doctor.''
    (4) For products labeled according to paragraph (b)(2) of this 
section for the prevention of athlete's foot. ``If irritation occurs, 
discontinue use and consult a doctor.''
    (5) For products containing the ingredient identified in 
Sec. 333.210(a) labeled according to paragraph (b)(1) of this section. 
The following statements must appear in boldface type as the first 
warnings under the ``Warnings'' heading. (i) ``Do not use on children 
under 2 years of age.'' (This warning is to be used in place of the 
warning in paragraph (c)(1)(i) of this section.)
    (ii) ``Do not use for diaper rash.''
    (d) Directions. The labeling of the product contains the following 
statements under the heading ``Directions'':
    (1) For products labeled according to paragraph (b)(1) of this 
section for the treatment of athlete's foot, jock itch, and ringworm. 
[Select one of the following: ``Clean'' or ``Wash''] ``the affected area 
and dry thoroughly. Apply'' (the word ``spray'' may be used to replace 
the word ``apply'' for aerosol products) ``a thin layer of the product 
over affected area twice daily (morning and night) or as directed by a 
doctor. Supervise children in the use of this product. For athlete's 
foot: Pay special attention to spaces between the toes; wear well-
fitting, ventilated shoes, and change shoes and socks at least once 
daily. For athlete's foot and ringworm, use daily for 4 weeks; for jock 
itch, use daily for 2 weeks. If condition persists longer, consult a 
doctor. This product is not effective on the scalp or nails.''
    (2) For products labeled according to paragraph (b)(2) of this 
section for the prevention of athlete's foot. ``To prevent athlete's 
foot,'' (select one of the following: ``clean'' or ``wash'') ``the feet 
and dry thoroughly. Apply'' (the word ``spray'' may be used to replace 
the word ``apply'' for aerosol products) ``a thin layer of the product 
to the feet once or twice daily (morning and/or night). Supervise 
children in the use of this product. Pay special attention to spaces 
between the toes; wear well-fitting, ventilated shoes, and change shoes 
and socks at least once daily.''
    (e) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this section.



Sec. 333.280  Professional labeling.

    The labeling provided to health professionals (but not to the 
general public) may contain the following additional indication:
    (a) For products containing haloprogin or miconazole nitrate 
identified in Sec. 333.210 (a) and (c). ``For the treatment of 
superficial skin infections caused by yeast (Candida albicans).''
    (b) [Reserved]



                  Subpart D--Topical Acne Drug Products

    Source:  56 FR 41019, Aug. 16, 1991, unless otherwise noted.



Sec. 333.301  Scope.

    (a) An over-the-counter acne drug product in a form suitable for 
topical application is generally recognized as safe and effective and is 
not misbranded if it meets each of the conditions in this subpart and 
each general condition established in Sec. 330.1 of this chapter.

[[Page 235]]

    (b) References in this subpart to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 333.303  Definitions.

    As used in this subpart:
    (a) Acne. A disease involving the oil glands and hair follicles of 
the skin which is manifested by blackheads, whiteheads, acne pimples, 
and acne blemishes.
    (b) Acne blemish. A flaw in the skin resulting from acne.
    (c) Acne drug product. A drug product used to reduce the number of 
acne blemishes, acne pimples, blackheads, and whiteheads.
    (d) Acne pimple. A small, prominent, inflamed elevation of the skin 
resulting from acne.
    (e) Blackhead. A condition of the skin that occurs in acne and is 
characterized by a black tip.
    (f) Whitehead. A condition of the skin that occurs in acne and is 
characterized by a small, firm, whitish elevation of the skin.



Sec. 333.310  Acne active ingredients.

    The active ingredient of the product consists of any of the 
following when labeled according to Sec. 333.350.
    (a) Resorcinol 2 percent when combined in accordance with 
Sec. 333.320(a).
    (b) Resorcinol monoacetate 3 percent when combined in accordance 
with Sec. 333.320(b).
    (c) Salicylic acid 0.5 to 2 percent.
    (d) Sulfur 3 to 10 percent.
    (e) Sulfur 3 to 8 percent when combined in accordance with 
Sec. 333.320.



Sec. 333.320  Permitted combinations of active ingredients.

    (a) Resorcinol identified in Sec. 333.310(a) when combined with 
sulfur identified in Sec. 333.310(e) provided the product is labeled 
according to Sec. 333.350.
    (b) Resorcinol monoacetate identified in Sec. 333.310(b) when 
combined with sulfur identified in Sec. 333.310(e) provided the product 
is labeled according to Sec. 333.350.



Sec. 333.350  Labeling of acne drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as an 
``acne medication,'' ``acne treatment,'' ``acne medication'' (insert 
dosage form, e.g., ``cream,'' ``gel,'' ``lotion,'' or ``ointment''), or 
``acne treatment'' (insert dosage form, e.g., ``cream,'' ``gel,'' 
``lotion,'' or ``ointment'').
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the phrase listed in paragraph (b)(1) of this 
section and may contain any of the additional phrases listed in 
paragraph (b)(2) of this section. Other truthful and nonmisleading 
statements, describing only the indications for use that have been 
established and listed in paragraph (b) of this section, may also be 
used, as provided in Sec. 330.1(c)(2) of this chapter, subject to the 
provisions of section 502 of the Federal Food, Drug, and Cosmetic Act 
(the act) relating to misbranding and the prohibition in section 301(d) 
of the act against the introduction or delivery for introduction into 
interstate commerce of unapproved new drugs in violation of section 
505(a) of the act.
    (1) ``For the'' (select one of the following: ``management'' or 
``treatment'') ``of acne.''
    (2) In addition to the information identified in paragraph (b)(1) of 
this section, the labeling of the product may contain any one or more of 
the following statements:
    (i) (Select one of the following: ``Clears,'' ``Clears up,'' 
``Clears up most,'' ``Dries,'' ``Dries up,'' ``Dries and clears,'' 
``Helps clear,'' ``Helps clear up,'' ``Reduces the number of,'' or 
``Reduces the severity of'') (select one or more of the following: 
``acne blemishes,'' ``acne pimples,'' ``blackheads,'' or ``whiteheads'') 
which may be followed by ``and allows skin to heal.''
    (ii) ``Penetrates pores to'' (select one of the following: 
``eliminate most,'' ``control,'' ``clear most,'' or ``reduce the number 
of'') (select one or more of the following: ``acne blemishes,'' ``acne 
pimples,'' ``blackheads,'' or ``whiteheads'').
    (iii) ``Helps keep skin clear of new'' (select one or more of the 
following: ``acne blemishes,'' ``acne pimples,'' ``blackheads,'' or 
``whiteheads'').
    (iv) ``Helps prevent new'' (select one or more of the following: 
``acne blemishes,'' ``acne pimples,'' ``blackheads,''

[[Page 236]]

or ``whiteheads'') which may be followed by ``from forming.''
    (v) ``Helps prevent the development of new'' (select one or more of 
the following: ``acne blemishes,'' ``acne pimples,'' ``blackheads,'' or 
``whiteheads'').
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) For products containing any ingredient identified in 
Sec. 333.310. (i) ``For external use only.''
    (ii) ``Using other topical acne medications at the same time or 
immediately following use of this product may increase dryness or 
irritation of the skin. If this occurs, only one medication should be 
used unless directed by a doctor.''
    (2) For products containing sulfur identified in Secs. 333.310 (d) 
and (e). ``Do not get into eyes. If excessive skin irritation develops 
or increases, discontinue use and consult a doctor.''
    (3) For products containing any combination identified in 
Sec. 333.320. ``Apply to affected areas only. Do not use on broken skin 
or apply to large areas of the body.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) ``Cleanse the skin thoroughly before applying medication. Cover 
the entire affected area with a thin layer one to three times daily. 
Because excessive drying of the skin may occur, start with one 
application daily, then gradually increase to two or three times daily 
if needed or as directed by a doctor. If bothersome dryness or peeling 
occurs, reduce application to once a day or every other day.''
    (2) The directions described in paragraph (d)(1) of this section are 
intended for products that are applied and left on the skin. Other 
products, such as soaps or masks, may be applied and removed and should 
have appropriate directions.
    (3) Optional directions. In addition to the required directions in 
paragraphs (d)(1) and (d)(2) of this section, the product may contain 
the following optional labeling: ``Sensitivity Test for a New User. 
Apply product sparingly to one or two small affected areas during the 
first 3 days. If no discomfort occurs, follow the directions stated: 
(select one of the following: `elsewhere on this label,' `above,' or 
`below.')''
    (e) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this section.



PART 336--ANTIEMETIC DRUG PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                      Subpart A--General Provisions

Sec.
336.1  Scope.
336.3  Definition.

                      Subpart B--Active Ingredients

336.10  Antiemetic active ingredients.

                           Subpart C--Labeling

336.50  Labeling of antiemetic drug products.
336.80  Professional labeling.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  52 FR 15892, Apr. 30, 1987, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 336.1  Scope.

    (a) An over-the-counter antiemetic drug product in a form suitable 
for oral administration is generally recognized as safe and effective 
and is not misbranded if it meets each of the conditions in this part 
and each of the general conditions established in Sec. 330.1.
    (b) References in this part to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 336.3  Definition.

    As used in this part:
    Antiemetic. An agent that prevents or treats nausea and vomiting.



                      Subpart B--Active Ingredients



Sec. 336.10  Antiemetic active ingredients.

    The active ingredient of the product consists of any of the 
following when used within the dosage limits established for each 
ingredient in Sec. 336.50(d):
    (a) Cyclizine hydrochloride.
    (b) Dimenhydrinate.

[[Page 237]]

    (c) Diphenhydramine hydrochloride.
    (d) Meclizine hydrochloride.



                           Subpart C--Labeling



Sec. 336.50  Labeling of antiemetic drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as an 
``antiemetic.''
    (b) Indications. The labeling of the product states the following 
under the heading ``Indications,'' ``For the prevention and treatment of 
the nausea, vomiting, or dizziness associated with motion sickness.'' 
Other truthful and nonmisleading statements, describing only the 
indications for use that have been established and listed in this 
paragraph (b), may also be used, as provided in Sec. 330.1(c)(2), 
subject to the provisions of section 502 of the act relating to 
misbranding and the prohibition in section 301(d) of the act against the 
introduction or delivery for introduction into interstate commerce of 
unapproved new drugs in violation of section 505(a) of the act.
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings:''
    (1) For products containing any ingredient identified in 
Sec. 336.10--(i) When labeled for use in adults and for those products 
that can be and are labeled for use in children under 12 years of age. 
``Do not take this product, unless directed by a doctor, if you have a 
breathing problem such as emphysema or chronic bronchitis, or if you 
have glaucoma or difficulty in urination due to enlargement of the 
prostate gland.''
    (ii) For those products that can be and are labeled only for 
children under 12 years of age. ``Do not give this product to children 
who have a breathing problem such as chronic bronchitis or who have 
glaucoma, without first consulting the child's doctor.''
    (2) For products containing cyclizine hydrochloride identified in 
Sec. 336.10(a). ``Do not give to children under 6 years of age unless 
directed by a doctor.''
    (3) For products containing dimenhydrinate identified in 
Sec. 336.10(b). ``Do not give to children under 2 years of age unless 
directed by a doctor.''
    (4) For products containing diphenhydramine hydrochloride identified 
in Sec. 336.10(c). ``Do not give to children under 6 years of age unless 
directed by a doctor.''
    (5) For products containing meclizine hydrochloride identified in 
Sec. 336.10(d). ``Do not give to children under 12 years of age unless 
directed by a doctor.''
    (6) For products containing cyclizine hydrochloride identified in 
Sec. 336.10(a) or meclizine hydrochloride identified in Sec. 330.10(d). 
``May cause drowsiness; alcohol, sedatives, and tranquilizers may 
increase the drowsiness effect. Avoid alcoholic beverages while taking 
this product. Do not take this product if you are taking sedatives or 
tranquilizers, without first consulting your doctor. Use caution when 
driving a motor vehicle or operating machinery.''
    (7) For products containing dimenhydrinate identified in 
Sec. 336.10(b) or diphenhydramine hydrochloride identified in 
Sec. 336.10(c). ``May cause marked drowsiness; alcohol, sedatives, and 
tranquilizers may increase the drowsiness effect. Avoid alcoholic 
beverages while taking this product. Do not take this product if you are 
taking sedatives or tranquilizers, without first consulting your doctor. 
Use caution when driving a motor vehicle or operating machinery.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) For products containing cyclizine hydrochloride identified in 
Sec. 336.10(a). Adults and children 12 years of age and over: Oral 
dosage is 50 milligrams every 4 to 6 hours, not to exceed 200 milligrams 
in 24 hours, or as directed by a doctor. Children 6 to under 12 years of 
age: Oral dosage is 25 milligrams every 6 to 8 hours, not to exceed 75 
milligrams in 24 hours, or as directed by a doctor.
    (2) For products containing dimenhydrinate identified in 
Sec. 336.10(b). Adults and children 12 years of age and over: Oral 
dosage is 50 to 100 milligrams every 4 to 6 hours, not to exceed 400 
milligrams in 24 hours, or as directed by a doctor. Children 6 to under 
12

[[Page 238]]

years of age: Oral dosage is 25 to 50 milligrams every 6 to 8 hours, not 
to exceed 150 milligrams in 24 hours, or as directed by a doctor. 
Children 2 to under 6 years of age: Oral dosage is 12.5 to 25 milligrams 
every 6 to 8 hours, not to exceed 75 milligrams in 24 hours, or as 
directed by a doctor.
    (3) For products containing diphenhydramine hydrochloride identified 
in Sec. 336.10(c). Adults and children 12 years of age and over: Oral 
dosage is 25 to 50 milligrams every 4 to 6 hours, not to exceed 300 
milligrams in 24 hours, or as directed by a doctor. Children 6 to under 
12 years of age: Oral dosage is 12.5 to 25 milligrams every 4 to 6 
hours, not to exceed 150 milligrams in 24 hours, or as directed by a 
doctor.
    (4) For products containing meclizine hydrochloride identified in 
Sec. 336.10(d). Adults and children 12 years of age and over: Oral 
dosage is 25 to 50 milligrams once daily, or as directed by a doctor.
    (e) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this section.
[52 FR 15892, Apr. 30, 1987, as amended at 53 FR 35809, Sept. 15, 1988; 
59 FR 16982, Apr. 11, 1994]



Sec. 336.80  Professional labeling.

    The labeling provided to health professionals (but not to the 
general public) may contain the following additional indications.
    (a) For products containing cyclizine hydrochloride, dimenhydrinate, 
and diphenhydramine hydrochloride identified in Sec. 336.10 (a), (b), 
and (c). ``For the treatment of vertigo of motion sickness.''
    (b) For products containing meclizine hydrochloride identified in 
Sec. 336.10(d). ``For the treatment of vertigo.''



PART 338--NIGHTTIME SLEEP-AID DRUG PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                      Subpart A--General Provisions

Sec.
338.1  Scope.
338.3  Definition.

                      Subpart B--Active Ingredients

338.10  Nighttime sleep-aid active ingredients.

                           Subpart C--Labeling

338.50  Labeling of nighttime sleep-aid drug products.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  54 FR 6826, Feb. 14, 1989, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 338.1  Scope.

    (a) An over-the-counter nighttime sleep-aid drug product in a form 
suitable for oral administration is generally recognized as safe and 
effective and is not misbranded if it meets each condition in this part 
and each general condition established in Sec. 330.1 of this chapter.
    (b) References in this part to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 338.3  Definition.

    As used in this part:
    Nighttime sleep-aid. A drug that is useful for the relief of 
occasional sleeplessness by individuals who have difficulty falling 
asleep.



                      Subpart B--Active Ingredients



Sec. 338.10  Nighttime sleep-aid active ingredients.

    The active ingredient of the product consists of any of the 
following when used within the dosage limits established for each 
ingredient in Sec. 338.50(d):
    (a) Diphenhydramine hydrochloride.
    (b) Diphenhydramine citrate.



                           Subpart C--Labeling



Sec. 338.50  Labeling of nighttime sleep-aid drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as a 
``nighttime sleep-aid.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' one or more of the phrases listed in this 
paragraph. Other truthful and nonmisleading statements, describing only 
the indications for use

[[Page 239]]

that have been established and listed in this paragraph (b), may also be 
used, as provided in Sec. 330.1(c)(2) of this chapter, subject to the 
provisions of section 502 of the act relating to misbranding and the 
prohibition in section 301(d) of the act against the introduction or 
delivery for introduction into interstate commerce of unapproved new 
drugs in violation of section 505(a) of the act.
    (1) (``Helps you'' or ``Reduces time to'') ``fall asleep if you have 
difficulty falling asleep.''
    (2) ``For relief of occasional sleeplessness.''
    (3) ``Helps to reduce difficulty falling asleep.''
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) ``Do not give to children under 12 years of age.''
    (2) ``If sleeplessness persists continuously for more than 2 weeks, 
consult your doctor. Insomnia may be a symptom of serious underlying 
medical illness.''
    (3) ``Do not take this product, unless directed by a doctor, if you 
have a breathing problem such as emphysema or chronic bronchitis, or if 
you have glaucoma or difficulty in urination due to enlargement of the 
prostate gland.''
    (4) ``Avoid alcoholic beverages while taking this product. Do not 
take this product if you are taking sedatives or tranquilizers, without 
first consulting your doctor.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) For products containing diphenhydramine hydrochloride identified 
in Sec. 338.10(a). Adults and children 12 years of age and over: Oral 
dosage is 50 milligrams at bedtime if needed, or as directed by a 
doctor.
    (2) For products containing diphenhydramine citrate identified in 
Sec. 338.10(b). Adults and children 12 years of age and over: Oral 
dosage is 76 milligrams at bedtime if needed, or as directed by a 
doctor.
    (e) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this section.
[54 FR 6826, Feb. 14, 1989, as amended at 59 FR 16983, Apr. 11, 1994]



PART 340--STIMULANT DRUG PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                      Subpart A--General Provisions

Sec.
340.1  Scope.
340.3  Definition.

                      Subpart B--Active Ingredient

340.10  Stimulant active ingredient.

                           Subpart C--Labeling

340.50  Labeling of stimulant drug products.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  53 FR 6105, Feb. 29, 1988, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 340.1  Scope.

    (a) An over-the-counter stimulant drug product in a form suitable 
for oral administration is generally recognized as safe and effective 
and is not misbranded if it meets each of the conditions in this part 
and each of the general conditions established in Sec. 330.1.
    (b) References in this part to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 340.3  Definition.

    As used in this part:
    Stimulant. A drug which helps restore mental alertness or 
wakefulness during fatigue or drowsiness.



                      Subpart B--Active Ingredient



Sec. 340.10  Stimulant active ingredient.

    The active ingredient of the product consists of caffeine when used 
within the dosage limits established in Sec. 340.50(d).

[[Page 240]]



                           Subpart C--Labeling



Sec. 340.50  Labeling of stimulant drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as an 
``altertness aid'' or a ``stimulant.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the following: ``Helps restore mental alertness 
or wakefulness when experiencing fatigue or drowsiness.'' Other truthful 
and nonmisleading statements, describing only the indications for use 
that have been established and listed in this paragraph (b), may also be 
used, as provided in Sec. 330.1(c)(2), subject to the provisions of 
section 502 of the Act relating to misbranding and the prohibition in 
section 301(d) of the Act against the introduction or delivery for 
introduction into interstate commerce of unapproved new drugs in 
violation of section 505(a) of the Act.
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) ``The recommended dose of this product contains about as much 
caffeine as a cup of coffee. Limit the use of caffeine-containing 
medications, foods, or beverages while taking this product because too 
much caffeine may cause nervousness, irritability, sleeplessness, and, 
occasionally, rapid heart beat.''
    (2) ``For occasional use only. Not intended for use as a substitute 
for sleep. If fatigue or drowsiness persists or continues to recur, 
consult a'' (select one of the following: ``physician'' or ``doctor'').
    (3) ``Do not give to children under 12 years of age.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'': Adults and children 12 
years of age and over: Oral dosage is 100 to 200 milligrams not more 
often than every 3 to 4 hours.



PART 341--COLD, COUGH, ALLERGY, BRONCHODILATOR, AND ANTIASTHMATIC DRUG PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                      Subpart A--General Provisions

Sec.
341.1  Scope.
341.3  Definitions.

                      Subpart B--Active Ingredients

341.12  Antihistamine active ingredients.
341.14  Antitussive active ingredients.
341.16  Bronchodilator active ingredients.
341.18  Expectorant active ingredient.
341.20  Nasal decongestant active ingredients.

                           Subpart C--Labeling

341.70  Labeling of OTC drug products containing ingredients that are 
          used for treating concurrent symptoms (in either a single-
          ingredient or combination drug product).
341.72  Labeling of antihistamine drug products.
341.74  Labeling of antitussive drug products.
341.76  Labeling of bronchodilator drug products.
341.78  Labeling of expectorant drug products.
341.80  Labeling of nasal decongestant drug products.
341.90  Professional labeling.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.



                      Subpart A--General Provisions



Sec. 341.1  Scope.

    (a) An over-the-counter cold, cough, allergy, bronchodilator, or 
antiasthmatic drug product in a form suitable for oral, inhalant, or 
topical administration is generally recognized as safe and effective and 
is not misbranded if it meets each of the conditions in this part and 
each of the general conditions established in Sec. 330.1.
    (b) References in this part to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.
[51 FR 35339, Oct. 2, 1986]

[[Page 241]]



Sec. 341.3  Definitions.

    As used in this part:
    (a) Bronchodilator drug. A drug used to overcome spasms that cause 
narrowing of the bronchial air tubes, such as in the symptomatic 
treatment of the wheezing and shortness of breath of asthma.
    (b) Oral antitussive drug. A drug that either is taken by mouth or 
is dissolved in the mouth in the form of a lozenge and acts systemically 
to relieve cough.
    (c) Topical antitussive drug. A drug that relieves cough when 
inhaled after being applied topically to the throat or chest in the form 
of an ointment or from a steam vaporizer, or when dissolved in the mouth 
in the form of a lozenge for a local effect.
    (d) Expectorant drug. A drug taken orally to promote or facilitate 
the removal of secretions from the respiratory airways.
    (e) Antihistamine drug. A drug used for the relief of the symptoms 
of hay fever and upper respiratory allergies (allergic rhinitis).
    (f) Oral nasal decongestant drug. A drug that is taken by mouth and 
acts systemically to reduce nasal congestion caused by acute or chronic 
rhinitis.
    (g) Topical nasal decongestant drug. A drug that when applied 
topically inside the nose, in the form of drops, jellies, or sprays, or 
when inhaled intranasally reduces nasal congestion caused by acute or 
chronic rhinitis.
    (h) Calibrated dropper. A dropper calibrated such that the volume 
error incurred in measuring any liquid does not exceed 15 percent under 
normal use conditions.
[51 FR 35339, Oct. 2, 1986, as amended at 54 FR 8509, Feb. 28, 1989; 55 
FR 40382, Oct. 3, 1990; 57 FR 58374, Dec. 9, 1992; 59 FR 43409, Aug. 23, 
1994]



                      Subpart B--Active Ingredients



Sec. 341.12  Antihistamine active ingredients.

    The active ingredient of the product consists of any of the 
following when used within the dosage limits established for each 
ingredient:
    (a) Brompheniramine maleate.
    (b) Chlorcyclizine hydrochloride.
    (c) Chlorpheniramine maleate.
    (d) Dexbrompheniramine maleate.
    (e) Dexchlorpheniramine maleate.
    (f) Diphenhydramine citrate.
    (g) Diphenhydramine hydrochloride.
    (h) Doxylamine succinate.
    (i) Phenindamine tartrate.
    (j) Pheniramine maleate.
    (k) Pyrilamine maleate.
    (l) Thonzylamine hydrochloride.
    (m) Triprolidine hydrochloride.
[57 FR 58374, Dec. 9, 1992, as amended at 59 FR 4218, Jan. 28, 1994]



Sec. 341.14  Antitussive active ingredients.

    The active ingredients of the product consist of any of the 
following when used within the dosage limits and in the dosage forms 
established for each ingredient in Sec. 341.74(d):
    (a) Oral antitussives. (1) Chlophedianol hydrochloride.
    (2) Codeine ingredients. The following ingredients may be used only 
in combination in accordance with Secs. 329.20(a) and 341.40 and 21 CFR 
1308.15(c).
    (i) Codeine.
    (ii) Codeine phosphate.
    (iii) Codeine sulfate.
    (3) Dextromethorphan.
    (4) Dextromethorphan hydrobromide.
    (5) Diphenhydramine citrate.
    (6) Diphenhydramine hydrochloride.
    (b) Topical antitussives.
    (1) Camphor.
    (2) Menthol.
[52 FR 30055, Aug. 12, 1987, as amended at 59 FR 29174, June 3, 1994]



Sec. 341.16  Bronchodilator active ingredients.

    The active ingredients of the product consist of any of the 
following when used within the dosage limits established for each 
ingredient:
    (a) Ephedrine.
    (b) Ephedrine hydrochloride.
    (c) Ephedrine sulfate.
    (d) Epinephrine.
    (e) Epinephrine bitartrate.
    (f) Racephedrine hydrochloride.
    (g) Racepinephrine hydrochloride.
[51 FR 35339, Oct. 2, 1986]

[[Page 242]]



Sec. 341.18  Expectorant active ingredient.

    The active ingredient of the product is guaifenesin when used within 
the dosage limits established in Sec. 341.78(d).
[54 FR 8509, Feb. 28, 1989]



Sec. 341.20  Nasal decongestant active ingredients.

    The active ingredient of the product consists of any of the 
following when used within the dosage limits and in the dosage forms 
established for each ingredient:
    (a) Oral nasal decongestants. (1) Phenylephrine hydrochloride.
    (2) Pseudoephedrine hydrochloride.
    (3) Pseudoephedrine sulfate.
    (b) Topical nasal decongestants. (1) [Reserved]
    (2) Ephedrine.
    (3) Ephedrine hydrochloride.
    (4) Ephedrine sulfate.
    (5) [Reserved]
    (6) Naphazoline hydrochloride.
    (7) Oxymetazoline hydrochloride.
    (8) Phenylephrine hydrochloride.
    (9) Propylhexedrine.
    (10) Xylometazoline hydrochloride.
[59 FR 43409, Aug. 23, 1994]



                           Subpart C--Labeling



Sec. 341.70  Labeling of OTC drug products containing ingredients that are used for treating concurrent symptoms (in either a single-ingredient or combination 
          drug product).

    The statements of identity, indications, warnings, and directions 
for use, respectively, applicable to each ingredient in the product may 
be combined to eliminate duplicative words or phrases so that the 
resulting information is clear and understandable.
    (a) For products containing diphenhydramine citrate and 
diphenhydramine hydrochloride identified in Sec. 341.14(a)(5) and 
(a)(6). The labeling of the product contains the established name of the 
drug, if any, and identifies the product as an ``antihistamine/cough 
suppressant'' or ``antihistamine/antitussive (cough suppressant).'' The 
indications shall be combined from Secs. 341.72(b) and 341.74(b). The 
warnings shall be combined from Secs. 341.72(c)(1), (c)(2), (c)(4), and 
(c)(6) and 341.74(c)(1), (c)(2), (c)(3), and (c)(4). Alternatively, all 
of the warnings in Sec. 341.74(c) shall be used. The directions for OTC 
labeling shall follow Secs. 341.74(d)(1)(iv) or (d)(1)(v), as 
applicable. The directions for professional labeling shall follow 
Sec. 341.90(j) or (k), as applicable.
    (b) (Reserved)
[61 FR 15703, Apr. 9, 1996]



Sec. 341.72  Labeling of antihistamine drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as an 
``antihistamine.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' any of the phrases listed in paragraph (b) of 
this section, as appropriate. Other truthful and nonmisleading 
statements, describing only the indications for use that have been 
established and listed in this paragraph, may also be used, as provided 
in Sec. 330.1(c)(2) of this chapter, subject to the provisions of 
section 502 of the Federal Food, Drug, and Cosmetic Act (the act) 
relating to misbranding and the prohibition in section 301(d) of the act 
against the introduction or delivery for introduction into interstate 
commerce of unapproved new drugs in violation of section 505(a) of the 
act.
    (1) ``Temporarily'' (select one of the following: ``relieves,'' 
``alleviates,'' ``decreases,'' ``reduces,'' or ``dries'') ``runny nose 
and'' (select one of the following: ``relieves,'' ``alleviates,'' 
``decreases,'' or ``reduces'') ``sneezing, itching of the nose or 
throat, and itchy, watery eyes due to hay fever'' (which may be followed 
by one or both of the following: ``or other upper respiratory 
allergies'' or ``(allergic rhinitis)'').
    (2) ``For the temporary relief of runny nose, sneezing, itching of 
the nose or throat, and itchy, watery eyes due to hay fever'' (which may 
be followed by one or both of the following: ``or other upper 
respiratory allergies'' or ``(allergic rhinitis)'').
    (c) Warnings. The labeling of the product contains the following 
warnings, under the heading ``Warnings'':
    (1) ``May cause excitability especially in children.''
    (2) ``Do not take this product, unless directed by a doctor, if you 
have a

[[Page 243]]

breathing problem such as emphysema or chronic bronchitis, or if you 
have glaucoma or difficulty in urination due to enlargement of the 
prostate gland.''
    (3) For products containing brompheniramine maleate, chlorcyclizine 
hydrochloride, chlorpheniramine maleate, dexbrompheniramine maleate, 
dexchlorpheniramine maleate, phenindamine tartrate, pheniramine maleate, 
pyrilamine maleate, thonzylamine hydrochloride, or triprolidine 
hydrochloride identified in Sec. 341.12(a), (b), (c), (d), (e), (i), 
(j), (k), (l), and (m). ``May cause drowsiness; alcohol, sedatives, and 
tranquilizers may increase the drowsiness effect. Avoid alcoholic 
beverages while taking this product. Do not take this product if you are 
taking sedatives or tranquilizers, without first consulting your doctor. 
Use caution when driving a motor vehicle or operating machinery.''
    (4) For products containing diphenhydramine citrate, diphenhydramine 
hydrochloride, or doxylamine succinate identified in Sec. 341.12(f), 
(g), and (h). ``May cause marked drowsiness; alcohol, sedatives, and 
tranquilizers may increase the drowsiness effect. Avoid alcoholic 
beverages while taking this product. Do not take this product if you are 
taking sedatives or tranquilizers, without first consulting your doctor. 
Use caution when driving a motor vehicle or operating machinery.''
    (5) For products containing phenindamine tartrate identified in 
Sec. 341.12(i). ``May cause nervousness and insomnia in some 
individuals.''
    (6) For products that are labeled only for use by children under 12 
years of age. The labeling of the product contains only the warnings 
identified in paragraphs (c)(1) and (c)(5) of this section as well as 
the following:
    (i) ``Do not give this product to children who have a breathing 
problem such as chronic bronchitis, or who have glaucoma, without first 
consulting the child's doctor.''
    (ii) For products containing brompheniramine maleate, 
chlorpheniramine maleate, dexbrompheniramine maleate, 
dexchlorpheniramine maleate, phenindamine tartrate, pheniramine maleate, 
pyrilamine maleate, thonzylamine hydrochloride, or triprolidine 
hydrochloride identified in Sec. 341.12(a), (c), (d), (e), (i), (j), 
(k), (l), and (m). ``May cause drowsiness. Sedatives and tranquilizers 
may increase the drowsiness effect. Do not give this product to children 
who are taking sedatives or tranquilizers, without first consulting the 
child's doctor.''
    (iii) For products containing diphenhydramine citrate, 
diphenhydramine hydrochloride, or doxylamine succinate identified in 
Sec. 341.12(f), (g), and (h). ``May cause marked drowsiness. Sedatives 
and tranquilizers may increase the drowsiness effect. Do not give this 
product to children who are taking sedatives or tranquilizers, without 
first consulting the child's doctor.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) For products containing brompheniramine maleate identified in 
Sec. 341.12(a). Adults and children 12 years of age and over: oral 
dosage is 4 milligrams every 4 to 6 hours, not to exceed 24 milligrams 
in 24 hours, or as directed by a doctor. Children 6 to under 12 years of 
age: oral dosage is 2 milligrams every 4 to 6 hours, not to exceed 12 
milligrams in 24 hours, or as directed by a doctor. Children under 6 
years of age: consult a doctor.
    (2) For products containing chlorcyclizine hydrochloride identified 
in Sec. 341.12(b). Adults and children 12 years of age and over: oral 
dosage is 25 milligrams every 6 to 8 hours, not to exceed 75 milligrams 
in 24 hours, or as directed by a doctor. Children under 12 years of age: 
consult a doctor.
    (3) For products containing chlorpheniramine maleate identified in 
Sec. 341.12(c). Adults and children 12 years of age and over: oral 
dosage is 4 milligrams every 4 to 6 hours, not to exceed 24 milligrams 
in 24 hours, or as directed by a doctor. Children 6 to under 12 years of 
age: oral dosage is 2 milligrams every 4 to 6 hours, not to exceed 12 
milligrams in 24 hours, or as directed by a doctor. Children under 6 
years of age: consult a doctor.
    (4) For products containing dexbrompheniramine maleate identified in 
Sec. 341.12(d). Adults and children 12 years

[[Page 244]]

of age and over: oral dosage is 2 milligrams every 4 to 6 hours, not to 
exceed 12 milligrams in 24 hours, or as directed by a doctor. Children 6 
to under 12 years of age: oral dosage is 1 milligram every 4 to 6 hours, 
not to exceed 6 milligrams in 24 hours, or as directed by a doctor. 
Children under 6 years of age: consult a doctor.
    (5) For products containing dexchlorpheniramine maleate identified 
in Sec. 341.12(e). Adults and children 12 years of age and over: oral 
dosage is 2 milligrams every 4 to 6 hours, not to exceed 12 milligrams 
in 24 hours, or as directed by a doctor. Children 6 to under 12 years of 
age: oral dosage is 1 milligram every 4 to 6 hours, not to exceed 6 
milligrams in 24 hours, or as directed by a doctor. Children under 6 
years of age: consult a doctor.
    (6) For products containing diphenhydramine citrate identified in 
Sec. 341.12(f). Adults and children 12 years of age and over: oral 
dosage is 38 to 76 milligrams every 4 to 6 hours, not to exceed 456 
milligrams in 24 hours, or as directed by a doctor. Children 6 to under 
12 years of age: oral dosage is 19 to 38 milligrams every 4 to 6 hours, 
not to exceed 228 milligrams in 24 hours, or as directed by a doctor. 
Children under 6 years of age: consult a doctor.
    (7) For products containing diphenhydramine hydrochloride identified 
in Sec. 341.12(g). Adults and children 12 years of age and over: oral 
dosage is 25 to 50 milligrams every 4 to 6 hours, not to exceed 300 
milligrams in 24 hours, or as directed by a doctor. Children 6 to under 
12 years of age: oral dosage is 12.5 to 25 milligrams every 4 to 6 
hours, not to exceed 150 milligrams in 24 hours, or as directed by a 
doctor. Children under 6 years of age: consult a doctor.
    (8) For products containing doxylamine succinate identified in 
Sec. 341.12(h). Adults and children 12 years of age and over: oral 
dosage is 7.5 to 12.5 milligrams every 4 to 6 hours, not to exceed 75 
milligrams in 24 hours, or as directed by a doctor. Children 6 to under 
12 years of age: oral dosage is 3.75 to 6.25 milligrams every 4 to 6 
hours, not to exceed 37.5 milligrams in 24 hours, or as directed by a 
doctor. Children under 6 years of age: consult a doctor.
    (9) For products containing phenindamine tartrate identified in 
Sec. 341.12(i). Adults and children 12 years of age and over: oral 
dosage is 25 milligrams every 4 to 6 hours, not to exceed 150 milligrams 
in 24 hours, or as directed by a doctor. Children 6 to under 12 years of 
age: oral dosage is 12.5 milligrams every 4 to 6 hours, not to exceed 75 
milligrams in 24 hours, or as directed by a doctor. Children under 6 
years of age: consult a doctor.
    (10) For products containing pheniramine maleate identified in 
Sec. 341.12(j). Adults and children 12 years of age and over: oral 
dosage is 12.5 to 25 milligrams every 4 to 6 hours, not to exceed 150 
milligrams in 24 hours, or as directed by a doctor. Children 6 to under 
12 years of age: oral dosage is 6.25 to 12.5 milligrams every 4 to 6 
hours, not to exceed 75 milligrams in 24 hours, or as directed by a 
doctor. Children under 6 years of age: consult a doctor.
    (11) For products containing pyrilamine maleate identified in 
Sec. 341.12(k). Adults and children 12 years of age and over: oral 
dosage is 25 to 50 milligrams every 6 to 8 hours, not to exceed 200 
milligrams in 24 hours, or as directed by a doctor. Children 6 to under 
12 years of age: oral dosage is 12.5 to 25 milligrams every 6 to 8 
hours, not to exceed 100 milligrams in 24 hours, or as directed by a 
doctor. Children under 6 years of age: consult a doctor.
    (12) For products containing thonzylamine hydrochloride identified 
in Sec. 341.12(l). Adults and children 12 years of age and over: oral 
dosage is 50 to 100 milligrams every 4 to 6 hours, not to exceed 600 
milligrams in 24 hours, or as directed by a doctor. Children 6 to under 
12 years of age: oral dosage is 25 to 50 milligrams every 4 to 6 hours, 
not to exceed 300 milligrams in 24 hours, or as directed by a doctor. 
Children under 6 years of age: consult a doctor.
    (13) For products containing triprolidine hydrochloride identified 
in Sec. 341.12(m). Adults and children 12 years of age and over: oral 
dosage is 2.5 milligrams every 4 to 6 hours, not to exceed 10 milligrams 
in 24 hours, or as directed by a doctor. Children 6 to under 12 years of 
age: oral dosage is 1.25 milligrams every 4 to 6 hours, not to exceed 5 
milligrams in 24 hours, or as directed by a

[[Page 245]]

doctor. Children under 6 years of age: consult a doctor.
    (e) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this section.
[57 FR 58374, Dec. 9, 1992, as amended at 59 FR 4218, Jan. 28, 1994]



Sec. 341.74  Labeling of antitussive drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as a 
``cough suppressant'' or an ``antitussive (cough suppressant).''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' any of the phrases listed in this paragraph 
(b), as appropriate. Other truthful and nonmisleading statements, 
describing only the indications for use that have been established and 
listed in this paragraph, may also be used, as provided in 
Sec. 330.1(c)(2), subject to the provisions of section 502 of the act 
relating to misbranding and the prohibition in section 301(d) of the act 
against the introduction or delivery for introduction into interstate 
commerce of unapproved new drugs in violation of section 505(a) of the 
act.
    (1) ``Temporarily'' (select one of the following: ``alleviates,'' 
``calms,'' ``controls,'' ``decreases,'' ``quiets,'' ``reduces,'' 
``relieves,'' or ``suppresses'') ``cough due to'' (select one of the 
following: ``minor bronchial irritation'' or ``minor throat and 
bronchial irritation'') (select one of the following: ``as may occur 
with,'' ``associated with,'' or ``occurring with'') (select one of the 
following: ``A cold'' or ``the common cold'') ``or inhaled irritants.''
    (2) ``Temporarily'' (select one of the following: ``alleviates,'' 
``calms,'' ``controls,'' ``decreases,'' ``quiets,'' ``reduces,'' 
``relieves,'' or ``suppresses'') ``cough'' (select one of the following: 
``as may occur with,'' ``associated with,'' or ``occurring with'') 
(select one of the following: ``A cold,'' ``the common cold,'' or 
``inhaled irritants'').
    (3) In addition to the required information identified in paragraphs 
(b) (1) and (2) of this section, the labeling of the product may contain 
any (one or more) of the following statements:
    (i) ``Cough suppressant which temporarily'' (select one of the 
following: ``Alleviates,'' ``controls,'' ``decreases,'' ``reduces,'' 
``relieves,'' or ``suppresses'') ``the impulse to cough.''
    (ii) ``Temporarily helps you cough less.''
    (iii) ``Temporarily helps to'' (select one of the following: 
``Alleviate,'' ``control,'' ``decrease,'' ``reduce,'' ``relieve,'' or 
``suppress'') ``the cough reflex that causes coughing.''
    (iv) ``Temporarily'' (select one of the following: ``Alleviates,'' 
``controls,'' ``decreases,'' ``reduces,'' ``relieves,'' or 
``suppresses'') ``the intensity of coughing.''
    (v) (Select one of the following: ``Alleviates,'' ``Controls,'' 
``Decreases,'' ``Reduces,'' ``Relieves,'' or ``Suppresses'') (select one 
of the following: ``Cough,'' ``the impulse to cough,'' or ``your 
cough'') ``to help you'' (select one of the following: ``Get to sleep,'' 
``sleep,'' or ``rest'').
    (vi) For products containing chlophedianol hydrochloride, codeine 
ingredients, dextromethorphan, or dextromethorphan hydrobromide 
identified in Sec. 341.14(a) (1), (2), (3), and (4). ``Calms the cough 
control center and relieves coughing.''
    (vii) For products containing chlophedianol hydrochloride, 
dextromethorphan, dextromethorphan hydrobromide, camphor, or menthol 
identified in Sec. 341.14(a) (1), (3), (4) and (b) (1) and (2). (a) 
``Nonnarcotic cough suppressant for the temporary'' (select one of the 
following: ``alleviation,'' ``control,'' ``decrease,'' ``reduction,'' 
``relief,'' or ``suppression'') ``of cough.''
    (b) (Select one of the following: ``Alleviates,'' ``Controls,'' 
``Decreases,'' ``Reduces,'' ``Relieves,'' or ``Suppresses'') ``cough 
impulses without narcotics.''
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) For oral and topical antitussives. ``A persistent cough may be a 
sign of a serious condition. If cough persists for more than 1 week, 
tends to recur, or is accompanied by fever, rash, or persistent 
headache, consult a doctor.''
    (2) For oral and topical antitussives labeled for adults or for 
adults and children under 12 years of age. ``Do not take this

[[Page 246]]

product for persistent or chronic cough such as occurs with smoking, 
asthma, or emphysema, or if cough is accompanied by excessive phlegm 
(mucus) unless directed by a doctor.''
    (3) For oral and topical antitussives labeled only for children 
under 12 years of age. ``Do not give this product for persistent or 
chronic cough such as occurs with asthma or if cough is accompanied by 
excessive phlegm (mucus) unless directed by a doctor.''
    (4) Oral antitussives--(i) For products containing codeine 
ingredients identified in Sec. 341.14(a)(2). ``May cause or aggravate 
constipation.''
    (ii) For products containing codeine ingredients identified in 
Sec. 341.14(a)(2) when labeled only for adults. ``Do not take this 
product if you have a chronic pulmonary disease or shortness of breath 
unless directed by a doctor.''
    (iii) For products containing codeine ingredients identified in 
Sec. 341.14(a)(2) when labeled only for children under 12 years of age. 
``Do not give this product to children who have a chronic pulmonary 
disease, shortness of breath, or who are taking other drugs unless 
directed by a doctor.''
    (iv) For products containing codeine ingredients identified in 
Sec. 341.14(a)(2) when labeled for use in adults and children under 12 
years of age. ``Adults and children who have a chronic pulmonary disease 
or shortness of breath, or children who are taking other drugs, should 
not take this product unless directed by a doctor.''
    (v) For products containing dextromethorphan or dextromethorphan 
hydrobromide as identified in Sec. 341.14(a)(3) and (a)(4) when labeled 
for adults or for adults and children under 12 years of age. ``Drug 
interaction precaution. Do not use this product if you are now taking a 
prescription monoamine oxidase inhibitor (MAOI) (certain drugs for 
depression, psychiatric or emotional conditions, or Parkinson's 
disease), or for 2 weeks after stopping the MAOI drug. If you are 
uncertain whether your prescription drug contains an MAOI, consult a 
health professional before taking this product.''
    (vi) For products containing dextromethorphan or dextromethorphan 
hydrobromide as identified in Sec. 341.14(a)(3) and (a)(4) when labeled 
only for children under 12 years of age. ``Drug interaction precaution. 
Do not give this product to a child who is taking a prescription 
monoamine oxidase inhibitor (MAOI) (certain drugs for depression, 
psychiatric or emotional conditions), or for 2 weeks after stopping the 
MAOI drug. If you are uncertain whether your child's prescription drug 
contains an MAOI, consult a health professional before giving this 
product.''
    (vii) For products containing diphenhydramine citrate or 
diphenhydramine hydrochloride identified in Sec. 341.14(a)(5) and 
(a)(6). ``May cause excitability especially in children.''
    (viii) For products containing diphenhydramine citrate or 
diphenhydramine hydrochloride identified in Sec. 341.14(a)(5) and (a)(6) 
when labeled only for children under 12 years of age--(A) ``Do not give 
this product to children who have a breathing problem such as chronic 
bronchitis, or who have glaucoma, without first consulting the child's 
doctor.''
    (B) ``May cause marked drowsiness. Sedatives and tranquilizers may 
increase the drowsiness effect. Do not give this product to children who 
are taking sedatives or tranquilizers, without first consulting the 
child's doctor.''
    (ix) For products containing diphenhydramine citrate or 
diphenhydramine hydrochloride identified in Sec. 341.14(a)(5) and (a)(6) 
when labeled for use in adults and children under 12 years of age--(A) 
``Do not take this product, unless directed by a doctor, if you have a 
breathing problem such as emphysema or chronic bronchitis, or if you 
have glaucoma or difficulty in urination due to enlargement of the 
prostate gland.''
    (B) ``May cause marked drowsiness; alcohol, sedatives, and 
tranquilizers may increase the drowsiness effect. Avoid alcoholic 
beverages while taking this product. Do not take this product if you are 
taking sedatives or tranquilizers, without first consulting your doctor. 
Use caution when driving a motor vehicle or operating machinery.''

[[Page 247]]

    (5) Topical antitussives--(i) For products containing camphor or 
menthol identified in Sec. 341.14(b) (1) and (2) in a suitable ointment 
vehicle. ``For external use only. Do not take by mouth or place in 
nostrils.''
    (ii) For products containing camphor or menthol identified in 
Sec. 341.14(b) (1) and (2) for steam inhalation use. ``For steam 
inhalation only. Do not take by mouth.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) Oral antitussives--(i) For products containing chlophedianol 
hydrochloride identified in Sec. 341.14(a)(1). Adults and children 12 
years of age and over: Oral dosage is 25 milligrams every 6 to 8 hours, 
not to exceed 100 milligrams in 24 hours, or as directed by a doctor. 
Children 6 to under 12 years of age: Oral dosage is 12.5 milligrams 
every 6 to 8 hours, not to exceed 50 milligrams in 24 hours, or as 
directed by a doctor. Children under 6 years of age: Consult a doctor.
    (ii) For products containing codeine ingredients identified in 
Sec. 341.14(a)(2). Adults and children 12 years of age and over: Oral 
dosage is 10 to 20 milligrams every 4 to 6 hours, not to exceed 120 
milligrams in 24 hours, or as directed by a doctor. Children 6 to under 
12 years of age: Oral dosage is 5 to 10 milligrams every 4 to 6 hours, 
not to exceed 60 milligrams in 24 hours, or as directed by a doctor. 
Children under 6 years of age: Consult a doctor. A special measuring 
device should be used to give an accurate dose of this product to 
children under 6 years of age. Giving a higher dose than recommended by 
a doctor could result in serious side effects for your child.
    (iii) For products containing dextromethorphan or dextromethorphan 
hydrobromide identified in Sec. 341.14(a) (3) and (4). The dosage is 
equivalent to dextromethorphan hydrobromide. Adults and children 12 
years of age and over: Oral dosage is 10 to 20 milligrams every 4 hours 
or 30 milligrams every 6 to 8 hours, not to exceed 120 milligrams in 24 
hours, or as directed by a doctor. Children 6 to under 12 years of age: 
Oral dosage is 5 to 10 milligrams every 4 hours or 15 milligrams every 6 
to 8 hours, not to exceed 60 milligrams in 24 hours, or as directed by a 
doctor. Children 2 to under 6 years of age: Oral dosage is 2.5 to 5 
milligrams every 4 hours or 7.5 milligrams every 6 to 8 hours, not to 
exceed 30 milligrams in 24 hours, or as directed by a doctor. Children 
under 2 years of age: Consult a doctor.
    (iv) For products containing diphenhydramine citrate identified in 
Sec. 341.14(a)(5). Adults and children 12 years of age and over: oral 
dosage is 38 milligrams every 4 hours, not to exceed 228 milligrams in 
24 hours, or as directed by a doctor. Children 6 to under 12 years of 
age: oral dosage is 19 milligrams every 4 hours, not to exceed 114 
milligrams in 24 hours, or as directed by a doctor. Children under 6 
years of age: consult a doctor.
    (v) For products containing diphenhydramine hydrochloride identified 
in Sec. 341.14(a)(6). Adults and children 12 years of age and over: oral 
dosage is 25 milligrams every 4 hours, not to exceed 150 milligrams in 
24 hours, or as directed by a doctor. Children 6 to under 12 years of 
age: oral dosage is 12.5 milligrams every 4 hours, not to exceed 75 
milligrams in 24 hours, or as directed by a doctor. Children under 6 
years of age: consult a doctor.
    (2) Topical antitussives--(i) For products containing camphor 
identified in Sec. 341.14(b)(1) in a suitable ointment vehicle. The 
product contains 4.7 to 5.3 percent camphor. Adults and children 2 to 
under 12 years of age: Rub on the throat and chest as a thick layer. The 
area of application may be covered with a warm, dry cloth if desired. 
However, clothing should be left loose about the throat and chest to 
help the vapors rise to reach the nose and mouth. Applications may be 
repeated up to three times daily or as directed by a doctor. Children 
under 2 years of age: consult a doctor.
    (ii) For products containing menthol identified in Sec. 341.14(b)(2) 
in a suitable ointment vehicle. The product contains 2.6 to 2.8 percent 
menthol. Adults and children 2 to under 12 years of age: Rub on the 
throat and chest as a thick layer. The area of application may be 
covered with a warm, dry cloth if desired. However, clothing should be 
left loose about the throat and chest to help the vapors rise to reach 
the nose

[[Page 248]]

and mouth. Applications may be repeated up to three times daily or as 
directed by a doctor. Children under 2 years of age: consult a doctor.
    (iii) For products containing menthol identified in 
Sec. 341.14(b)(2) in a lozenge. The product contains 5 to 10 milligrams 
menthol. Adults and children 2 to under 12 years of age: Allow lozenge 
to dissolve slowly in the mouth. May be repeated every hour as needed or 
as directed by a doctor. Children under 2 years of age: Consult a 
doctor.
    (iv) For products containing camphor identified in Sec. 341.14(b)(1) 
for steam inhalation use. The product contains 6.2 percent camphor. 
Adults and children 2 to under 12 years of age: Add 1 tablespoonful of 
solution, for each quart of water, directly to the water in a hot steam 
vaporizer, bowl, or wash basin; or add 1\1/2\ teaspoonsful of solution, 
for each pint of water, to an open container of boiling water. Breathe 
in the medicated vapors. May be repeated up to three times daily or as 
directed by a doctor. Children under 2 years of age: consult a doctor.
    (v) For products containing menthol identified in Sec. 341.14(b)(2) 
for steam inhalation use. The product contains 3.2 percent menthol. 
Adults and children 2 to under 12 years of age: Add 1 tablespoonful of 
solution, for each quart of water, directly to the water in a hot steam 
vaporizer, bowl, or wash basin; or add 1\1/2\ teaspoonsful of solution, 
for each pint of water, to an open container of boiling water. Breathe 
in the medicated vapors. May be repeated up to three times daily or as 
directed by a doctor. Children under 2 years of age: consult a doctor.
    (e) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this section.
    (f) Exemption from the general accidental overdose warning. The 
labeling for antitussive drug products containing the active ingredient 
identified in Sec. 341.14(b)(2) marketed in accordance with 
Sec. 341.74(d)(2)(iii) is exempt from the requirement in Sec. 330.1(g) 
of this chapter that the labeling bear the general warning statement 
``In case of accidental overdose, seek professional assistance or 
contact a poison control center immediately.'' The labeling must 
continue to bear the first part of the general warning in Sec. 330.1(g) 
of this chapter, which states, ``Keep this and all drugs out of the 
reach of children.''
[52 FR 30055, Aug. 12, 1987; 52 FR 35610, Sept. 22, 1987; 53 FR 35809, 
Sept. 15, 1988; 55 FR 27808, July 6, 1990; 55 FR 40383, Oct. 3, 1990; 58 
FR 54236, Oct. 20, 1993; 59 FR 29174, June 3, 1994; 59 FR 36051, July 
15, 1994]



Sec. 341.76  Labeling of bronchodilator drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as a 
``bronchodilator.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the phrase listed in paragraph (b)(1) of this 
section. Other truthful and nonmisleading statements, describing only 
the indications for use that have been established and listed in this 
paragraph (b), may also be used, as provided in Sec. 330.1(c)(2), 
subject to the provisions of section 502 of the act relating to 
misbranding and the prohibition in section 301(d) of the act against the 
introduction or delivery for introduction into interstate commerce of 
unapproved new drugs in violation of section 505(a) of the act.
    (1) ``For temporary relief of shortness of breath, tightness of 
chest, and wheezing due to bronchial asthma.''
    (2) In addition to the required information identified in paragraph 
(b)(1) of this section, the labeling of the product may contain one or 
more of the following statements:
    (i) ``For the'' (select one of the following: ``temporary relief'' 
or ``symptomatic control'') ``of bronchial asthma.''
    (ii) ``Eases breathing for asthma patients'' (which may be followed 
by: ``by reducing spasms of bronchial muscles'').
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) ``Do not use this product unless a diagnosis of asthma has been 
made by a doctor.''
    (2) ``Do not use this product if you have heart disease, high blood 
pressure, thyroid disease, diabetes, or difficulty in urination due to 
enlargement of the prostate gland unless directed by a doctor.''

[[Page 249]]

    (3) ``Do not use this product if you have ever been hospitalized for 
asthma or if you are taking any prescription drug for asthma unless 
directed by a doctor.''
    (4) ``Drug interaction precaution. Do not use this product if you 
are now taking a prescription monoamine oxidase inhibitor (MAOI) 
(certain drugs for depression, psychiatric or emotional conditions, or 
Parkinson's disease), or for 2 weeks after stopping the MAOI drug. If 
you are uncertain whether your prescription drug contains an MAOI, 
consult a health professional before taking this product.''
    (5) For products containing ephedrine, ephedrine hydrochloride, 
ephedrine sulfate, or racephedrine hydrochloride identified in 
Sec. 341.16 (a), (b), (c), and (f). (i) ``Do not continue to use this 
product, but seek medical assistance immediately if symptoms are not 
relieved within 1 hour or become worse.''
    (ii) ``Some users of this product may experience nervousness, 
tremor, sleeplessness, nausea, and loss of appetite. If these symptoms 
persist or become worse, consult your doctor.''
    (6) For products containing epinephrine, epinephrine bitartrate, or 
racepinephrine hydrochloride identified in Sec. 341.16 (d), (e), and 
(g). (i) ``Do not use this product more frequently or at higher doses 
than recommended unless directed by a doctor. [first sentence in 
boldface type] Excessive use may cause nervousness and rapid heart beat, 
and, possibly, adverse effects on the heart.''
    (ii) ``Do not continue to use this product, but seek medical 
assistance immediately if symptoms are not relieved within 20 minutes or 
become worse.'' [sentence in boldface type]
    (iii) For products intended for use in a hand-held rubber bulb 
nebulizer. ``Do not use this product if it is brown in color or 
cloudy.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) For products containing ephedrine, ephedrine hydrochloride, 
ephedrine sulfate, or racephedrine hydrochloride identified in 
Sec. 341.16 (a), (b), (c), and (f). Adults and children 12 years of age 
and over: Oral dosage is 12.5 to 25 milligrams every 4 hours, not to 
exceed 150 milligrams in 24 hours, or as directed by a doctor. Do not 
exceed recommended dose unless directed by a doctor. Children under 12 
years of age: Consult a doctor.
    (2) For products containing epinephrine, epinephrine bitartrate, and 
racepinephrine hydrochloride identified in Sec. 341.16(d), (e), and (g) 
for use in a hand-held rubber bulb nebulizer. The ingredient is used in 
an aqueous solution at a concentration equivalent to 1 percent 
epinephrine. Inhalation dosage for adults, children 12 years of age and 
over, and children 4 to under 12 years of age: 1 to 3 inhalations not 
more often than every 3 hours. The use of this product by children 
should be supervised by an adult. Children under 4 years of age: Consult 
a doctor.

(Collection of information requirement approved by the Office of 
Management and Budget under control number 0910-0237)

[51 FR 35339, Oct. 2, 1986, as amended at 52 FR 7126, Mar. 9, 1987; 52 
FR 7830, Mar. 13, 1987; 53 FR 35810, Sept. 15, 1988; 58 FR 54242, Oct. 
20, 1993; 61 FR 25146, May 20, 1996; 62 FR 9684, Mar. 4, 1997]



Sec. 341.78  Labeling of expectorant drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as an 
``expectorant.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the following: ``Helps loosen phlegm (mucus) 
and thin bronchial secretions to'' (select one or more of the following: 
``rid the bronchial passageways of bothersome mucus,'' ``drain bronchial 
tubes,'' and ``make coughs more productive''). Other truthful and 
nonmisleading statements, describing only the indications for use that 
have been established and listed in this paragraph (b), may also be 
used, as provided in Sec. 330.1(c)(2) of this chapter, subject to the 
provisions of section 502 of the act relating to misbranding and the 
prohibition in section 301(d) of the act against the introduction or 
delivery for introduction into interstate commerce of unapproved new 
drugs in violation of section 505(a) of the act.
    (c) Warnings. The labeling of the product contains the following 
warnings, under the heading ``Warnings'':

[[Page 250]]

    (1) ``A persistent cough may be a sign of a serious condition. If 
cough persists for more than 1 week, tends to recur, or is accompanied 
by a fever, rash, or persistent headache, consult a doctor.''
    (2) For expectorant drug products labeled for adults or for adults 
and children under 12 years of age. ``Do not take this product for 
persistent or chronic cough such as occurs with smoking, asthma, chronic 
bronchitis, or emphysema, or where cough is accompanied by excessive 
phlegm (mucus) unless directed by a doctor.''
    (3) For expectorant drug products labeled only for children under 12 
years of age. ``Do not give this product for persistent or chronic cough 
such as occurs with asthma or if cough is accompanied by excessive 
phlegm (mucus) unless directed by a doctor.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'' for products containing 
guaifenesin identified in Sec. 341.18: Adults and children 12 years of 
age and over: oral dosage is 200 to 400 milligrams every 4 hours not to 
exceed 2,400 milligrams in 24 hours. Children 6 to under 12 years of 
age: oral dosage is 100 to 200 milligrams every 4 hours not to exceed 
1,200 milligrams in 24 hours. Children 2 to under 6 years of age: oral 
dosage is 50 to 100 milligrams every 4 hours not to exceed 600 
milligrams in 24 hours. Children under 2 years of age: consult a doctor.
    (e) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this section.
[54 FR 8509, Feb. 28, 1989, as amended at 57 FR 29177, June 30, 1992]



Sec. 341.80  Labeling of nasal decongestant drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as a 
``nasal decongestant.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the phrase listed in paragraph (b)(1) of this 
section, as appropriate, and may contain any additional phrases listed 
in paragraph (b)(2) of this section. Other truthful and nonmisleading 
statements, describing only the indications for use that have been 
established and listed in paragraphs (b)(1) and (b)(2) of this section, 
may also be used, as provided in Sec. 330.1(c)(2) of this chapter, 
subject to the provisions of section 502 of the Federal Food, Drug, and 
Cosmetic Act (the act) relating to misbranding and the prohibition in 
section 301(d) of the act against the introduction or delivery for 
introduction into interstate commerce of unapproved new drugs in 
violation of section 505(a) of the act.
    (1) (Select one of the following: ``For the temporary relief of 
nasal congestion'' or ``Temporarily relieves nasal congestion'') (which 
may be followed by any of the following in paragraphs (b)(1) (i), (ii), 
and (iii) of this section):
    (i) ``due to'' (select one of the following: ``the common cold'' or 
``a cold'').
    (ii) ``due to'' (select one of the following: ``hay fever,'' ``hay 
fever (allergic rhinitis),'' ``hay fever or other upper respiratory 
allergies,'' or ``hay fever or other upper respiratory allergies 
(allergic rhinitis)'').
    (iii) ``associated with sinusitis.''
    (2) In addition to the information identified in paragraph (b)(1) of 
this section, the labeling of the product may contain any (one or more) 
of the following statements:
    (i) (Select one of the following: ``For the temporary relief of'' or 
``Temporarily relieves'') (select one of the following: ``stuffy nose,'' 
``stopped up nose,'' ``nasal stuffiness,'' or ``clogged up nose.'')
    (ii) (Select one of the following: ``Reduces swelling of,'' 
``Decongests,'' or ``Helps clear'') ``nasal passages; shrinks swollen 
membranes.''
    (iii) ``Temporarily restores freer breathing through the nose.''
    (iv) ``Helps decongest sinus openings and passages; temporarily 
relieves sinus congestion and pressure.''
    (v) ``Promotes nasal and/or sinus drainage; temporarily relieves 
sinus congestion and pressure.''
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) Oral nasal decongestants--(i) For products containing 
phenylephrine hydrochloride, pseudoephedrine hydrochloride, or 
pseudoephedrine sulfate identified in Sec. 341.20 (a)(1), (a)(2), and 
(a)(3) when labeled for adults. (A) ``Do not exceed

[[Page 251]]

recommended dosage. [first sentence in boldface type] If nervousness, 
dizziness, or sleeplessness occur, discontinue use and consult a 
doctor.''
    (B) ``If symptoms do not improve within 7 days or are accompanied by 
fever, consult a doctor.''
    (C) ``Do not take this product if you have heart disease, high blood 
pressure, thyroid disease, diabetes, or difficulty in urination due to 
enlargement of the prostate gland unless directed by a doctor.''
    (D) ``Drug interaction precaution. Do not use this product if you 
are now taking a prescription monoamine oxidase inhibitor (MAOI) 
(certain drugs for depression, psychiatric or emotional conditions, or 
Parkinson's disease), or for 2 weeks after stopping the MAOI drug. If 
you are uncertain whether your prescription drug contains an MAOI, 
consult a health professional before taking this product.''
    (ii) For products containing phenylephrine hydrochloride, 
pseudoephedrine hydrochloride, or pseudoephedrine sulfate identified in 
Sec. 341.20 (a)(1), (a)(2), and (a)(3) when labeled for children under 
12 years of age. (A) ``Do not exceed recommended dosage. [first sentence 
in boldface type] If nervousness, dizziness, or sleeplessness occur, 
discontinue use and consult a doctor.''
    (B) ``If symptoms do not improve within 7 days or are accompanied by 
fever, consult a doctor.''
    (C) ``Do not give this product to a child who has heart disease, 
high blood pressure, thyroid disease, or diabetes unless directed by a 
doctor.''
    (D) ``Drug interaction precaution. Do not give this product to a 
child who is taking a prescription monoamine oxidase inhibitor (MAOI) 
(certain drugs for depression, psychiatric or emotional conditions), or 
for 2 weeks after stopping the MAOI drug. If you are uncertain whether 
your child's prescription drug contains an MAOI, consult a health 
professional before giving this product.''
    (iii) For oral nasal decongestant products labeled for both adults 
and children under 12 years of age. The labeling of the product contains 
the warnings identified in paragraph (c)(1)(i) of this section.
    (2) Topical nasal decongestants--(i) For products containing any 
topical nasal decongestant identified in Sec. 341.20(b) when labeled for 
adults. (A) ``Do not exceed recommended dosage.'' [sentence in boldface 
type]
    (B) ``This product may cause temporary discomfort such as burning, 
stinging, sneezing, or an increase in nasal discharge.''
    (C) ``The use of this container by more than one person may spread 
infection.''
    (ii) [Reserved]
    (iii) For products containing ephedrine, ephedrine hydrochloride, 
ephedrine sulfate, naphazoline hydrochloride, oxymetazoline 
hydrochloride, phenylephrine hydrochloride, or xylometazoline 
hydrochloride identified in Sec. 341.20 (b)(2), (b)(3), (b)(4), (b)(6), 
(b)(7), (b)(8), and (b)(10) when used as nasal sprays, drops, or jellies 
and when labeled for adults. (A) ``Do not use this product for more than 
3 days. Use only as directed. Frequent or prolonged use may cause nasal 
congestion to recur or worsen. If symptoms persist, consult a doctor.''
    (B) ``Do not use this product if you have heart disease, high blood 
pressure, thyroid disease, diabetes, or difficulty in urination due to 
enlargement of the prostate gland unless directed by a doctor.''
    (iv) For products containing naphazoline hydrochloride identified in 
Sec. 341.20(b)(6) at a concentration of 0.05 percent. ``Do not use this 
product in children under 12 years of age because it may cause sedation 
if swallowed.''
    (v) For products containing propylhexedrine identified in 
Sec. 341.20(b)(9) when used in an inhalant dosage form and when labeled 
for adults. ``Do not use this product for more than 3 days. Use only as 
directed. Frequent or prolonged use may cause nasal congestion to recur 
or worsen. If symptoms persist, consult a doctor.''
    (vi) For products containing any topical nasal decongestant 
identified in Sec. 341.20(b) when labeled for children under 12 years of 
age. The labeling of the product contains the warnings identified in 
paragraph (c)(2)(i) of this section.
    (vii) [Reserved]
    (viii) For products containing ephedrine, ephedrine hydrochloride, 
ephedrine sulfate, naphazoline hydrochloride,

[[Page 252]]

oxymetazoline hydrochloride, phenylephrine hydrochloride, or 
xylometazoline hydrochloride identified in Sec. 341.20(b)(2), (b)(3), 
(b)(4), (b)(6), (b)(7), (b)(8), and (b)(10) when used as nasal sprays, 
drops, or jellies and when labeled for children under 12 years of age. 
(A) ``Do not use this product for more than 3 days. Use only as 
directed. Frequent or prolonged use may cause nasal congestion to recur 
or worsen. If symptoms persist, consult a doctor.''
    (B) ``Do not use this product in a child who has heart disease, high 
blood pressure, thyroid disease, or diabetes unless directed by a 
doctor.''
    (ix) For products containing propylhexedrine identified in 
Sec. 341.20(b)(9) when used in an inhalant dosage form and when labeled 
for children under 12 years of age. ``Do not use this product for more 
than 3 days. Use only as directed. Frequent or prolonged use may cause 
nasal congestion to recur or worsen. If symptoms persist, consult a 
doctor.''
    (x) For topical nasal decongestant products labeled for both adults 
and for children under 12 years of age. The labeling of the product 
contains the applicable warnings identified in paragraphs (c)(2)(i), 
(c)(2)(ii), (c)(2)(iii), and (c)(2)(v) of this section.
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) Oral nasal decongestants--(i) For products containing 
phenylephrine hydrochloride identified in Sec. 341.20(a)(1). Adults and 
children 12 years of age and over: 10 milligrams every 4 hours not to 
exceed 60 milligrams in 24 hours. Children 6 to under 12 years of age: 5 
milligrams every 4 hours not to exceed 30 milligrams in 24 hours. 
Children 2 to under 6 years of age: 2.5 milligrams every 4 hours not to 
exceed 15 milligrams in 24 hours. Children under 2 years of age: consult 
a doctor.
    (ii) For products containing pseudoephedrine hydrochloride or 
pseudoephedrine sulfate identified in Sec. 341.20 (a)(2) and (a)(3). 
Adults and children 12 years of age and over: 60 milligrams every 4 to 6 
hours not to exceed 240 milligrams in 24 hours. Children 6 to under 12 
years of age: 30 milligrams every 4 to 6 hours not to exceed 120 
milligrams in 24 hours. Children 2 to under 6 years of age: 15 
milligrams every 4 to 6 hours not to exceed 60 milligrams in 24 hours. 
Children under 2 years of age: consult a doctor.
    (2) Topical nasal decongestants--(i) [Reserved]
    (ii) For products containing ephedrine, ephedrine hydrochloride, or 
ephedrine sulfate identified in Sec. 341.20(b) (2), (3), and (4)--(A) 
Nasal drops or sprays--For a 0.5-percent aqueous solution. Adults and 
children 12 years of age and over: 2 or 3 drops or sprays in each 
nostril not more often than every 4 hours. Children 6 to under 12 years 
of age (with adult supervision): 1 or 2 drops or sprays in each nostril 
not more often than every 4 hours. Children under 6 years of age: 
consult a doctor.
    (B) Nasal jelly--For a 0.5-percent water-based jelly. Adults and 
children 6 to under 12 years of age (with adult supervision): place a 
small amount in each nostril and inhale well back into the nasal 
passages. Use not more often than every 4 hours.
    (iii) For products containing naphazoline hydrochloride identified 
in Sec. 341.20(b)(6)--(A) Nasal drops or sprays--(1) For a 0.05-percent 
aqueous solution. Adults and children 12 years of age and over: 1 or 2 
drops or sprays in each nostril not more often than every 6 hours. Do 
not give to children under 12 years of age unless directed by a doctor.
    (2) For a 0.025-percent aqueous solution. Children 6 to under 12 
years of age (with adult supervision): 1 or 2 drops or sprays in each 
nostril not more often than every 6 hours. Children under 6 years of 
age: consult a doctor.
    (B) Nasal jelly--(1) For a 0.05-percent water-based jelly. Adults 
and children 12 years of age and over: place a small amount in each 
nostril and inhale well back into the nasal passages. Use not more often 
than every 6 hours. Do not give to children under 12 years of age unless 
directed by a doctor.
    (2) For a 0.025-percent water-based jelly. Children 6 to under 12 
years of age (with adult supervision): place a small amount in each 
nostril and inhale well back into the nasal passages. Use not more often 
than every 6 hours. Children under 6 years of age: consult a doctor.

[[Page 253]]

    (iv) For products containing oxymetazoline hydrochloride identified 
in Sec. 341.20(b)(7)--(A) Nasal drops or sprays--(1) For a 0.05-percent 
aqueous solution. Adults and children 6 to under 12 years of age (with 
adult supervision): 2 or 3 drops or sprays in each nostril not more 
often than every 10 to 12 hours. Do not exceed 2 doses in any 24-hour 
period. Children under 6 years of age: consult a doctor.
    (2) A 0.025-percent aqueous solution in a container having either a 
calibrated dropper or a metered-dose spray that delivers no more than 
0.027 milligrams of oxymetazoline per three drops or three sprays. 
Children 2 to under 6 years of age (with adult supervision): 2 or 3 
drops or sprays in each nostril not more often than every 10 to 12 
hours. Use only recommended amount. Do not exceed 2 doses in any 24-hour 
period. [previous two sentences in boldface type] Children under 2 years 
of age: consult a doctor.
    (B) Nasal jelly--For a 0.05-percent water-based jelly. Adults and 
children 6 to under 12 years of age (with adult supervision): place a 
small amount in each nostril and inhale well back into the nasal 
passages. Use not more often than every 10 to 12 hours. Do not exceed 2 
doses in any 24-hour period. Children under 6 years of age: consult a 
doctor.
    (v) For products containing phenylephrine hydrochloride identified 
in Sec. 341.20(b)(8)--(A) Nasal drops or sprays--(1) For a 1-percent 
aqueous solution. Adults and children 12 years of age and over: 2 or 3 
drops or sprays in each nostril not more often than every 4 hours. Do 
not give to children under 12 years of age unless directed by a doctor.
    (2) For a 0.5-percent aqueous solution. Adults and children 12 years 
of age and over: 2 or 3 drops or sprays in each nostril not more often 
than every 4 hours. Do not give to children under 12 years of age unless 
directed by a doctor.
    (3) For a 0.25-percent aqueous solution. Adults and children 6 to 
under 12 years of age (with adult supervision): 2 or 3 drops or sprays 
in each nostril not more often than every 4 hours. Children under 6 
years of age: consult a doctor.
    (4) A 0.125-percent aqueous solution in a container having either a 
calibrated dropper or a metered-dose spray that delivers no more than 
0.135 milligrams of phenylephrine per three drops or three sprays. 
Children 2 to under 6 years of age (with adult supervision): 2 or 3 
drops or sprays in each nostril not more often than every 4 hours. Use 
only recommended amount. [previous sentence in boldface type] Children 
under 2 years of age: consult a doctor.
    (B) Nasal jelly--(1) For a 1-percent water-based jelly. Adults and 
children 12 years of age and over: place a small amount in each nostril 
and inhale well back into the nasal passages. Use not more often than 
every 4 hours. Do not give to children under 12 years of age unless 
directed by a doctor.
    (2) For a 0.5-percent water-based jelly. Adults and children 12 
years of age and over: place a small amount in each nostril and inhale 
well back into the nasal passages. Use not more often than every 4 
hours. Do not give to children under 12 years of age unless directed by 
a doctor.
    (3) For a 0.25-percent water-based jelly. Adults and children 6 to 
under 12 years of age (with adult supervision): place a small amount in 
each nostril and inhale well back into the nasal passages. Use not more 
often than every 4 hours. Children under 6 years of age: consult a 
doctor.
    (vi) For products containing propylhexedrine identified in 
Sec. 341.20(b)(9) when used in an inhalant dosage form. The product 
delivers in each 800 milliliters of air 0.40 to 0.50 milligrams of 
propylhexedrine. Adults and children 6 to under 12 years of age (with 
adult supervision): 2 inhalations in each nostril not more often than 
every 2 hours. Children under 6 years of age: consult a doctor.
    (vii) For products containing xylometazoline hydrochloride 
identified in Sec. 341.20(b)(10)--(A) Nasal drops or sprays--(1) For a 
0.1-percent aqueous solution. Adults and children 12 years of age and 
over: 2 or 3 drops or sprays in each nostril not more often than every 8 
to 10 hours. Do not give to children under 12 years of age unless 
directed by a doctor.
    (2) A 0.05-percent aqueous solution in a container having either a 
calibrated dropper or a metered-dose spray that delivers no more than 
0.054 milligrams of

[[Page 254]]

xylometazoline per three drops or three sprays. Children 6 to under 12 
years of age (with adult supervision): 2 or 3 drops or sprays in each 
nostril not more often than every 8 to 10 hours. Children 2 to under 6 
years of age (with adult supervision): 2 or 3 drops or sprays in each 
nostril not more often than every 8 to 10 hours. Use only recommended 
amount. Do not exceed 3 doses in any 24-hour period. [previous two 
sentences in boldface type] Children under 2 years of age: consult a 
doctor.
    (B) Nasal jelly--(1) For a 0.1-percent water-based jelly. Adults and 
children 12 years of age and over: place a small amount in each nostril 
and inhale well back into the nasal passages. Use not more often than 
every 8 to 10 hours. Do not give to children under 12 years of age 
unless directed by a doctor.
    (2) For a 0.05-percent water-based jelly. Children 6 to under 12 
years of age (with adult supervision): place a small amount in each 
nostril and inhale well back into the nasal passages. Use not more often 
than every 8 to 10 hours. Children under 6 years of age: consult a 
doctor.
    (viii) Other required statements--For products containing 
propylhexedrine identified in Sec. 341.20(b)(9) when used in an inhalant 
dosage form. (A) ``This inhaler is effective for a minimum of 3 months 
after first use.''
    (B) ``Keep inhaler tightly closed.''
[59 FR 43409, Aug. 23, 1994]



Sec. 341.90  Professional labeling.

    The labeling of the product provided to health professionals (but 
not to the general public) may contain the following additional dosage 
information for products containing the active ingredients identified 
below:
    (a) For products containing ephedrine, ephedrine hydrochloride, 
ephedrine sulfate, or racephedrine hydrochloride identified in 
Sec. 341.16 (a), (b), (c), and (f). Children 6 to under 12 years of age: 
oral dosage is 6.25 to 12.5 milligrams every 4 hours, not to exceed 75 
milligrams in 24 hours. Children 2 to under 6 years of age: oral dosage 
is 0.3 to 0.5 milligram per kilogram of body weight every 4 hours, not 
to exceed 2 milligrams per kilogram of body weight in 24 hours.
    (b) For products containing chlophedianol hydrochloride identified 
in 341.14(a)(1). Children 2 to under 6 years of age: oral dosage is 12.5 
milligrams every 6 to 8 hours, not to exceed 50 milligrams in 24 hours.
    (c) For products containing codeine ingredients identified in 
Sec. 341.14(a)(2). (1) Children 2 to under 6 years of age: Oral dosage 
is 1 milligram per kilogram body weight per day administered in four 
equal divided doses. The average body weight for each age may also be 
used to determine dosage as follows: For children 2 years of age 
(average body weight, 12 kilograms), the oral dosage is 3 milligrams 
every 4 to 6 hours, not to exceed 12 milligrams in 24 hours; for 
children 3 years of age (average body weight, 14 kilograms), the oral 
dosage is 3.5 milligrams every 4 to 6 hours, not to exceed 14 milligrams 
in 24 hours; for children 4 years of age (average body weight, 16 
kilograms), the oral dosage is 4 milligrams every 4 to 6 hours, not to 
exceed 16 milligrams in 24 hours: for children 5 years of age (average 
body weight, 18 kilograms), the oral dosage is 4.5 milligrams every 4 to 
6 hours, not to exceed 18 milligrams in 24 hours. The manufacturer must 
relate these dosages for its specific product dosages for its specific 
product to the use of the calibrated measuring device discussed in 
paragraph (c)(3) of this section. If age is used to determine the dose, 
the directions must include instructions to reduce the dose for low-
weight children.
    (2) Parents should be instructed to obtain and use a calibrated 
measuring device for administering the drug to the child, to use extreme 
care in measuring the dosage, and not exceed the recommended daily 
dosage.
    (3) A dispensing device (such as a dropper calibrated for age or 
weight) should be dispensed along with the product when it is intended 
for use in children 2 to under 6 years of age to prevent possible 
overdose due to improper measuring of the dose.
    (4) Codeine is not recommended for use in children under 2 years of 
age. Children under 2 years may be more susceptible to the respiratory 
depressant effects of codeine, including respiratory arrest, coma, and 
death.

[[Page 255]]

    (d) The following labeling indication may be used for products 
containing guaifenesin identified in Sec. 341.18 when used as a single 
ingredient product. ``Helps loosen phlegm and thin bronchial secretions 
in patients with stable chronic bronchitis.''
    (e) For products containing brompheniramine maleate identified in 
Sec. 341.12(a). Children 2 to under 6 years of age: oral dosage is 1 
milligram every 4 to 6 hours, not to exceed 6 milligrams in 24 hours.
    (f) For products containing chlorcyclizine hydrochloride identified 
in Sec. 341.12(b). Children 6 to under 12 years of age: oral dosage is 
12.5 milligrams every 6 to 8 hours, not to exceed 37.5 milligrams in 24 
hours. Children 2 to under 6 years of age: oral dosage is 6.25 
milligrams every 6 to 8 hours, not to exceed 18.75 milligrams in 24 
hours.
    (g) For products containing chlorpheniramine maleate identified in 
Sec. 341.12(c). Children 2 to under 6 years of age: oral dosage is 1 
milligram every 4 to 6 hours, not to exceed 6 milligrams in 24 hours.
    (h) For products containing dexbrompheniramine maleate identified in 
Sec. 341.12(d). Children 2 to under 6 years of age: oral dosage is 0.5 
milligram every 4 to 6 hours, not to exceed 3 milligrams in 24 hours.
    (i) For products containing dexchlorpheniramine maleate identified 
in Sec. 341.12(e). Children 2 to under 6 years: oral dosage is 0.5 
milligram every 4 to 6 hours, not to exceed 3 milligrams in 24 hours.
    (j) For products containing diphenhydramine citrate identified in 
Sec. 341.12(f). Children 2 to under 6 years of age: oral dosage is 9.5 
milligrams every 4 to 6 hours, not to exceed 57 milligrams in 24 hours.
    (k) For products containing diphenhydramine hydrochloride identified 
in Sec. 341.12(g). Children 2 to under 6 years of age: oral dosage is 
6.25 milligrams every 4 to 6 hours, not to exceed 37.5 mg in 24 hours.
    (l) For products containing doxylamine succinate identified in 
Sec. 341.12(h). Children 2 to under 6 years of age: oral dosage is 1.9 
to 3.125 milligrams every 4 to 6 hours, not to exceed 18.75 milligrams 
in 24 hours.
    (m) For products containing phenindamine tartrate identified in 
Sec. 341.12(i). Children 2 to under 6 years of age: oral dosage is 6.25 
milligrams every 4 to 6 hours, not to exceed 37.5 milligrams in 24 
hours.
    (n) For products containing pheniramine maleate identified in 
Sec. 341.12(j). Children 2 to under 6 years of age: oral dosage is 3.125 
to 6.25 milligrams every 4 to 6 hours, not to exceed 37.5 milligrams in 
24 hours.
    (o) For products containing pyrilamine maleate identified in 
Sec. 341.12(k). Children 2 to under 6 years of age: oral dosage is 6.25 
to 12.5 milligrams every 6 to 8 hours, not to exceed 50 milligrams in 24 
hours.
    (p) For products containing thonzylamine hydrochloride identified in 
Sec. 341.12(l). Children 2 to under 6 years of age: oral dosage is 12.5 
to 25 milligrams every 4 to 6 hours, not to exceed 150 milligrams in 24 
hours.
    (q) For products containing triprolidine hydrochloride identified in 
Sec. 341.12(m). Children 4 to under 6 years of age: oral dosage is 0.938 
milligram every 4 to 6 hours, not to exceed 3.744 milligrams in 24 
hours. Children 2 to under 4 years of age: oral dosage is 0.625 
milligram every 4 to 6 hours, not to exceed 2.5 milligrams in 24 hours. 
Infants 4 months to under 2 years of age: oral dosage is 0.313 milligram 
every 4 to 6 hours, not to exceed 1.252 milligrams in 24 hours.
    (r) For products containing diphenhydramine citrate identified in 
Sec. 341.14(a)(5). Children 2 to under 6 years of age: oral dosage is 
9.5 milligrams every 4 hours, not to exceed 57 milligrams in 24 hours.
    (s) For products containing diphenhydramine hydrochloride identified 
in Sec. 341.14(a)(6). Children 2 to under 6 years of age: oral dosage is 
6.25 milligrams every 4 hours, not to exceed 37.5 milligrams in 24 
hours.
[51 FR 35339, Oct. 2, 1986, as amended at 52 FR 30057, Aug. 12, 1987; 54 
FR 8509, Feb. 28, 1989; 57 FR 58376, Dec. 9, 1992; 59 FR 4218, Jan. 28, 
1994; 59 FR 29174, June 3, 1994; 59 FR 36051, July 15, 1994]

[[Page 256]]



PART 344--TOPICAL OTIC DRUG PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                      Subpart A--General Provisions

Sec.
344.1  Scope.
344.3  Definitions.

                      Subpart B--Active Ingredients

344.10  Topical otic active ingredient.

                           Subpart C--Labeling

344.50  Labeling of topical otic drug products.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  51 FR 28660, Aug. 8, 1986, unless otherwise noted:



                      Subpart A--General Provisions



Sec. 344.1  Scope.

    (a) An over-the-counter topical otic drug product in a form suitable 
for topical administration is generally recognized as safe and effective 
and is not misbranded if it meets each of the conditions in this part in 
addition to each of the general conditions established in Sec. 330.1.
    (b) References in this part to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 344.3  Definitions.

    As used in this part:
    (a) Anhydrous glycerin. An ingredient that may be prepared by 
heating glycerin U.S.P. at 150 deg. C for 2 hours to drive off the 
moisture content.
    (b) Earwax removal aid. A drug used in the external ear canal that 
aids in the removal of excessive earwax.



                      Subpart B--Active Ingredients



Sec. 344.10  Topical otic active ingredient.

    The active ingredient of the product consists of carbamide peroxide 
6.5 percent formulated in an anhydrous glycerin vehicle.



                           Subpart C--Labeling



Sec. 344.50  Labeling of topical otic drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as an 
``earwax removal aid.''
    (b) Indication. The labeling of the product states, under the 
heading ``Indication,'' the following: ``For occasional use as an aid 
to'' (which may be followed by: ``soften, loosen, and'') ``remove 
excessive earwax.'' Other truthful and nonmisleading statements, 
describing only the indications for use that have been established and 
listed in this paragraph (b), may also be used, as provided in 
Sec. 330.1(c)(2), subject to the provisions of section 502 of the act 
relating to misbranding and the prohibition in section 301(d) of the act 
against the introduction or delivery for introduction into interstate 
commerce of unapproved new drugs in violation of section 505(a) of the 
act.
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) ``Do not use if you have ear drainage or discharge, ear pain, 
irritation, or rash in the ear or are dizzy; consult a doctor.''
    (2) ``Do not use if you have an injury or perforation (hole) of the 
ear drum or after ear surgery unless directed by a doctor.''
    (3) ``Do not use for more than 4 days; if excessive earwax remains 
after use of this product, consult a doctor.''
    (4) ``Avoid contact with the eyes.''
    (d) Directions. The labeling of the product contains the following 
statement under the heading ``Directions'': FOR USE IN THE EAR ONLY. 
Adults and children over 12 years of age: tilt head sideways and place 5 
to 10 drops into ear. Tip of applicator should not enter ear canal. Keep 
drops in ear for several minutes by keeping head tilted or placing 
cotton in the ear. Use twice daily for up to 4 days if needed, or as 
directed by a doctor. Any wax remaining after treatment may be removed 
by gently flushing the ear with warm water, using a soft rubber bulb ear 
syringe. Children under 12 years of age: consult a doctor.

[[Page 257]]

    (e) Optional wording. The word ``physician'' may be substituted for 
the word ``doctor'' in any of the labeling statements in this section.
[51 FR 28660, Aug. 8, 1986; 52 FR 7830, Mar. 13, 1987]



PART 346--ANORECTAL DRUG PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                      Subpart A--General Provisions

Sec.
346.1  Scope.
346.3  Definitions.

                      Subpart B--Active Ingredients

346.10  Local anesthetic active ingredients.
346.12  Vasoconstrictor active ingredients.
346.14  Protectant active ingredients.
346.16  Analgesic, anesthetic, and antipruritic active ingredients.
346.18  Astringent active ingredients.
346.20  Keratolytic active ingredients.
346.22  Permitted combinations of anorectal active ingredients.

                           Subpart C--Labeling

346.50  Labeling of anorectal drug products.
346.52  Labeling of permitted combinations of anorectal active 
          ingredients.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  55 FR 31779, Aug. 3, 1990, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 346.1  Scope.

    (a) An over-the-counter anorectal drug product in a form suitable 
for external (topical) or intrarectal (rectal) administration is 
generally recognized as safe and effective and is not misbranded if it 
meets each condition in this part and each general condition established 
in Sec. 330.1 of this chapter.
    (b) References in this part to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 212 unless otherwise 
noted.



Sec. 346.3  Definitions.

    As used in this part:
    (a) Analgesic, anesthetic drug. A topically (externally) applied 
drug that relieves pain by depressing cutaneous sensory receptors.
    (b) Anorectal drug. A drug that is used to relieve symptoms caused 
by anorectal disorders in the anal canal, perianal area, and/or the 
lower rectal areas.
    (c) Antipruritic drug. A topically (externally) applied drug that 
relieves itching by depressing cutaneous sensory receptors.
    (d) Astringent drug. A drug that is applied topically (externally) 
to the skin or mucous membranes for a local and limited protein 
coagulant effect.
    (e) External use. Topical application of an anorectal drug product 
to the skin of the perianal area and/or the skin of the anal canal.
    (f) Intrarectal use. Topical application of an anorectal drug 
product to the mucous membrane of the rectum.
    (g) Keratolytic drug. A drug that causes desquamation (loosening) 
and debridement or sloughing of the surface cells of the epidermis.
    (h) Local anesthetic drug. A drug that produces local disappearance 
of pain, burning, itching, irritation, and/or discomfort by reversibly 
blocking nerve conduction when applied to nerve tissue in appropriate 
concentrations.
    (i) Protectant drug. A drug that provides a physical barrier, 
forming a protective coating over skin or mucous membranes.
    (j) Vasoconstrictor. A drug that causes temporary constriction of 
blood vessels.



                      Subpart B--Active Ingredients



Sec. 346.10  Local anesthetic active ingredients.

    The active ingredient of the product consists of any of the 
following when used in the concentration or within the concentration 
range established for each ingredient:
    (a) Benzocaine 5 to 20 percent.
    (b) Benzyl alcohol 1 to 4 percent.
    (c) Dibucaine 0.25 to 1 percent.
    (d) Dibucaine hydrochloride 0.25 to 1 percent.
    (e) Dyclonine hydrochloride 0.5 to 1 percent.
    (f) Lidocaine 2 to 5 percent.
    (g) Pramoxine hydrochloride 1 percent.
    (h) Tetracaine 0.5 to 1 percent.

[[Page 258]]

    (i) Tetracaine hydrochloride 0.5 to 1 percent.



Sec. 346.12  Vasoconstrictor active ingredients.

    The active ingredient of the product consists of any of the 
following when used in the concentration or within the concentration 
range established for each ingredient.
    (a) Ephedrine sulfate 0.1 to 1.25 percent.
    (b) Epinephrine 0.005 to 0.01 percent.
    (c) Epinephrine hydrochloride 0.005 to 0.01 percent.
    (d) Phenylephrine hydrochloride 0.25 percent.



Sec. 346.14  Protectant active ingredients.

    (a) The following active ingredients may be used as the sole 
protectant active ingredient in a product if the ingredient as 
identified constitutes 50 percent or more by weight of the final 
product. In addition, the following active ingredients may be used in 
concentrations of less than 50 percent by weight only when used in 
combinations in accordance with Sec. 346.22 (a), (b), or (n).
    (1) Aluminum hydroxide gel.
    (2) Cocoa butter.
    (3) Glycerin in a 20- to 45-percent (weight/weight) aqueous solution 
so that the final product contains not less than 10 and not more than 45 
percent glycerin (weight/weight). Any combination product containing 
glycerin must contain at least this minimum amount of glycerin.
    (4) Hard fat.
    (5) Kaolin.
    (6) Lanolin.
    (7) Mineral oil.
    (8) Petrolatum.
    (9) Topical starch.
    (10) White petrolatum.
    (b) The following active ingredients may not be used as a sole 
protectant ingredient but may be used in combination with one, two, or 
three other protectant active ingredients in accordance with Sec. 346.22 
(a), (b), (n), and (o) and with the following limitations:
    (1) Calamine not to exceed 25 percent by weight per dosage unit 
(based on the zinc oxide content of calamine).
    (2) Cod liver oil, provided that the product is labeled so that the 
amount of the product that is used in a 24-hour period represents a 
quantity that provides 10,000 U.S.P. units of vitamin A and 400 U.S.P. 
units of cholecalciferol.
    (3) Shark liver oil, provided that the product is labeled so that 
the amount of the product that is used in a 24-hour period represents a 
quantity that provides 10,000 U.S.P. units of vitamin A and 400 U.S.P. 
units of cholecalciferol.
    (4) Zinc oxide not to exceed 25 percent by weight per dosage unit.



Sec. 346.16  Analgesic, anesthetic, and antipruritic active ingredients.

    The active ingredient of the product consists of any of the 
following when used within the concentration range established for each 
ingredient:
    (a) Camphor 0.1 to 3 percent.
    (b) Juniper tar 1 to 5 percent.
    (c) Menthol 0.1 to 1 percent.



Sec. 346.18  Astringent active ingredients.

    The active ingredient of the product consists of any of the 
following when used within the concentration range established for each 
ingredient:
    (a) Calamine, within a concentration range of 5 to 25 percent by 
weight per dosage unit (based on the zinc oxide content of calamine).
    (b) Witch hazel, 10 to 50 percent.
    (c) Zinc oxide, within a concentration range of 5 to 25 percent by 
weight per dosage unit.
[55 FR 31779, Aug. 3, 1990, as amended at 59 FR 28767, June 3, 1994]



Sec. 346.20  Keratolytic active ingredients.

    The active ingredient of the product consists of any of the 
following when used within the concentration range established for each 
ingredient:
    (a) Alcloxa 0.2 to 2 percent.
    (b) Resorcinol 1 to 3 percent.



Sec. 346.22  Permitted combinations of anorectal active ingredients.

    (a) Any two, three, or four protectants identified in (a) 
Sec. 346.14 may be combined, except aluminum hydroxide gel in 
Sec. 346.14(a)(1) and kaolin in Sec. 346.14(a)(5) may not be combined 
with any ingredient in Sec. 346.14(a) (2), (4), (6), (7), (8) and (10), 
and (b) (2) and (3), provided that the combined percentage by weight of 
all protectants in the combination is at least 50 percent of

[[Page 259]]

the final product (e.g., 1 gram of a 2-gram dosage unit). Any protectant 
ingredient included in the combination must be present at a level that 
contributes at least 12.5 percent by weight (e.g., 0.25 gram of a 2-gram 
dosage unit), except cod liver oil and shark liver oil. If an ingredient 
in Sec. 346.14(b) is included in the combination, it must not exceed the 
concentration limit specified in Sec. 346.14(b).
    (b) Any single anorectal ingredient identified in Sec. 346.10, 
346.12, 346.16, 346.18, or 346.20 may be combined with up to four 
protectants in accordance with paragraph (a) of this section.
    (c) Any single local anesthetic identified in Sec. 346.10 may be 
combined with any single vasoconstrictor identified in Sec. 346.12.
    (d) Any single local anesthetic identified in Sec. 346.10 may be 
combined with any single astringent identified in Sec. 346.18.
    (e) Any single local anesthetic identified in Sec. 346.10 may be 
combined with any single keratolytic identified in Sec. 346.20.
    (f) Any single vasoconstrictor identified in Sec. 346.12 may be 
combined with any single astringent identified in Sec. 346.18.
    (g) Any single analgesic, anesthetic, and antipruritic identified in 
Sec. 346.16 may be combined with any single astringent identified in 
Sec. 346.18.
    (h) Any single analgesic, anesthetic, and antipruritic identified in 
Sec. 346.16 may be combined with any single keratolytic identified in 
Sec. 346.20.
    (i) Any single astringent identified in Sec. 346.18 may be combined 
with any single keratolytic identified in Sec. 346.20.
    (j) Any single local anesthetic identified in Sec. 346.10 may be 
combined with any single vasoconstrictor identified in Sec. 346.12 and 
with any single astringent identified in Sec. 346.18.
    (k) Any single local anesthetic identified in Sec. 346.10 may be 
combined with any single astringent identified in Sec. 346.18 and with 
any single keratolytic identified in Sec. 346.20.
    (l) Any single vasoconstrictor identified in Sec. 346.12 may be 
combined with any single analgesic, anesthetic, and antipruritic 
identified in Sec. 346.16 and with any single astringent identified in 
Sec. 346.18.
    (m) Any single analgesic, anesthetic, and antipruritic identified in 
Sec. 346.16 may be combined with any single astringent identified in 
Sec. 346.18 and with any single keratolytic identified in Sec. 346.20.
    (n) Any combination of ingredients listed in paragraphs (c) through 
(m) of this section may be combined with up to four protectants in 
accordance with paragraph (a) of this section.
    (o) Any product containing calamine for use as a protectant and/or 
as an astringent and/or containing zinc oxide for use as a protectant 
and/or as an astringent may not have a total weight of zinc oxide 
exceeding 25 percent by weight per dosage unit.



                           Subpart C--Labeling



Sec. 346.50  Labeling of anorectal drug products.

    The labeling of the product contains the following information for 
anorectal ingredients identified in Secs. 346.10, 346.12, 346.14, 
346.16, 346.18, and 346.20, and for combinations of anorectal 
ingredients identified in Sec. 346.22. Unless otherwise specified, the 
labeling in this subpart is applicable to anorectal drug products for 
both external and intrarectal use.
    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as 
``anorectal (hemorrhoidal),'' ``hemorrhoidal,'' ``hemorrhoidal 
(anorectal) (insert dosage form, e.g., cream, lotion, or ointment).''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' any of the phrases listed in paragraph (b) of 
this section, as appropriate. Other truthful and nonmisleading 
statements, describing only the indications for use that have been 
established and listed in this paragraph, may also be used, as provided 
in Sec. 330.1(c)(2) of this chapter, subject to the provisions of 
section 502 of the Federal Food, Drug, and Cosmetic Act (the act) 
relating to misbranding and the prohibition in section 301(d) of the act 
against the introduction or delivery for introduction into interstate 
commerce of unapproved new drugs in violation of section 505(a) of the 
act.
    (1) (``For the temporary relief of,'' ``Gives temporary relief of,'' 
or ``Helps

[[Page 260]]

relieve the'') (As an option, select one or both of the following: 
``local'' or ``anorectal'') [select one or more of the following: 
``discomfort,'' ``itching,'' or ``itching and discomfort,'' followed by: 
``in the perianal area'' or ``associated with'' (select one or more of 
the following: ``hemorrhoids,'' ``anorectal disorders,'' ``inflamed 
hemorrhoidal tissues,'' ``anorectal inflammation,'' ``hemorrhoidal 
tissues,'' or ``piles (hemorrhoids).'')]
    (2) Additional indications. Indications applicable to each active 
ingredient of the product may be combined to eliminate duplicative words 
or phrases so that the resulting indication is clear and understandable. 
In addition to the indication identified in paragraph (b)(1) of this 
section, the labeling of the product intended for external or 
intrarectal use may also contain the following indications, as 
appropriate.
    (i) For products for external use only containing any ingredient 
identified in Sec. 346.10. ``For the temporary relief of'' (select one 
or more of the following: ``pain,'' ``soreness,'' or ``burning'').
    (ii) For products containing epinephrine or epinephrine 
hydrochloride identified in Sec. 346.12 (b) and (c) for external use 
only, and for products containing ephedrine sulfate or phenylephrine 
hydrochloride identified in Sec. 346.12 (a) and (d).
    (A) ``Temporarily reduces the swelling associated with'' (select one 
of the following: ``irritated hemorrhoidal tissue and other anorectal 
disorders'' or ``irritation in hemorrhoids and other anorectal 
disorders'').
    (B) ``Temporarily shrinks hemorrhoidal tissue.''
    (iii) For products for external use only containing glycerin 
identified in Sec. 346.14(a)(3) and for products for external and/or 
intrarectal use containing any protectant identified in Sec. 346.14(a) 
(2), (4), (6) through (10), and (b) (1) through (4).
    (A) ``Temporarily forms a protective coating over inflamed tissues 
to help prevent drying of tissues.''
    (B) ``Temporarily protects irritated areas.''
    (C) ``Temporarily relieves burning.''
    (D) ``Provides temporary relief from skin irritations.''
    (E) ``Temporarily provides a coating for relief of anorectal 
discomforts.''
    (F) ``Temporarily protects the inflamed, irritated anorectal 
surface'' (select one of the following: ``to help make bowel movements 
less painful'' or ``from irritation and abrasion during bowel 
movement'').
    (G) ``Temporarily protects inflamed perianal skin.''
    (H) ``Temporarily relieves the symptoms of perianal skin 
irritation.''
    (iv) For products containing aluminum hydroxide gel identified in 
Sec. 346.14(a)(1) and for products containing kaolin identified in 
Sec. 346.14(a)(5). ``For the temporary relief of itching associated with 
moist anorectal conditions.''
    (v) For products for external use only containing any analgesic, 
anesthetic, and antipruritic identified in Sec. 346.16.
    (A) ``For the temporary relief of'' (select one or both of the 
following: ``pain'' or ``burning'').
    (B) ``Can help distract from pain.''
    (C) ``May provide a cooling sensation.''
    (vi) For products for external use only containing witch hazel 
identified in Sec. 346.18(b), and for products for external use and/or 
intrarectal use containing calamine or zinc oxide identified in 
Sec. 346.18 (a) and (c).
    (A) ``Aids in protecting irritated anorectal areas.''
    (B) ``Temporary relief of'' (select one or both of the following: 
``irritation'' or ``burning'').
    (vii) For products for external use only containing any ingredient 
identified in Sec. 346.20. The indication in paragraph (b)(1) of this 
section applies.
    (c) Warnings. Warnings applicable to each active ingredient of the 
product may be combined to eliminate duplicative words or phrases so 
that the resulting warning is clear and understandable. The labeling of 
the product contains the following warnings under the heading 
``Warnings'':
    (1) ``If condition worsens or does not improve within 7 days, 
consult a doctor.''
    (2) ``Do not exceed the recommended daily dosage unless directed by 
a doctor.''
    (3) ``In case of bleeding, consult a doctor promptly.''

[[Page 261]]

    (4) For products for external use only. ``Do not put this product 
into the rectum by using fingers or any mechanical device or 
applicator.''
    (5) For products for intrarectal use to be used with a special 
applicator such as a pile pipe or other mechanical device. ``Do not use 
this product with an applicator if the introduction of the applicator 
into the rectum causes additional pain. Consult a doctor promptly.''
    (6) For products for external use only containing any local 
anesthetic identified in Sec. 346.10, menthol identified in 
Sec. 346.16(c), or resorcinol identified in Sec. 346.20(b). ``Certain 
persons can develop allergic reactions to ingredients in this product. 
If the symptom being treated does not subside or if redness, irritation, 
swelling, pain, or other symptoms develop or increase, discontinue use 
and consult a doctor.''
    (7) For products containing any vasoconstrictor identified in 
Sec. 346.12. (i) ``Do not use this product if you have heart disease, 
high blood pressure, thyroid disease, diabetes, or difficulty in 
urination due to enlargement of the prostate gland unless directed by a 
doctor.''
    (ii) ``Drug interaction precaution. Do not use this product if you 
are presently taking a prescription drug for high blood pressure or 
depression, without first consulting your doctor.''
    (iii) For products containing ephedrine sulfate identified in 
Sec. 346.12(a). ``Some users of this product may experience nervousness, 
tremor, sleeplessness, nausea, and loss of appetite. If these symptoms 
persist or become worse, consult your doctor.''
    (8) For products containing aluminum hydroxide gel identified in 
Sec. 346.14(a)(1) and for products containing kaolin identified in 
Sec. 346.14(a)(5). ``Remove petrolatum or greasy ointment before using 
this product because they interfere with the ability of this product to 
adhere properly to the skin area.''
    (9) For products for external use only containinq resorcinol 
identified in Sec. 346.20(b). ``Do not use on open wounds near the 
anus.''
    (d) Directions. Directions applicable to each active ingredient of 
the product may be combined to eliminate duplicative words or phrases so 
that the resulting information is clear and understandable. The labeling 
of the product contains the following information under the heading 
``Directions'':
    (1) ``Adults: When practical, cleanse the affected area'' (select 
one or both of the following: ``with mild soap and warm water and rinse 
thoroughly'' or ``by patting or blotting with an appropriate cleansing 
pad''). ``Gently dry by patting or blotting with toilet tissue or a soft 
cloth before application of this product.'' [Other appropriate 
directions in this section may be inserted here.] ``Children under 12 
years of age: consult a doctor.''
    (2) For products for external use only. ``Apply externally to the 
affected area'' (insert appropriate time interval of administration as 
identified in paragraphs (d)(6), (7), (8), or (9) of this section).
    (3) For products for external use that are pads containing anorectal 
ingredients. ``Gently apply to the affected area by patting and then 
discard.''
    (4) For products for intrarectal use that are wrapped suppositories. 
``Remove wrapper before inserting into the rectum.''
    (5) For products for intrarectal use that are to be used with a 
special applicator such as a pile pipe or other mechanical device. ``FOR 
INTRARECTAL USE: Attach applicator to tube. Lubricate applicator well, 
then gently insert applicator into the rectum.''
    (6) For products for external use only containing any of the local 
anesthetics identified in Sec. 346.10; analgesics, anesthetics, and 
antipruritics identified in Sec. 346.16; or alcloxa or resorcinol 
identified in Sec. 346.20. Apply to the affected area up to 6 times 
daily.
    (i) For products for external use only containing dibucaine or 
dibucaine hydrochloride identified in Sec. 346.10 (c) and (d). Apply to 
the affected area up to 3 or 4 times daily.
    (ii) For products for external use only containing pramoxine 
hydrochloride identified in Sec. 346.10(g). Apply to the affected area 
up to 5 times daily.
    (7) For products containing vasoconstrictors identified in 
Sec. 346.12. Apply to the affected area up to 4 times daily.
    (8) For products for external use only containing glycerin 
identified in Sec. 346.14(a)(3) or witch hazel identified in 
Sec. 346.18(b), and for products for external and/or intrarectal use 
containing any

[[Page 262]]

protectant identified in Sec. 346.14(a)(1), (2), (4), (5), (6), (7), and 
(9), and (b)(1), (2), (3), and (4), or any astringent identified in 
Sec. 346.18(a) and (c). Apply to the affected area up to 6 times daily 
or after each bowel movement.
    (9) For products containing petrolatum or white petrolatum 
identified in Sec. 346.14(a)(8) and (10). Apply liberally to the 
affected area as often as necessary.
    (e) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this section.
[55 FR 31779, Aug. 3, 1990, as amended at 59 FR 28767, June 3, 1994]



Sec. 346.52  Labeling of permitted combinations of anorectal active ingredients.

    Indications, warnings, and directions for use, respectively, 
applicable to each ingredient in the product may be combined to 
eliminate duplicative words or phrases so that the resulting information 
is clear and understandable.
    (a) Statement of identity. For a combination drug product that has 
an established name, the labeling of the product states the established 
name of the combination drug product, followed by the statement of 
identity established in Sec. 346.50(a). For a combination drug product 
that does not have an established name, the labeling of the product 
states the statement of identity established in Sec. 346.50(a).
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the indication(s) for each ingredient in the 
combination, as established in the indications sections of this subpart.
    (c) Warnings. The labeling of the product states, under the heading 
``Warnings,'' the warning(s) for each ingredient in the combination, as 
established in the warnings sections of this subpart.
    (d) Directions. The labeling of the product states, under the 
heading ``Directions,'' directions that conform to the directions 
established for each ingredient in the directions sections of this 
subpart. When the time intervals or age limitations for administration 
of the individual ingredients differ, the directions for the combination 
product may not exceed any maximum dosage limits established for the 
individual ingredients in the applicable OTC drug monograph.



PART 347--SKIN PROTECTANT DRUG PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                   Subpart A--Astringent Drug Products

Sec.
347.1  Scope.
347.3  Definitions.
347.10  Astringent active ingredients.
347.50  Labeling of astringent drug products.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  58 FR 54462, Oct. 21, 1993, unless otherwise noted.



                   Subpart A--Astringent Drug Products



Sec. 347.1  Scope.

    (a) An over-the-counter skin protectant drug product in a form 
suitable for topical administration is generally recognized as safe and 
effective and is not misbranded if it meets each condition in this part 
and each general condition established in Sec. 330.1 of this chapter.
    (b) References in this part to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 347.3  Definitions.

    As used in this part:
    (a) Astringent drug product means a drug product that is applied to 
the skin or mucous membranes for a local and limited protein coagulant 
effect.
    (b) [Reserved]



Sec. 347.10  Astringent active ingredients.

    The active ingredient of the product consists of any one of the 
following within the specified concentration established for each 
ingredient:
    (a) Aluminum acetate, 0.13 to 0.5 percent (depending on the 
formulation and concentration of the marketed product, the manufacturer 
must provide adequate directions so that the resulting solution to be 
used by the consumer contains 0.13 to 0.5 percent aluminum acetate).

[[Page 263]]

    (b) Aluminum sulfate, 46 to 63 percent (the concentration is based 
on the anhydrous equivalent).
    (c) Witch hazel.
[58 FR 54462, Oct. 21, 1993, as amended at 59 FR 28768, June 3, 1994]



Sec. 347.50  Labeling of astringent drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as an 
``astringent.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications'' any of the phrases listed in this paragraph (b), 
as appropriate. Other truthful and nonmisleading statements describing 
only the indications for use that have been established and listed in 
this paragraph (b) may also be used, as provided in Sec. 330.1(c)(2) of 
this chapter, subject to the provisions of section 502 of the Federal 
Food, Drug, and Cosmetic Act (the act) relating to misbranding and the 
prohibition in section 301(d) of the act against the introduction or 
delivery for introduction into interstate commerce of unapproved new 
drugs in violation of section 505(a) of the act.
    (1) For products containing aluminum acetate identified in 
Sec. 347.10(a). ``For temporary relief of minor skin irritations due 
to'' (select one or more of the following: ``poison ivy,'' ``poison 
oak,'' ``poison sumac,'' ``insect bites,'' ``athlete's foot,'' or 
``rashes caused by soaps, detergents, cosmetics, or jewelry'').
    (2) For products containing aluminum sulfate identified in 
Sec. 347.10(b) for use as a styptic pencil. ``Stops bleeding caused by 
minor surface cuts and abrasions as may occur during shaving.''
    (3) For products containing witch hazel identified in 
Sec. 347.10(c). (i) ``For relief of minor skin irritations due to'' 
(select one or more of the following: ``insect bites,'' ``minor cuts,'' 
or ``minor scrapes'').
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) ``For external use only. Avoid contact with the eyes.''
    (2) For products containing aluminum acetate identified in 
Sec. 347.10(a) or witch hazel identified in Sec. 347.10(c). ``If 
condition worsens or symptoms persist for more than 7 days, discontinue 
use of the product and consult a'' (select one of the following: 
``physician'' or ``doctor'').
    (3) For products containing aluminum acetate identified in 
Sec. 347.10(a) used as a compress or wet dressing. ``Do not cover 
compress or wet dressing with plastic to prevent evaporation.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) For products containing aluminum acetate identified in 
Sec. 347.10(a)--(i) For products used as a soak. ``For use as a soak: 
Soak affected area in the solution for 15 to 30 minutes. Discard 
solution after each use. Repeat 3 times a day.''
    (ii) For products used as a compress or wet dressing. ``For use as a 
compress or wet dressing: saturate a clean, soft, white cloth (such as a 
diaper or torn sheet) in the solution, gently squeeze, and apply loosely 
to the affected area. Saturate the cloth in the solution every 15 to 30 
minutes and apply to the affected area. Discard solution after each use. 
Repeat as often as necessary.''
    (2) For products containing aluminum sulfate identified in 
Sec. 347.10(b) for use as a styptic pencil. ``Moisten tip of pencil with 
water and apply to the affected area. Dry pencil after use.''
    (3) For products containing witch hazel identified in 
Sec. 347.10(c). ``Apply to the affected area as often as necessary.''
[58 FR 54462, Oct. 21, 1993, as amended at 59 FR 28768, June 3, 1994]



PART 348--EXTERNAL ANALGESIC DRUG PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                      Subpart A--General Provisions

Sec.
348.1  Scope.
348.3  Definitions.

                      Subpart B--Active Ingredients

348.10  Analgesic, anesthetic, and antipruritic active ingredients.

                           Subpart C--Labeling

348.50  Labeling of external analgesic drug

[[Page 264]]

          products.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  57 FR 27656, June 19, 1992, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 348.1  Scope.

    (a) An over-the-counter external analgesic drug product in a form 
suitable for topical administration is generally recognized as safe and 
effective and is not misbranded if it meets each condition in this part 
and each general condition established in Sec. 330.1 of this chapter.
    (b) References in this part to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 348.3  Definitions.

    As used in this part:
    (a) Male genital desensitizing drug product. A drug product applied 
to the penis to help in temporarily slowing the onset of ejaculation.
    (b) [Reserved]



                      Subpart B--Active Ingredients



Sec. 348.10  Analgesic, anesthetic, and antipruritic active ingredients.

    The active ingredient of the product consists of any of the 
following within the specified concentration established for each 
ingredient:
    (a) Male genital desensitizers. (1) Benzocaine, 3 to 7.5 percent in 
a water-soluble base.
    (2) Lidocaine in a metered spray with approximately 10 milligrams 
per spray.
    (b) [Reserved]



                           Subpart C--Labeling



Sec. 348.50  Labeling of external analgesic drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as 
follows:
    (1) For products containing any ingredient identified in 
Sec. 348.10(a). ``Male genital desensitizer.''
    (2) [Reserved]
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' any of the phrases listed in paragraph (b) of 
this section. Other truthful and nonmisleading statements, describing 
only the indications for use that have been established and listed in 
paragraph (b) of this section, may also be used, as provided in 
Sec. 330.1(c)(2) of this chapter, subject to the provisions of section 
502 of the Federal Food, Drug, and Cosmetic Act (the act) relating to 
misbranding and the prohibition in section 301(d) of the act against the 
introduction or delivery for introduction into interstate commerce of 
unapproved new drugs in violation of section 505(a) of the act.
    (1) For products containing any ingredient identified in 
Sec. 348.10(a). (i) ``Helps in the prevention of premature 
ejaculation.''
    (ii) ``For temporary male genital desensitization, helping to slow 
the onset of ejaculation.''
    (iii) ``Helps in temporarily'' (select one of the following: 
``retarding the onset of,'' ``slowing the onset of,'' or ``prolonging 
the time until'') followed by ``ejaculation.''
    (iv) ``For reducing oversensitivity in the male in advance of 
intercourse.''
    (2) [Reserved]
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) For products containing any ingredient identified in 
Sec. 348.10(a). (i) ``Premature ejaculation may be due to a condition 
requiring medical supervision. If this product, used as directed, does 
not provide relief, discontinue use and consult a doctor.''
    (ii) ``Avoid contact with the eyes.''
    (iii) ``If you or your partner develop a rash or irritation, such as 
burning or itching, discontinue use. If symptoms persist, consult a 
doctor.''
    (2) [Reserved]
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) For products containing any ingredient identified in 
Sec. 348.10(a)--(i) For products containing benzocaine identified in 
Sec. 348.10(a)(1). ``Apply a small amount to

[[Page 265]]

head and shaft of penis before intercourse, or use as directed by a 
doctor. Wash product off after intercourse.''
    (ii) For products containing lidocaine identified in 
Sec. 348.10(a)(2). ``Apply 3 or more sprays, not to exceed 10, to head 
and shaft of penis before intercourse, or use as directed by a doctor. 
Wash product off after intercourse.''
    (2) [Reserved]
    (e) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this section.



PART 349--OPHTHALMIC DRUG PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                      Subpart A--General Provisions

Sec.
349.1  Scope.
349.3  Definitions.

                      Subpart B--Active Ingredients

349.10  Ophthalmic astringent.
349.12  Ophthalmic demulcents.
349.14  Ophthalmic emollients.
349.16  Ophthalmic hypertonicity agent.
349.18  Ophthalmic vasoconstrictors.
349.20  Eyewashes.
349.30  Permitted combinations of active ingredients.

                           Subpart C--Labeling

349.50  Labeling of ophthalmic drug products.
349.55  Labeling of ophthalmic astringent drug products.
349.60  Labeling of ophthalmic demulcent drug products.
349.65  Labeling of ophthalmic emollient drug products.
349.70  Labeling of ophthalmic hypertonicity drug products.
349.75  Labeling of ophthalmic vasoconstrictor drug products.
349.78  Labeling of eyewash drug products.
349.79  Labeling of permitted combinations of active ingredients.
349.80  Professional labeling.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  53 FR 7090, Mar. 4, 1988, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 349.1  Scope.

    (a) An over-the-counter ophthalmic drug product in a form suitable 
for topical administration is generally recognized as safe and effective 
and is not misbranded if it meets each of the conditions in this part 
and each of the general conditions established in Sec. 330.1.
    (b) References in this part to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 349.3  Definitions.

    As used in this part:
    (a) Ophthalmic drug product. A drug product, which should be sterile 
in accordance with Sec. 200.50, to be applied to the eyelid or instilled 
in the eye.
    (b) Astringent. A locally acting pharmacologic agent which, by 
precipitating protein, helps to clear mucus from the outer surface of 
the eye.
    (c) Buffering agent. A substance which stabilizes the pH of 
solutions against changes produced by introduction of acids or bases 
from such sources as drugs, body fluids, tears, etc.
    (d) Demulcent. An agent, usually a water-soluble polymer, which is 
applied topically to the eye to protect and lubricate mucous membrane 
surfaces and relieve dryness and irritation.
    (e) Emollient. An agent, usually a fat or oil, which is applied 
locally to eyelids to protect or soften tissues and to prevent drying 
and cracking.
    (f) Eyewash, eye lotion, irrigating solution. A sterile aqueous 
solution intended for washing, bathing, or flushing the eye.
    (g) Hypertonicity agent. An agent which exerts an osmotic gradient 
greater than that present in body tissues and fluids, so that water is 
drawn from the body tissues and fluids across semipermeable membranes. 
Applied topically to the eye, a hypertonicity agent creates an osmotic 
gradient which draws water out of the cornea.
    (h) Isotonicity. A state or quality in which the osmotic pressure in 
two fluids is equal.
    (i) Vasoconstrictor. A pharmacologic agent which, when applied 
topically to the mucous membranes of the eye,

[[Page 266]]

causes transient constriction of conjunctival blood vessels.



                      Subpart B--Active Ingredients



Sec. 349.10  Ophthalmic astringent.

    The active ingredient and its concentration in the product is as 
follows: Zinc sulfate, 0.25 percent.



Sec. 349.12  Ophthalmic demulcents.

    The active ingredients of the product consist of any of the 
following, within the established concentrations for each ingredient:
    (a) Cellulose derivatives:
    (1) Carboxymethylcellulose sodium, 0.2 to 2.5 percent.
    (2) Hydroxyethyl cellulose, 0.2 to 2.5 percent.
    (3) Hydroxypropyl methylcellulose, 0.2 to 2.5 percent.
    (4) Methylcellulose, 0.2 to 2.5 percent.
    (b) Dextran 70, 0.1 percent when used with another polymeric 
demulcent agent in this section.
    (c) Gelatin, 0.01 percent.
    (d) Polyols, liquid:
    (1) Glycerin, 0.2 to 1 percent.
    (2) Polyethylene glycol 300, 0.2 to 1 percent.
    (3) Polyethylene glycol 400, 0.2 to 1 percent.
    (4) Polysorbate 80, 0.2 to 1 percent.
    (5) Propylene glycol, 0.2 to 1 percent.
    (e) Polyvinyl alcohol, 0.1 to 4 percent.
    (f) Povidone, 0.1 to 2 percent.



Sec. 349.14  Ophthalmic emollients.

    The active ingredients of the product consist of any of the 
following:
    (a) Lanolin preparations:
    (1) Anhydrous lanolin, 1 to 10 percent in combination with one or 
more oleaginous emollient agents included in the monograph.
    (2) Lanolin, 1 to 10 percent in combination with one or more 
oleaginous emollient agents included in the monograph.
    (b) Oleaginous ingredients:
    (1) Light mineral oil, up to 50 percent in combination with one or 
more other emollient agents included in the monograph.
    (2) Mineral oil, up to 50 percent in combination with one or more 
other emollient agents included in the monograph.
    (3) Paraffin, up to 5 percent in combination with one or more other 
emollient agents included in the monograph.
    (4) Petrolatum, up to 100 percent.
    (5) White ointment, up to 100 percent.
    (6) White petrolatum, up to 100 percent.
    (7) White wax, up to 5 percent in combination with one or more other 
emollient agents included in the monograph.
    (8) Yellow wax, up to 5 percent in combination with one or more 
other emollient agents included in the monograph.



Sec. 349.16  Ophthalmic hypertonicity agent.

    The active ingredient and its concentration in the product is as 
follows: Sodium chloride, 2 to 5 percent.



Sec. 349.18  Ophthalmic vasoconstrictors.

    The active ingredient of the product consists of one of the 
following, within the established concentration for each ingredient:
    (a) Ephedrine hydrochloride, 0.123 percent.
    (b) Naphazoline hydrochloride, 0.01 to 0.03 percent.
    (c) Phenylephrine hydrochloride, 0.08 to 0.2 percent.
    (d) Tetrahydrozoline hydrochloride, 0.01 to 0.05 percent.



Sec. 349.20  Eyewashes.

    These products contain water, tonicity agents to establish 
isotonicity with tears, agents for establishing pH and buffering to 
achieve the same pH as tears, and a suitable preservative agent.



Sec. 349.30  Permitted combinations of active ingredients.

    The following combinations are permitted provided each active 
ingredient is present within the established concentration, and the 
product is labeled in accordance with Sec. 349.79.
    (a) Any single ophthalmic astringent active ingredient identified in 
Sec. 349.10 may be combined with any single ophthalmic vasoconstrictor 
active ingredient identified in Sec. 349.18.

[[Page 267]]

    (b) Any two or three ophthalmic demulcent active ingredients 
identified in Sec. 349.12 may be combined.
    (c) Any single ophthalmic demulcent active ingredient identified in 
Sec. 349.12 or any ophthalmic demulcent combination identified in 
paragraph (b) of this section may be combined with any single ophthalmic 
vasoconstrictor identified in Sec. 349.18.
    (d) Any single ophthalmic astringent active ingredient identified in 
Sec. 349.10 may be combined with any single ophthalmic vasoconstrictor 
active ingredient identified in Sec. 349.18 and any single ophthalmic 
demulcent identified in Sec. 349.12 or ophthalmic demulcent combination 
identified in paragraph (b) of this section.
    (e) Any two or more emollient active ingredients identified in 
Sec. 349.14 may be combined as necessary to give the product proper 
consistency for application to the eye.



                           Subpart C--Labeling



Sec. 349.50  Labeling of ophthalmic drug products.

    (a) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this part.
    (b) Where applicable, indications in this part applicable to each 
ingredient in the product may be combined to eliminate duplicative words 
or phrases so that the resulting information is clear and 
understandable. Other truthful and nonmisleading statements, describing 
only the indications for use that have been established and listed in 
this part, may also be used, as provided in Sec. 330.1(c)(2), subject to 
the provisions of section 502 of the act relating to misbranding and the 
prohibition in section 301(d) of the act against the introduction or 
delivery for introduction into interstate commerce of unapproved new 
drugs in violation of section 505(a) of the act.
    (c) The labeling of the product contains the following warnings, 
under the heading ``Warnings'':
    (1) For ophthalmic drug products packaged in multi-use containers. 
``To avoid contamination, do not touch tip of container to any surface. 
Replace cap after using.''
    (2) For ophthalmic drug products packaged in single-use containers. 
``To avoid contamination, do not touch tip of container to any surface. 
Do not reuse. Once opened, discard.''
    (3) For ophthalmic drug products containing mercury compounds used 
as a preservative. ``This product contains (name and quantity of 
mercury-containing ingredient) as a preservative. Do not use this 
product if you are sensitive to'' (select one of the following: 
``mercury'' or ``(insert name of mercury-containing ingredient) or any 
other ingredient containing mercury).''



Sec. 349.55  Labeling of ophthalmic astringent drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as an 
``astringent'' (select one of the following: ``eye'' or ``ophthalmic'') 
``(insert dosage form, e.g., drops).''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the following phrase: ``For the temporary 
relief of discomfort from minor eye irritations.''
    (c) Warnings. In addition to the warnings in Sec. 349.50, the 
labeling of the product contains the following warnings under the 
heading ``Warnings'' for products containing any ingredient identified 
in Sec. 349.10:
    (1) ``If you experience eye pain, changes in vision, continued 
redness or irritation of the eye, or if the condition worsens or 
persists for more than 72 hours, discontinue use and consult a doctor.''
    (2) ``If solution changes color or becomes cloudy, do not use.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'': Instill 1 to 2 drops in 
the affected eye(s) up to four times daily.



Sec. 349.60  Labeling of ophthalmic demulcent drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug(s), if any, and identifies the product as a 
``lubricant'' or ``demulcent (lubricant)'' (select one of the following: 
``eye'' or ``ophthalmic'') ``(insert dosage form, e.g., drops).''

[[Page 268]]

    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' one or more of the following phrases:
    (1) ``For the temporary relief of burning and irritation due to 
dryness of the eye.''
    (2) ``For the temporary relief of discomfort due to minor 
irritations of the eye or to exposure to wind or sun.''
    (3) ``For use as a protectant against further irritation or to 
relieve dryness of the eye.''
    (4) ``For use as a lubricant to prevent further irritation or to 
relieve dryness of the eye.''
    (c) Warnings. In addition to the warnings in Sec. 349.50, the 
labeling of the product contains the following warnings under the 
heading ``Warnings'' for products containing any ingredient identified 
in Sec. 349.12:
    (1) ``If you experience eye pain, changes in vision, continued 
redness or irritation of the eye, or if the condition worsens or 
persists for more than 72 hours, discontinue use and consult a doctor.''
    (2) ``If solution changes color or becomes cloudy, do not use.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'': Instill 1 or 2 drops in 
the affected eye(s) as needed.



Sec. 349.65  Labeling of ophthalmic emollient drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug(s), if any, and identifies the product as a 
``lubricant'' or ``emollient (lubricant)'' (select one of the following: 
``eye'' or ``ophthalmic'') ``(insert dosage form, e.g., ointment).''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' one or more of the following phrases:
    (1) ``For the temporary relief of burning and irritation due to 
dryness of the eye.''
    (2) ``For the temporary relief of discomfort due to minor 
irritations of the eye or to exposure to wind or sun.''
    (3) ``For use as a protectant against further irritation or to 
relieve dryness of the eye.''
    (4) ``For use as a lubricant to prevent further irritation or to 
relieve dryness of the eye.''
    (c) Warnings. In addition to the warnings in Sec. 349.50, the 
labeling of the product contains the following warnings under the 
heading ``Warnings'' for products containing any ingredient identified 
in Sec. 349.14: ``If you experience eye pain, changes in vision, 
continued redness or irritation of the eye, or if the condition worsens 
or persists for more than 72 hours, discontinue use and consult a 
doctor.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'': Pull down the lower lid of 
the affected eye and apply a small amount (one-fourth inch) of ointment 
to the inside of the eyelid.



Sec. 349.70  Labeling of ophthalmic hypertonicity drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as a 
``hypertonicity'' (select one of the following: ``eye'' or 
``ophthalmic'') ``(insert dosage form, e.g., drops).''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the following phrase: ``For the temporary 
relief of corneal edema.''
    (c) Warnings. In addition to the warnings in Sec. 349.50, the 
labeling of the product contains the following warnings under the 
heading ``Warnings'' for products containing any ingredient identified 
in Sec. 349.16:
    (1) ``Do not use this product except under the advice and 
supervision of a doctor. If you experience eye pain, changes in vision, 
continued redness or irritation of the eye, or if the condition worsens 
or persists, consult a doctor.''
    (2) ``This product may cause temporary burning and irritation on 
being instilled into the eye.''
    (3) ``If solution changes color or becomes cloudy, do not use.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'': Instill 1 or 2 drops in 
the affected eye(s) every 3 or 4 hours, or as directed by a doctor.

[[Page 269]]



Sec. 349.75  Labeling of ophthalmic vasoconstrictor drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug(s), if any, and identifies the product as a 
``redness reliever'' or ``vasoconstrictor (redness reliever)'' (select 
one of the following: ``eye'' or ``ophthalmic'') ``(insert dosage form, 
e.g., drops).''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the following phrase: ``Relieves redness of the 
eye due to minor eye irritations.''
    (c) Warnings. In addition to the warnings in Sec. 349.50, the 
labeling of the product contains the following warnings under the 
heading ``Warnings'' for products containing any ingredient identified 
in Sec. 349.18:
    (1) ``If you experience eye pain, changes in vision, continued 
redness or irritation of the eye, or if the condition worsens or 
persists for more than 72 hours, discontinue use and consult a doctor.''
    (2) ``If you have glaucoma, do not use this product except under the 
advice and supervision of a doctor.''
    (3) ``Overuse of this product may produce increased redness of the 
eye.''
    (4) ``If solution changes color or becomes cloudy, do not use.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'': Instill 1 to 2 drops in 
the affected eye(s) up to four times daily.



Sec. 349.78  Labeling of eyewash drug products.

    (a) Statement of identity. The labeling of the product identifies 
the product with one or more of the following terms: ``eyewash,'' ``eye 
lotion,'' or ``eye irrigating solution.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' one of the following phrases:
    (1) ``For'' (select one of the following: ``flushing,'' 
``irrigating,'' ``cleansing,'' ``washing,'' or ``bathing'') ``the eye to 
remove'' (select one or more of the following: ``loose foreign 
material,'' ``air pollutants (smog or pollen),'' or ``chlorinated 
water'').
    (2) ``For'' (select one of the following: ``flushing,'' 
``irrigating,'' ``cleansing,'' ``washing,'' or ``bathing'') ``the eye to 
help relieve'' (select one or more of the following: ``irritation,'' 
``discomfort,'' ``burning,'' ``stinging,'' ``smarting,'' or ``itching'') 
``by removing'' (select one or more of the following: ``loose foreign 
material,'' ``air pollutants (smog or pollen),'' or ``chlorinated 
water'').
    (c) Warnings. In addition to the warnings in Sec. 349.50, the 
labeling of the product contains the following warnings under the 
heading ``Warnings'' for all eyewash products:
    (1) ``If you experience eye pain, changes in vision, continued 
redness or irritation of the eye, or if the condition worsens or 
persists, consult a doctor.''
    (2) ``Obtain immediate medical treatment for all open wounds in or 
near the eyes.''
    (3) ``If solution changes color or becomes cloudy, do not use.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) For eyewash products intended for use with an eyecup. Rinse cup 
with clean water immediately before each use. Avoid contamination of rim 
and inside surfaces of cup. Fill cup half full and apply the cup to the 
affected eye, pressing tightly to prevent the escape of the liquid, and 
tilt the head backward. Open eyelids wide and rotate eyeball to ensure 
thorough bathing with the wash or lotion. Rinse cup with clean water 
after each use.
    (2) For eyewash products intended for use with a nozzle applicator. 
Flush the affected eye as needed, controlling the rate of flow of 
solution by pressure on the bottle.



Sec. 349.79  Labeling of permitted combinations of active ingredients.

    Statements of identity, indications, warnings, and directions for 
use, respectively, applicable to each ingredient in the product may be 
combined to eliminate duplicative words or phrases so that the resulting 
information is clear and understandable.
    (a) Statement of identity. For a combination drug product that has 
an established name, the labeling of the product states the established 
name of

[[Page 270]]

the combination drug product, followed by the statement of identity for 
each ingredient in the combination, as established in the statement of 
identity sections of this part. For a combination drug product that does 
not have an established name, the labeling of the product states the 
statement of identity for each ingredient in the combination, as 
established in the statement of identity sections of this part.
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the indication(s) for each ingredient in the 
combination, as established in the indications sections of this part.
    (c) Warnings. The labeling of the product states, under the heading 
``Warnings,'' the warning(s) for each ingredient in the combination, as 
established in the warnings sections of this part.
    (d) Directions. The labeling of the product states, under the 
heading ``Directions,'' directions that conform to the directions 
established for each ingredient in the directions sections of this part. 
When the time intervals or age limitations for administration of the 
individual ingredients differ, the directions for the combination 
product may not exceed any maximum dosage limits established for the 
individual ingredients in the applicable OTC drug monograph.



Sec. 349.80  Professional labeling.

    The labeling of any OTC ophthalmic demulcent drug product provided 
to health professionals (but not to the general public) may contain 
instructions for the use of these products in professional eye 
examinations (i.e. gonioscopy, electroretinography).



PART 355--ANTICARIES DRUG PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                      Subpart A--General Provisions

Sec.
355.1  Scope.
355.3  Definitions.

                      Subpart B--Active Ingredients

355.10  Anticaries active ingredients.
355.20  Packaging conditions.

                           Subpart C--Labeling

355.50  Labeling of anticaries drug products.
355.55  Principal display panel of all fluoride rinse drug products.
335.60  Professional labeling.

                      Subpart D--Testing Procedures

355.70  Testing procedures for fluoride dentifrice drug products.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  60 FR 52507, Oct. 6, 1995, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 355.1  Scope.

    (a) An over-the-counter anticaries drug product in a form suitable 
for topical administration to the teeth is generally recognized as safe 
and effective and is not misbranded if it meets each condition in this 
part and each general condition established in Sec. 330.1 of this 
chapter.
    (b) References in this part to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 355.3  Definitions.

    As used in this part:
    (a) Abrasive. Solid materials that are added to dentifrices to 
facilitate mechanical removal of dental plaque, debris, and stain from 
tooth surfaces.
    (b) Anhydrous glycerin. An ingredient that may be prepared by 
heating glycerin U.S.P. at 150 -C for 2 hours to drive off the moisture 
content.
    (c) Anticaries drug. A drug that aids in the prevention and 
prophylactic treatment of dental cavities (decay, caries).
    (d) Dental caries. A disease of calcified tissues of teeth 
characterized by demineralization of the inorganic portion and 
destruction of the organic matrix.
    (e) Dentifrice. An abrasive-containing dosage form (gel, paste, or 
powder) for delivering an anticaries drug to the teeth.
    (f) Fluoride. The inorganic form of the chemical element fluorine in 
combination with other elements.

[[Page 271]]

    (g) Fluoride ion. The negatively charged atom of the chemical 
element fluorine.
    (h) Fluoride supplement. A special treatment rinse dosage form that 
is intended to be swallowed, and is promoted to health professionals for 
use in areas where the water supply contains 0 to 0.7 parts per million 
(ppm) fluoride ion.
    (i) Preventive treatment gel. A dosage form for delivering an 
anticaries drug to the teeth. Preventive treatment gels are formulated 
in an anhydrous glycerin base with suitable thickening agents included 
to adjust viscosity. Preventive treatment gels do not contain abrasives.
    (j) Treatment rinse. A liquid dosage form for delivering an 
anticaries drug to the teeth.
    (k) Treatment rinse concentrated solution. A fluoride treatment 
rinse in a concentrated form to be mixed with water before using to 
result in the appropriate fluoride concentration specified in the 
monograph.
    (l) Treatment rinse effervescent tablets. A fluoride treatment rinse 
prepared by adding an effervescent tablet (a concentrated solid dosage 
form) to water before using to result in the appropriate fluoride 
concentration specified in the monograph.
    (m) Treatment rinse powder. A fluoride treatment rinse prepared by 
adding the powder (a concentrated solid dosage form) to water before 
using to result in the appropriate fluoride concentration specified in 
the monograph.
[60 FR 52507, Oct. 6, 1995, as amended at 61 FR 52286, Oct. 7, 1996]



                      Subpart B--Active Ingredients



Sec. 355.10  Anticaries active ingredients.

    The active ingredient of the product consists of any of the 
following when used in the concentration and dosage form established for 
each ingredient:
    (a) Sodium fluoride--(1) Dentifrices containing 850 to 1,150 ppm 
theoretical total fluorine in a gel or paste dosage form. Sodium 
fluoride 0.188 to 0.254 percent with an available fluoride ion 
concentration  650 parts per million (ppm).
    (2) Dentifrices containing 850 to 1,150 ppm theoretical total 
fluorine in a powdered dosage form. Sodium fluoride 0.188 to 0.254 
percent with an available fluoride ion concentration of  850 
ppm for products containing the abrasive sodium bicarbonate and a 
poured-bulk density of 1.0 to 1.2 grams per milliliter.
    (3) Treatment rinses. (i) An aqueous solution of acidulated 
phosphate fluoride derived from sodium fluoride acidulated with a 
mixture of sodium phosphate, monobasic, and phosphoric acid to a level 
of 0.1 molar phosphate ion and a pH of 3.0 to 4.5 and which yields an 
effective fluoride ion concentration of 0.02 percent.
    (ii) An aqueous solution of acidulated phosphate fluoride derived 
from sodium fluoride acidulated with a mixture of sodium phosphate, 
dibasic, and phosphoric acid to a pH of 3.5 and which yields an 
effective fluoride ion concentration of 0.01 percent.
    (iii) Sodium fluoride 0.02 percent aqueous solution with a pH of 
approximately 7.
    (iv) Sodium fluoride 0.05 percent aqueous solution with a pH of 
approximately 7.
    (v) Sodium fluoride concentrate containing adequate directions for 
mixing with water before using to result in a 0.02-percent or 0.05-
percent aqueous solution with a pH of approximately 7.
    (b) Sodium monofluorophosphate--(1) Dentifrices containing 850 to 
1,150 ppm theoretical total fluorine in a gel or paste dosage form. 
Sodium monofluorophosphate 0.654 to 0.884 percent with an available 
fluoride ion concentration (consisting of PO3 F= 
and F- combined)  800 ppm.
    (2) Dentifrices containing 1,500 ppm theoretical total fluorine in a 
gel or paste dosage form. Sodium monofluorophosphate 1.153 percent with 
an available fluoride ion concentration (consisting of PO3 
F= and F- combined)  1,275 ppm.
    (c) Stannous fluoride--(1) Dentifrices containing 850 to 1,150 ppm 
theoretical total fluorine in a gel or paste dosage form. (i) Stannous 
fluoride 0.351 to 0.474 percent with an available fluoride ion 
concentration  700 ppm for products containing abrasives 
other than calcium pyrophosphate.
    (ii) Stannous fluoride 0.351 to 0.474 percent with an available 
fluoride ion concentration  290 ppm for products

[[Page 272]]

containing the abrasive calcium pyrophosphate.
    (2) Preventive treatment gel. Stannous fluoride 0.4 percent in an 
anhydrous glycerin gel, made from anhydrous glycerin and the addition of 
suitable thickening agents to adjust viscosity.
    (3) Treatment rinse. Stannous fluoride concentrate marketed in a 
stable form and containing adequate directions for mixing with water 
immediately before using to result in a 0.1-percent aqueous solution.
[60 FR 52507, Oct. 6, 1995, as amended at 61 FR 52286, Oct. 7, 1996]



Sec. 355.20  Packaging conditions.

    (a) Package size limitation. Due to the toxicity associated with 
fluoride active ingredients, the following package size limitations are 
required for anticaries drug products:
    (1) Dentifrices. Dentifrice (toothpastes and tooth powders) packages 
shall not contain more than 276 milligrams (mg) total fluorine per 
package.
    (2) Preventive treatment gels and treatment rinses. Preventive 
treatment gel and treatment rinse packages shall not contain more than 
120 mg total fluorine per package.
    (3) Exception. Package size limitations do not apply to anticaries 
drug products marketed for professional office use only and labeled in 
accord with Sec. 355.60.
    (b) Tight container packaging. To minimize moisture contamination, 
all fluoride powdered dentifrices shall be packaged in a tight container 
as defined as a container that protects the contents from contamination 
by extraneous liquids, solids, or vapors, from loss of the article, and 
from efflorescence, deliquescence, or evaporation under the ordinary or 
customary conditions of handling, shipment, storage, and distribution, 
and is capable of tight reclosure.



                           Subpart C--Labeling



Sec. 355.50  Labeling of anticaries drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as: 
(select one or both of the following: `anticavity' or `fluoride') 
(select one of the following as appropriate: ``dentifrice,'' 
``toothpaste,'' ``tooth polish,'' ``tooth powder;'' (optional: 
``dental'') ``preventive treatment gel;'' or (optional: ``treatment'' or 
``dental'')) (select one of the following: ``rinse,'' ``concentrated 
solution,'' ``rinse powder,'' or ``rinse effervescent tablets''). The 
word ``mouthwash'' may be substituted for the word ``rinse'' in this 
statement of identity if the product also has a cosmetic use, as defined 
in section 201(i) of the Federal Food, Drug, and Cosmetic Act (the act) 
(21 U.S.C. 321(i)).
    (b) Indication. The labeling of the product states, under the 
heading ``Indication,'' the following: ``Aids in the prevention of 
dental (select one of the following: ``cavities,'' ``decay,'' ``caries 
(decay),'' or ``caries (cavities)''). Other truthful and nonmisleading 
statements, describing only the indication for use that has been 
established and listed in this paragraph (b), may also be used, as 
provided in Sec. 330.1(c)(2) of this chapter, subject to the provisions 
of section 502 of the Federal Food, Drug, and Cosmetic Act (the act) 
relating to misbranding and the prohibition in section 301(d) of the act 
against the introduction or delivery for introduction into interstate 
commerce of unapproved new drugs in violation of section 505(a) of the 
act.
    (c) Warning. The labeling of the product contains the following 
warning under the heading ``Warning'':
    (1) For all fluoride dentifrice (gel, paste, and powder) products. 
``Keep out of the reach of children under 6 years of age. If you 
accidentally swallow more than used for brushing, seek professional 
assistance or contact a Poison Control Center immediately.'' These 
warnings shall be used in place of the general warning statements 
required by Sec. 330.1(g) of this chapter.
    (2) For all fluoride rinse and preventive treatment gel products. 
``Keep this and all drugs out of the reach of children. If you 
accidentally swallow more than used for'' (select appropriate word: 
``brushing'' or ``rinsing''), ``seek professional assistance or contact 
a Poison Control Center immediately.'' These warnings shall be used in 
place of the general warning statements required by Sec. 330.1(g) of 
this chapter.

[[Page 273]]

    (d) Directions. The labeling of the product contains the following 
statements under the heading ``Directions'':
    (1) For anticaries dentifrice products--(i) Gel or paste dosage form 
with a theoretical total fluorine concentration of 850 to 1,150 ppm 
identified in Sec. 355.10(a)(1), (b)(1), and (c)(1). Adults and children 
2 years of age and older: Brush teeth thoroughly, preferably after each 
meal or at least twice a day, or as directed by a dentist or doctor. 
Instruct children under 6 years of age in good brushing and rinsing 
habits (to minimize swallowing). Supervise children as necessary until 
capable of using without supervision. Children under 2 years of age: 
Consult a dentist or doctor.
    (ii) Gel or paste dosage form with a theoretical total fluorine 
concentration of 1,500 ppm identified in Sec. 355.10(b)(2). Adults and 
children 6 years of age and older: Brush teeth thoroughly, preferably 
after each meal or at least twice a day, or as directed by a dentist or 
doctor. Instruct children under 12 years of age in good brushing and 
rinsing habits (to minimize swallowing). Supervise children as necessary 
until capable of using without supervision. Children under 6 years of 
age: Do not use unless directed by a dentist or doctor.
    (iii) Powdered dosage form with a theoretical total fluorine 
concentration of 850 to 1,150 ppm identified in Sec. 355.10(a)(2). 
Adults and children 6 years of age and older: Apply powder to a wet 
toothbrush; completely cover all bristles. Brush for at least 30 
seconds. Reapply powder as before and brush again. Rinse and spit out 
thoroughly. Brush teeth, preferably after each meal or at least twice a 
day, or as directed by a dentist or doctor. Instruct children under 12 
years of age in good brushing and rinsing habits (to minimize 
swallowing). Supervise children as necessary until capable of using 
without supervision. Children under 6 years of age: Do not use unless 
directed by a dentist or doctor.
    (2) For anticaries treatment rinse products--(i) For acidulated 
phosphate fluoride solution containing 0.02 percent fluoride ion, sodium 
fluoride 0.05 percent, sodium fluoride concentrate, and stannous 
fluoride concentrate identified in Sec. 355.10(a)(3)(i), (a)(3)(iv), 
(a)(3)(v), and (c)(3). Adults and children 6 years of age and older: Use 
once a day after brushing your teeth with a toothpaste. Vigorously swish 
10 milliliters of rinse between your teeth for 1 minute and then spit 
out. Do not swallow the rinse. Do not eat or drink for 30 minutes after 
rinsing. Instruct children under 12 years of age in good rinsing habits 
(to minimize swallowing). Supervise children as necessary until capable 
of using without supervision. Children under 6 years of age: Consult a 
dentist or doctor.
    (ii) For acidulated phosphate fluoride solution containing 0.01 
percent fluoride ion and sodium fluoride 0.02 percent aqueous solution 
identified in Sec. 355.10(a)(3)(ii) and (a)(3)(iii). Adults and children 
6 years of age and older: Use twice a day after brushing your teeth with 
a toothpaste. Vigorously swish 10 milliliters of rinse between your 
teeth for 1 minute and then spit out. Do not swallow the rinse. Do not 
eat or drink for 30 minutes after rinsing. Instruct children under 12 
years of age in good rinsing habits (to minimize swallowing). Supervise 
children as necessary until capable of using without supervision. 
Children under 6 years of age: consult a dentist or doctor.
    (3) For stannous fluoride treatment rinse products. (i) ``Use 
immediately after preparing the rinse.''
    (ii) For powder or effervescent tablets used to prepare treatment 
rinses. ``Do not use as a rinse until all the'' (select one of the 
following: ``powder'' or ``tablet'') ``has dissolved.''
    (4) For anticaries preventive treatment gel products. Adults and 
children 6 years of age and older: Use once a day after brushing your 
teeth with a toothpaste. Apply the gel to your teeth and brush 
thoroughly. Allow the gel to remain on your teeth for 1 minute and then 
spit out. Do not swallow the gel. Do not eat or drink for 30 minutes 
after brushing. Instruct children under 12 years of age in the use of 
this product (to minimize swallowing). Supervise children as necessary 
until capable of using without supervision. Children under 6 years of 
age: consult a dentist or doctor.

[[Page 274]]

    (5) For all concentrated treatment rinse solutions, powders, and 
effervescent tablets. The following statement shall appear as the first 
statement under directions: ``Do not use before mixing with water.''
    (e) Additional labeling statements for anticaries drug products. The 
following statements need not appear under warnings, but are required to 
appear on the label of anticaries drugs products as applicable.
    (1) For all preventive treatment gels. ``This is a(n)'' (select one 
or both of the following: ``anticavity'' or ``fluoride'') ``preventive 
treatment gel, not a toothpaste. Read directions carefully before 
using.''
    (2) For all stannous fluoride treatment rinse, preventive treatment 
gel, and dentifrice products. ``This product may produce surface 
staining of the teeth. Adequate toothbrushing may prevent these stains 
which are not harmful or permanent and may be removed by your dentist.''
    (f) Optional additional labeling statements--(1) For fluoride 
treatment rinses and preventive treatment gels. The following labeling 
statement may appear in the required boxed area designated ``APPROVED 
USES'': ``The combined daily use of a fluoride preventive treatment'' 
(select one of the following: ``rinse'' or ``gel'') ``and a fluoride 
toothpaste can help reduce the incidence of dental cavities.''
    (2) For dentifrice products containing 1,500 ppm theoretical total 
fluorine. ``Adults and children over 6 years of age may wish to use this 
extra-strength fluoride dentifrice if they reside in a nonfluoridated 
area or if they have a greater tendency to develop cavities.''
[60 FR 52507, Oct. 6, 1995; 60 FR 57927, Nov. 24, 1995; 61 FR 51187, 
Oct. 7, 1996]



Sec. 355.55  Principal display panel of all fluoride rinse drug products.

    In addition to the statement of identity required in Sec. 355.50, 
the following statement shall be prominently placed on the principal 
display panel: ``IMPORTANT: Read directions for proper use.''



Sec. 355.60  Professional labeling.

    (a) The labeling for anticaries fluoride treatment rinses identified 
in Sec. 355.10(a)(3) and (c)(3) that are specially formulated so they 
may be swallowed (fluoride supplements) and are provided to health 
professionals (but not to the general public) may contain the following 
additional dosage information: Children 3 to under 14 years of age: As a 
supplement in areas where the water supply is nonfluoridated (less than 
0.3 parts per million (ppm)), clean the teeth with a toothpaste and 
rinse with 5 milliliters (mL) of 0.02 percent or 10 mL of 0.01 percent 
fluoride ion rinse daily, then swallow. When the water supply contains 
0.3 to 0.7 ppm fluoride ion, reduce the dose to 2.5 mL of 0.02 percent 
or 5 mL of 0.01 percent fluoride ion rinse daily.
    (b) The labeling for products marketed to health to health 
professionals in package sizes larger than those specified in 
Sec. 355.20 shall include the statements: ``For Professional Office Use 
Only'' and ``This product is not intended for home or unsupervised 
consumer use.''



                      Subpart D--Testing Procedures



Sec. 355.70  Testing procedures for fluoride dentifrice drug products.

    (a) A fluoride dentifrice drug product shall meet the biological 
test requirements for animal caries reduction and one of the following 
tests: Enamel solubility reduction or fluoride enamel uptake. The 
testing procedures for these biological tests are labeled Biological 
Testing Procedures for Fluoride Dentifrices; these testing procedures 
are on file under Docket No. 80N-0042 in the Dockets Management Branch 
(HFA-305), Food and Drug Administration, rm. 1-23, 12420 Parklawn Dr., 
Rockville, MD 20857, and are available on request to that office.
    (b) The United States Pharmacopeia fluoride dentifrice reference 
standards along with reference standard stability profiles (total 
fluoride, available fluoride ion, pH, and specific gravity) required to 
be used in the biological tests are available to any purchaser upon 
written request to the United States Pharmacopeial Convention, Inc., 
1260

[[Page 275]]

Twinbrook Parkway, Rockville, MD 20852.
    (c) Alternative testing procedures may be used. Any proposed 
modification or alternative testing procedures shall be submitted as a 
petition in accord with Sec. 10.30 of this chapter. The petition should 
contain data to support the modification or data demonstrating that an 
alternative testing procedure provides results of equivalent accuracy. 
All information submitted will be subjected to the disclosure rules in 
part 20 of this chapter.



PART 357--MISCELLANEOUS INTERNAL DRUG PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                          Subpart A  [Reserved]

                  Subpart B--Anthelmintic Drug Products

Sec.
357.101  Scope.
357.103  Definition.
357.110  Anthelmintic active ingredient.
357.150  Labeling of anthelmintic drug products.
357.152  Package inserts for anthelmintic drug products.
357.180  Professional labeling.

               Subpart C--Cholecystokinetic Drug Products

357.201  Scope.
357.203  Definition.
357.210  Cholecystokinetic active ingredients.
357.250  Labeling of cholecystokinetic drug products.
357.280  Professional labeling.

                        Subparts D-H  [Reserved]

           Subpart I--Deodorant Drug Products for Internal Use

357.801  Scope.
357.803  Definitions.
357.810  Active ingredients for deodorant drug products for internal 
          use.
357.850  Labeling of deodorant drug products for internal use.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.



                          Subpart A  [Reserved]



                  Subpart B--Anthelmintic Drug Products

    Source:  51 FR 27759, Aug. 1, 1986, unless otherwise noted.



Sec. 357.101  Scope.

    (a) An over-the-counter anthelmintic drug product in a form suitable 
for oral administration is generally recognized as safe and effective 
and is not misbranded if it meets each condition in this subpart and 
each general condition established in Sec. 330.1.
    (b) References in this subpart to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 357.103  Definition.

    As used in this subpart:
    Anthelmintic. An agent that is destructive to worms.



Sec. 357.110  Anthelmintic active ingredient.

    The active ingredient of the product is pyrantel pamoate when used 
within the dosage limits established in Sec. 357.150(d)(1).



Sec. 357.150  Labeling of anthelmintic drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as a 
``pinworm treatment.''
    (b) Indication. The labeling of the product states, under the 
heading ``Indication,'' the following: ``For the treatment of 
pinworms.'' Other truthful and nonmisleading statements, describing only 
the indications for use that have been established and listed in this 
paragraph (b), may also be used, as provided in Sec. 330.1(c)(2), 
subject to the provisions of section 502 of the act relating to 
misbranding and the prohibition in section 301(d) of the act against the 
introduction or delivery for introduction into interstate commerce of 
unapproved new drugs in violation of section 505(a) of the act.

[[Page 276]]

    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) ``Abdominal cramps, nausea, vomiting, diarrhea, headache, or 
dizziness sometimes occur after taking this drug. If any of these 
conditions persist consult a doctor.''
    (2) ``If you are pregnant or have liver disease, do not take this 
product unless directed by a doctor.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) Adults, children 12 years of age and over, and children 2 years 
to under 12 years of age: Oral dosage is a single dose of 5 milligrams 
of pyrantel base per pound, or 11 milligrams per kilogram, of body 
weight not to exceed 1 gram. Dosing information should be converted to 
easily understood directions for the consumer using the following dosage 
schedule:

                                                                        
------------------------------------------------------------------------
                                              Dosage (taken as a single 
                  Weight                              dose) \1\         
------------------------------------------------------------------------
Less than 25 pounds or under 2 years old..  Do not use unless directed  
                                             by a doctor.               
 25 to 37 pounds..........................  125 milligrams.             
 38 to 62 pounds..........................  250 milligrams.             
 63 to 87 pounds..........................  375 milligrams.             
 88 to 112 pounds.........................  500 milligrams.             
113 to 137 pounds.........................  625 milligrams.             
138 to 162 pounds.........................  750 milligrams.             
163 to 187 pounds.........................  875 milligrams.             
188 pounds and over.......................  1,000 milligrams.           
------------------------------------------------------------------------
\1\ Depending on the product, the label should state the quantity of    
  drug as a liquid measurement (e.g., teaspoonsful) or as the number of 
  dosage units (e.g., tablets) to be taken for the varying body weights.
  (If appropriate, it is recommended that a measuring cup graduated by  
  body weight and/or liquid measurement be provided with the product.)  
  Manufacturers should present this information as appropriate for their
  product and may vary the format of this chart as necessary.           

    (2) ``Read package insert carefully before taking this medication. 
Take only according to directions and do not exceed the recommended 
dosage unless directed by a doctor. Medication should only be taken on 
time as a single dose; do not repeat treatment unless directed by a 
doctor. When one individual in a household has pinworms, the entire 
household should be treated unless otherwise advised. See Warnings. If 
any worms other than pinworms are present before or after treatment, 
consult a doctor. If any symptoms or pinworms are still present after 
treatment, consult a doctor.
    (3) ``This product can be taken any time of day, with or without 
meals. It may be taken alone or with milk or fruit juice. Use of a 
laxative is not necessary prior to, during, or after medication.''
    (e) Optional wording. The word ``physician'' may be substituted for 
the word ``doctor'' in any of the labeling statements in this section.
[51 FR 27759, Aug. 1, 1986; 52 FR 7831, Mar. 13, 1987, as amended at 53 
FR 35810, Sept. 15, 1988]



Sec. 357.152  Package inserts for anthelmintic drug products.

    The labeling of the product contains a consumer package insert which 
includes the following information:
    (a) A discussion of the symptoms suggestive of pinworm infestation, 
including a statement that pinworms must be visually identified before 
taking this medication.
    (b) A detailed description of how to find and identify the pinworm.
    (c) A commentary on the life cycle of the pinworm.
    (d) A commentary on the ways in which pinworms may be spread from 
person to person and hygienic procedures to follow to avoid such 
spreading.
    (e) The appropriate labeling information contained in Sec. 357.150

(Collection of information requirement approved by the Office of 
Management and Budget under control number 0910-0232)

[51 FR 27759, Aug. 1, 1986, as amended at 52 FR 2515, Jan 23, 1987]



Sec. 357.180  Professional labeling.

    The labeling provided to health professionals (but not to the 
general public) may contain an additional indication: ``For the 
treatment of common roundworm infestation.''



               Subpart C--Cholecystokinetic Drug Products



Sec. 357.201  Scope.

    (a) An over-the-counter cholecystokinetic drug product in a form 
suitable for oral administration is generally recognized as safe and 
effective and is not misbranded if it meets each of the conditions in 
this subpart

[[Page 277]]

in addition to each of the general conditions established in Sec. 330.1.
    (b) References in this subpart to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.
[48 FR 27005, June 10, 1983]



Sec. 357.203  Definition.

    As used in this subpart:
    Cholecystokinetic drug product. A drug product that causes 
contraction of the gallbladder and is used during the course of 
diagnostic gallbladder studies (cholecystography).
[48 FR 27005, June 10, 1983]



Sec. 357.210  Cholecystokinetic active ingredients.

    The active ingredient of the product consists of any of the 
following when used within the specified concentration and dosage form 
established for each ingredient:
    (a) 50-percent aqueous emulsion of corn oil.
    (b) Hydrogenated soybean oil in a suitable, water-dispersible 
powder. The hydrogenated soybean oil is food-grade, partially 
hydrogenated with a melting point of 41 to 43.5  deg. C, an iodine value 
of 65 to 69, and a fatty acid composition as follows:

                                                                        
------------------------------------------------------------------------
                                                               Percent  
                         Fatty acid                          composition
------------------------------------------------------------------------
Myristic acid..............................................         0.1 
Palmitic acid..............................................        10.0 
Palmitoleic acid...........................................         0.1 
Stearic acid...............................................        13.5 
Oleic acid.................................................        72.0 
Linoleic acid..............................................         3.8 
Linolenic acid.............................................         0.1 
Arachidic acid.............................................         0.5 
Behenic acid...............................................         0.2 
------------------------------------------------------------------------

[54 FR 8321, Feb. 28, 1989]



Sec. 357.250  Labeling of cholecystokinetic drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as a 
``gallbladder diagnostic agent.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the following: ``For the contraction of the 
gallbladder during diagnostic gallbladder studies.'' Other truthful and 
nonmisleading statements, describing only the indications for use that 
have been established and listed in this paragraph (b), may also be 
used, as provided in Sec. 330.1(c)(2), subject to the provisions of 
section 502 of the act relating to misbranding and the prohibition in 
section 301(d) of the act against the introduction or delivery for 
introduction into interstate commerce of unapproved new drugs in 
violation of section 505(a) of the act.
    (c) Warnings. [Reserved]
    (d) Directions. The labeling of the product contains the following 
statements under the heading ``Directions'':
    (1) ``Take only when instructed by a doctor:''
    (2) For products containing 50-percent aqueous emulsion of corn oil.
    (i) ``Shake well before using.''
    (ii) Oral dosage is 60 milliliters 20 minutes before diagnostic 
gallbladder x-ray or as directed by a doctor.
    (3) For products containing hydrogeneated soybean oil. Oral dosage 
is 12.4 grams in a suitable, water-dispersible powder in 2 to 3 ounces 
of water. Stir briskly to prepare a suspension before using. Drink 20 
minutes before diagnostic gallbladder x-ray or as directed by a doctor.
    (e) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this section.
[48 FR 27005, June 10, 1983, as amended at 51 FR 16267, May 1, 1986; 52 
FR 7830, Mar. 13, 1987; 54 FR 8321, Feb. 28, 1989]



Sec. 357.280  Professional labeling.

    The labeling provided to health professionals (but not to the 
general public) may contain the following information for ingredients 
identified in Sec. 357.210: Indication. ``For visualization of biliary 
ducts during cholecystography.''
[54 FR 8321, Feb. 28, 1989]



                         Subparts D-H [Reserved]



           Subpart I--Deodorant Drug Products for Internal Use

    Source:  55 FR 19865, May 11, 1990, unless otherwise noted.

[[Page 278]]



Sec. 357.801  Scope.

    (a) An over-the-counter deodorant drug product for internal use in a 
form suitable for oral administration is generally recognized as safe 
and effective and is not misbranded if it meets each condition in this 
subpart and each general condition established in Sec. 330.1 of this 
chapter.
    (b) References in this subpart to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 357.803  Definitions.

    As used in this subpart:
    (a) Colostomy. An external operative opening of the colon.
    (b) Deodorant for internal use. An ingredient taken internally to 
reduce odors arising from conditions such as colostomies, ileostomies, 
or fecal incontinence.
    (c) Ileostomy. An external operative opening from the ileum.
    (d) Incontinence. An inability to retain urine or feces.



Sec. 357.810  Active ingredients for deodorant drug products for internal use.

    The active ingredient of the product consists of either of the 
following when used within the dosage limits established for each 
ingredient in Sec. 357.850(d):
    (a) Bismuth subgallate.
    (b) Chlorophyllin copper complex.



Sec. 357.850  Labeling of deodorant drug products for internal use.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as a 
``deodorant for internal use'' or as a ``colostomy or ileostomy 
deodorant.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' any of the phrases listed in paragraph (b) of 
this section as appropriate. Other truthful and nonmisleading 
statements, describing only the indications for use that have been 
established and listed in paragraph (b) of this section may also be 
used, as provided in Sec. 330.1(c)(2) of this chapter, subject to the 
provisions of section 502 of the Federal Food, Drug, and Cosmetic Act 
(the act) relating to misbranding and the prohibition in section 301(d) 
of the act against the introduction or delivery for introduction into 
interstate commerce of unapproved new drugs in violation of section 
505(a) of the act.
    (1) For products containing bismuth subgallate identified in 
Sec. 357.810(a). ``An aid to reduce odor from a colostomy or 
ileostomy.''
    (2) For products containing chlorophyllin copper complex identified 
in Sec. 357.810(b). (i) ``An aid to reduce odor from a colostomy or 
ileostomy.''
    (ii) ``An aid to reduce fecal odor due to incontinence.''
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'': (1) For products containing 
chlorophyllin copper complex identified in Sec. 357.810(b). (i) ``If 
cramps or diarrhea occurs, reduce the dosage. If symptoms persist, 
consult your doctor.''
    (ii) The warning required by Sec. 330.1(g) of this chapter 
concerning overdose is not required on products containing chlorophyllin 
copper complex identified in Sec. 357.810(b).
    (2) [Reserved]
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions.''
    (1) For products containing bismuth subgallate identified in 
Sec. 357.810(a). Adults and children 12 years of age and over: Oral 
dosage is 200 to 400 milligrams up to 4 times daily. Children under 12 
years of age: consult a doctor.
    (2) For products containing chlorophyllin copper complex identified 
in Sec. 357.810(b). Adults and children 12 years of age and over: Oral 
dosage is 100 to 200 milligrams daily in divided doses as required. If 
odor is not controlled, take up to an additional 100 milligrams daily in 
divided doses as required. The smallest effective dose should be used. 
Do not exceed 300 milligrams daily. Children under 12 years of age: 
consult a doctor.

[[Page 279]]



PART 358--MISCELLANEOUS EXTERNAL DRUG PRODUCTS FOR OVER-THE-COUNTER HUMAN USE--Table of Contents




                          Subpart A  [Reserved]

                  Subpart B--Wart Remover Drug Products

Sec.
358.101  Scope.
358.103  Definitions.
358.110  Wart remover active ingredients.
358.150  Labeling of wart remover drug products.

                        Subparts C-E  [Reserved]

            Subpart F--Corn and Callus Remover Drug Products

358.501  Scope.
358.503  Definitions.
358.510  Corn and callus remover active ingredients.
358.550  Labeling of corn and callus remover drug products.

                  Subpart G--Pediculicide Drug Products

358.601  Scope.
358.603  Definition.
358.610  Pediculicide active ingredients.
358.650  Labeling of pediculicide drug products.

    Subpart H--Drug Products for the Control of Dandruff, Seborrheic 
                        Dermatitis, and Psoriasis

358.701  Scope.
358.703  Definitions.
358.710  Active ingredients for the control of dandruff, seborrheic 
          dermatitis, or psoriasis.
358.720  Permitted combinations of active ingredients.
358.750  Labeling of drug products for the control of dandruff, 
          seborrheic dermatitis, or psoriasis.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 360, 371.

    Source:  55 FR 33255, Aug. 14, 1990, unless otherwise noted.



                          Subpart A  [Reserved]



                  Subpart B--Wart Remover Drug Products



Sec. 358.101  Scope.

    (a) An over-the-counter wart remover drug product in a form suitable 
for topical application is generally recognized as safe and effective 
and is not misbranded if it meets each of the conditions in this subpart 
and each of the general conditions established in Sec. 330.1 of this 
chapter.
    (b) References in this subpart to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 358.103  Definitions.

    As used in this subpart:
    (a) Wart remover drug product. A topical agent used for the removal 
of common or plantar warts.
    (b) Collodion-like vehicle. A solution containing pyroxylin 
(nitrocellulose) in an appropriate nonaqueous solvent that leaves a 
transparent cohesive film when applied to the skin in a thin layer.
    (c) Plaster vehicle. A fabric, plastic, or other suitable backing 
material in which medication is usually incorporated for topical 
application to the skin.



Sec. 358.110  Wart remover active ingredients.

    The product consists of any of the following active ingredients 
within the specified concentration and in the dosage form established 
for each ingredient.
    (a) Salicylic acid 12 to 40 percent in a plaster vehicle.
    (b) Salicylic acid 5 to 17 percent in a collodion-like vehicle.
    (c) Salicylic acid 15 percent in a karaya gum, glycol plaster 
vehicle.



Sec. 358.150  Labeling of wart remover drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as a 
``wart remover.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' any of the phrases listed in paragraph (b) of 
this section. Other truthful and nonmisleading statements, describing 
only the indications for use that have been established in paragraph (b) 
of this section, may also be used, as provided in Sec. 330.1(c)(2) of 
this chapter, subject to the provisions of section 502 of the Federal 
Food,

[[Page 280]]

Drug, and Cosmetic Act (the act) relating to misbranding and the 
prohibition in section 301(d) of the act against the introduction or 
delivery for introduction into interstate commerce of unapproved new 
drugs in violation of section 505(a) of the act.
    (1) ``For the removal of common warts. The common wart is easily 
recognized by the rough `cauliflower-like' appearance of the surface.''
    (2) ``For the removal of plantar warts on the bottom of the foot. 
The plantar wart is recognized by its location only on the bottom of the 
foot, its tenderness, and the interruption of the footprint pattern.''
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) For products containing any ingredient identified in 
Sec. 358.110. (i) ``For external use only.''
    (ii) ``Do not use this product on irritated skin, on any area that 
is infected or reddened, if you are a diabetic, or if you have poor 
blood circulation.''
    (iii) ``If discomfort persists, see your doctor.''
    (iv) ``Do not use on moles, birthmarks, warts with hair growing from 
them, genital warts, or warts on the face or mucous membranes.''
    (2) For any product formulated in a flammable vehicle. (i) The 
labeling should contain an appropriate flammability signal word, e.g. 
``extremely flammable,'' ``flammable,'' ``combustible,'' consistent with 
16 CFR 1500.3(b)(10).
    (ii) ``Keep away from fire or flame.''
    (3) For any product formulated in a volatile vehicle. ``Cap bottle 
tightly and store at room temperature away from heat.''
    (4) For any product formulated in a collodion-like vehicle. (i) ``If 
product gets into the eye, flush with water for 15 minutes.''
    (ii) ``Avoid inhaling vapors.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) For products containing salicylic acid identified in 
Sec. 358.110(a). ``Wash affected area.'' (Optional: ``May soak wart in 
warm water for 5 minutes.'') ``Dry area thoroughly.'' (If appropriate: 
``Cut plaster to fit wart.'') ``Apply medicated plaster. Repeat 
procedure every 48 hours as needed (until wart is removed) for up to 12 
weeks.''
    (2) For products containing salicylic acid identified in 
Sec. 358.110(b). ``Wash affected area.'' (Optional: ``May soak wart in 
warm water for 5 minutes.'') ``Dry area thoroughly. Apply'' (select one 
of the following, as appropriate: ``one drop'' or ``small amount'') ``at 
a time with'' (select one of the following, as appropriate: 
``applicator'' or ``brush'') ``to sufficiently cover each wart. Let dry. 
Repeat this procedure once or twice daily as needed (until wart is 
removed) for up to 12 weeks.''
    (3) For products containing salicylic acid identified in 
Sec. 358.110(c). ``Wash affected area.'' (Optional: ``May soak wart in 
warm water for 5 minutes.'') ``Dry area thoroughly. Gently smooth wart 
surface with emery file supplied.'' (If appropriate: ``Cut plaster to 
fit wart.'') ``Apply a drop of warm water to the wart, keeping the 
surrounding skin dry. Apply medicated plaster at bedtime and leave in 
place for at least 8 hours. In the morning, remove plaster and discard. 
Repeat procedure every 24 hours as needed (until wart is removed) for up 
to 12 weeks.''
    (e) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this section.
    (f) The phrase ``or podiatrist'' may be used in addition to the word 
``doctor'' in any of the labeling statements in this section when a 
product is labeled with the indication identified in Sec. 358.150(b)(2).
[55 FR 33255, Aug. 14, 1990; 55 FR 37403, Sept. 11, 1990, as amended at 
57 FR 44495, Sept. 28, 1992; 59 FR 60317, Nov. 23, 1994]



                         Subparts C-E [Reserved]



            Subpart F--Corn and Callus Remover Drug Products

    Source:  55 FR 33261, Aug. 14, 1990, unless otherwise noted.



Sec. 358.501  Scope.

    (a) An over-the-counter corn and callus remover drug product in a 
form suitable for topical application is generally recognized as safe 
and effective

[[Page 281]]

and is not misbranded if it meets each of the conditions in this subpart 
and each of the general conditions established in Sec. 330.1 of this 
chapter.
    (b) References in this subpart to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 358.503  Definitions.

    As used in this subpart:
    (a) Corn and callus remover drug product. A topical agent used for 
the removal of corns and calluses.
    (b) Collodion-like vehicle. A solution containing pyroxylin 
(nitrocellulose) in an appropriate nonaqueous solvent that leaves a 
transparent cohesive film when applied to the skin in a thin layer.
    (c) Plaster vehicle. A fabric, plastic, or other suitable backing 
material in which medication is usually incorporated for topical 
application to the skin.



Sec. 358.510  Corn and callus remover active ingredients.

    The product consists of any of the following active ingredients 
within the specified concentrations and in the dosage form established 
for each ingredient.
    (a) Salicylic acid 12 to 40 percent in a plaster vehicle.
    (b) Salicylic acid 12 to 17.6 percent in a collodion-like vehicle.



Sec. 358.550  Labeling of corn and callus remover drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as a 
``corn and callus remover.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the phrase listed in paragraph (b)(1) of this 
section and may contain the additional phrase listed in paragraph (b)(2) 
of this section. Other truthful and nonmisleading statements, describing 
only the indications for use that have been established in paragraph (b) 
of this section, may also be used, as provided in Sec. 330.1(c)(2) of 
this chapter, subject to the provisions of section 502 of the Federal 
Food, Drug, and Cosmetic Act (the act) relating to misbranding and the 
prohibition in section 301(d) of the act against the introduction or 
delivery for introduction into interstate commerce of unapproved new 
drugs in violation of section 505(a) of the act.
    (1) ``For the removal of corns and calluses.''
    (2) In addition to the information identified in paragraph (b)(1) of 
this section, the labeling of the product may contain the following 
statement: ``Relieves pain by removing corns and calluses.''
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) For products containing any ingredient identified in 
Sec. 358.510. (i) ``For external use only.''
    (ii) ``Do not use this product on irritated skin, on any area that 
is infected or reddened, if you are a diabetic, or if you have poor 
blood circulation.''
    (iii) ``If discomfort persists, see your doctor or podiatrist.''
    (2) For any product formulated in a flammable vehicle. (i) The 
labeling should contain an appropriate flammability signal word, e.g., 
``extremely flammable,'' ``flammable,'' ``combustible,'' consistent with 
16 CFR 1500.3(b)(10).
    (ii) ``Keep away from fire or flame.''
    (3) For any product formulated in a volatile vehicle. ``Cap bottle 
tightly and store at room temperature away from heat.''
    (4) For any product formulated in a collodion-like vehicle. (i) ``If 
product gets into the eye, flush with water for 15 minutes.''
    (ii) ``Avoid inhaling vapors.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) For products containing salicylic acid identified in 
Sec. 358.510(a). ``Wash affected area and dry thoroughly.'' (If 
appropriate: ``Cut plaster to fit corn/callus.'') ``Apply medicated 
plaster. After 48 hours remove the medicated plaster. Repeat this 
procedure every 48 hours as needed for up to 14 days (until corn/callus 
is removed).'' (Optional: ``May soak corn/callus in warm water for 5 
minutes to assist in removal.'')

[[Page 282]]

    (2) For products containing salicylic acid identified in 
Sec. 358.510(b). ``Wash affected area and dry thoroughly. Apply'' 
(select one of the following, as appropriate: ``one drop'' or ``small 
amount'') ``at a time with'' (select one of the following, as 
appropriate: ``applicator'' or ``brush'') ``to sufficiently cover each 
corn/callus. Let dry. Repeat this procedure once or twice daily as 
needed for up to 14 days (until corn/callus is removed).'' (Optional: 
``May soak corn/callus in warm water for 5 minutes to assist in 
removal.'')
    (e) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this section.
[55 FR 33261, Aug. 14, 1990, as amended at 57 FR 44494, Sept. 28, 1992]



                  Subpart G--Pediculicide Drug Products

    Source:  58 FR 65455, Dec. 14, 1993, unless otherwise noted.



Sec. 358.601  Scope.

    (a) An over-the-counter pediculicide drug product in a form suitable 
for topical application is generally recognized as safe and effective 
and is not misbranded if it meets each condition in this subpart and 
each general condition established in Sec. 330.1 of this chapter.
    (b) References in this subpart to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 358.603  Definition.

    As used in this subpart:
    Pediculicide drug product. A drug product for the treatment of head, 
pubic (crab), and body lice.



Sec. 358.610  Pediculicide active ingredients.

    The active ingredients of the product consist of the combination of 
pyrethrum extract (0.17 to 0.33 percent) with piperonyl butoxide (2 to 4 
percent) in a nonaerosol dosage formulation.



Sec. 358.650  Labeling of pediculicide drug products.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product as a 
``pediculicide (lice treatment)'' or ``lice treatment.''
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the following: ``For the treatment of head, 
pubic (crab), and body lice.'' Other truthful and nonmisleading 
statements, describing only the indications for use that have been 
established and listed in paragraph (b) of this section, may also be 
used, as provided in Sec. 330.1(c)(2) of this chapter, subject to the 
provisions of section 502 of the Federal Food, Drug, and Cosmetic Act 
(the act) relating to misbranding and the prohibition in section 301(d) 
of the act against the introduction or delivery for introduction into 
interstate commerce of unapproved new drugs in violation of section 
505(a) of the act.
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) ``Use with caution on persons allergic to ragweed.''
    (2) ``For external use only. Do not use near the eyes or permit 
contact with mucous membranes, such as inside the nose, mouth, or 
vagina, as irritation may occur. Keep out of eyes when rinsing hair. 
Adults and children: Close eyes tightly and do not open eyes until 
product is rinsed out. Also, protect children's eyes with washcloth, 
towel or other suitable material, or by a similar method. If product 
gets into the eyes, immediately flush with water.''
    (3) ``If skin irritation or infection is present or develops, 
discontinue use and consult a doctor. Consult a doctor if infestation of 
eyebrows or eyelashes occurs.''
    (4) The word ``physician'' may be substituted for the word 
``doctor'' in any of the warning statements in this paragraph.
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions'':
    (1) For all products. ``Important: Read warnings before using.'' 
[sentence in all capital letters and boldface type]
    (2) For nonshampoo products. ``Apply to affected area until all the 
hair is thoroughly wet with product. Allow

[[Page 283]]

product to remain on area for 10 minutes but no longer. Wash area 
thoroughly with warm water and soap or shampoo. A fine-toothed comb or a 
special lice/nit removing comb may be used to help remove dead lice or 
their eggs (nits) from hair. A second treatment must be done in 7 to 10 
days to kill any newly hatched lice.''
    (3) For products formulated for use as a shampoo. ``Apply to 
affected area until all the hair is thoroughly wet with product. Allow 
product to remain on area for 10 minutes but no longer. Add sufficient 
warm water to form a lather and shampoo as usual. Rinse thoroughly. A 
fine-toothed comb or a special lice/nit removing comb may be used to 
help remove dead lice or their eggs (nits) from hair. A second treatment 
must be done in 7 to 10 days to kill any newly hatched lice.''
    (e) Other required statements.
    (1) ``Head Lice: Head lice live on the scalp and lay small white 
eggs (nits) on the hair shaft close to the scalp. The nits are most 
easily found on the nape of the neck or behind the ears. All personal 
headgear, scarfs, coats, and bed linen should be disinfected by machine 
washing in hot water and drying, using the hot cycle of a dryer for at 
least 20 minutes. Personal articles of clothing or bedding that cannot 
be washed may be dry-cleaned, sealed in a plastic bag for a period of 
about 2 weeks, or sprayed with a product specifically designed for this 
purpose. Personal combs and brushes may be disinfected by soaking in hot 
water (above 130  deg. F) for 5 to 10 minutes. Thorough vacuuming of 
rooms inhabited by infected patients is recommended.''
    (2) ``Pubic (Crab) Lice: Pubic lice may be transmitted by sexual 
contact; therefore, sexual partners should be treated simultaneously to 
avoid reinfestation. The lice are very small and look almost like brown 
or grey dots on the skin. Pubic lice usually cause intense itching and 
lay small white eggs (nits) on the hair shaft generally close to the 
skin surface. In hairy individuals, pubic lice may be present on the 
short hairs of the thighs and trunk, underarms, and occasionally on the 
beard and mustache. Underwear should be disinfected by machine washing 
in hot water; then drying, using the hot cycle for at least 20 
minutes.''
    (3) ``Body Lice: Body lice and their eggs are generally found in the 
seams of clothing, particularly in the waistline and armpit area. They 
move to the skin to feed, then return to the seams of the clothing where 
they lay their eggs. Clothing worn and not laundered before treatment 
should be disinfected by the same procedure as described for head lice, 
except that sealing clothing in a plastic bag is not recommended for 
body lice because the nits (eggs) from these lice can remain dormant for 
a period of up to 30 days.''



    Subpart H--Drug Products for the Control of Dandruff, Seborrheic 
                        Dermatitis, and Psoriasis

    Source:  56 FR 63568, Dec. 4, 1991, unless otherwise noted.



Sec. 358.701  Scope.

    (a) An over-the-counter dandruff, seborrheic dermatitis, or 
psoriasis drug product in a form suitable for topical application is 
generally recognized as safe and effective and is not misbranded if it 
meets each of the conditions in this subpart and each general condition 
established in Sec. 330.1 of this chapter.
    (b) References in this subpart to regulatory sections of the Code of 
Federal Regulations are to chapter I of title 21 unless otherwise noted.



Sec. 358.703  Definitions.

    As used in this subpart:
    (a) Coal tar. The tar used for medicinal purposes that is obtained 
as a byproduct during the destructive distillation of bituminous coal at 
temperatures in the range of 900  deg.C to 1,100  deg.C. It may be 
further processed using either extraction with alcohol and suitable 
dispersing agents and maceration times or fractional distillation with 
or without the use of suitable organic solvents.
    (b) Dandruff. A condition involving an increased rate of shedding of 
dead epidermal cells of the scalp.
    (c) Psoriasis. A condition of the scalp or body characterized by 
irritation,

[[Page 284]]

itching, redness, and extreme excess shedding of dead epidermal cells.
    (d) Seborrheic dermatitis. A condition of the scalp or body 
characterized by irritation, itching, redness, and excess shedding of 
dead epidermal cells.
    (e) Selenium sulfide, micronized. Selenium sulfide that has been 
finely ground and that has a median particle size of approximately 5 
micrometers (m), with not more than 0.1 percent of the 
particles greater than 15 m and not more than 0.1 percent of 
the particles less than 0.5 m.
[56 FR 63568, Dec. 4, 1991, as amended at 59 FR 4001, Jan. 28, 1994]



Sec. 358.710  Active ingredients for the control of dandruff, seborrheic dermatitis, or psoriasis.

    The active ingredient of the product consists of any of the 
following within the specified concentration established for each 
ingredient:
    (a) Active ingredients for the control of dandruff. (1) Coal tar, 
0.5 to 5 percent. When a coal tar solution, derivative, or fraction is 
used as the source of the coal tar, the labeling shall specify the 
identity and concentration of the coal tar source used and the 
concentration of the coal tar present in the final product.
    (2) Pyrithione zinc, 0.3 to 2 percent when formulated to be applied 
and then washed off after brief exposure.
    (3) Pyrithione zinc, 0.1 to 0.25 percent when formulated to be 
applied and left on the skin or scalp.
    (4) Salicylic acid, 1.8 to 3 percent.
    (5) Selenium sulfide, 1 percent.
    (6) Selenium sulfide, micronized, 0.6 percent.
    (7) Sulfur, 2 to 5 percent.
    (b) Active ingredients for the control of seborrheic dermatitis. (1) 
Coal tar, 0.5 to 5 percent. When a coal tar solution, derivative, or 
fraction is used as the source of the coal tar, the labeling shall 
specify the identity and concentration of the coal tar source used and 
the concentration of the coal tar present in the final product.
    (2) Pyrithione zinc, 0.95 to 2 percent when formulated to be applied 
and then washed off after brief exposure.
    (3) Pyrithione zinc, 0.1 to 0.25 percent when formulated to be 
applied and left on the skin or scalp.
    (4) Salicylic acid, 1.8 to 3 percent.
    (5) Selenium sulfide, 1 percent.
    (c) Active ingredients for the control of psoriasis. (1) Coal tar, 
0.5 to 5 percent. When a coal tar solution, derivative, or fraction is 
used as the source of the coal tar, the labeling shall specify the 
identity and concentration of the coal tar source used and the 
concentration of the coal tar present in the final product.
    (2) Salicylic acid, 1.8 to 3 percent.
[56 FR 63568, Dec. 4, 1991, as amended at 59 FR 4001, Jan. 28, 1994]



Sec. 358.720  Permitted combinations of active ingredients.

    Salicylic acid identified in Sec. 358.710(a) (4) may be combined 
with sulfur identified in Sec. 358.710(a)(6) provided each ingredient is 
present within the established concentration and the product is labeled 
for the control of dandruff.



Sec. 358.750  Labeling of drug products for the control of dandruff, seborrheic dermatitis, or psoriasis.

    (a) Statement of identity. The labeling of the product contains the 
established name of the drug, if any, and identifies the product with 
one or more of the following, as appropriate:
    (1) ``Dandruff (insert product form)'' or ``antidandruff (insert 
product form)''.
    (2) ``Seborrheic dermatitis (insert product form)''.
    (3) ``Psoriasis (insert product form)''.
    (b) Indications. The labeling of the product states, under the 
heading ``Indications,'' the phrase listed in paragraph (b)(1) of this 
section and may contain any of the terms listed in paragraph (b)(2) or 
(b)(3) of this section. Other truthful and nonmisleading statements, 
describing only the indications for use that have been established and 
listed in paragraph (b) of this section, may also be used, as provided 
in Sec. 330.1(c)(2) of this chapter, subject to the provisions of 
section 502 of the Federal Food, Drug, and Cosmetic Act (the act) 
relating to misbranding and the prohibition in section 301(d) of the act 
against the introduction or delivery for introduction into interstate 
commerce of unapproved new drugs in violation of section 505(a) of the 
act.

[[Page 285]]

    (1) (``For relief of'' or ``Controls'') ``the symptoms of'' (select 
one or more of the following, as appropriate: ``dandruff,'' ``seborrheic 
dermatitis,'' and/or ``psoriasis.'')
    (2) The following terms or phrases may be used in place of or in 
addition to the words ``For the relief of'' or ``Controls'' in the 
indications in paragraph (b)(1) of this section: ``fights,'' 
``reduces,'' ``helps eliminate,'' ``helps stop,'' ``controls recurrence 
of,'' ``fights recurrence of,'' ``helps prevent recurrence of,'' 
``reduces recurrence of,'' ``helps eliminate recurrence of,'' ``helps 
stop recurrence of.''
    (3) The following terms may be used in place of the words ``the 
symptoms of'' in the indications in paragraph (b)(1) of this section: 
(``skin'' and/or ``scalp,'' as appropriate) (select one or more of the 
following: ``itching,'' ``irritation,'' ``redness,'' ``flaking,'' 
``scaling,'') ``associated with.''
    (c) Warnings. The labeling of the product contains the following 
warnings under the heading ``Warnings'':
    (1) For products containing any ingredient identified in 
Sec. 358.710. (i) ``For external use only.''
    (ii) ``Avoid contact with the eyes. If contact occurs, rinse eyes 
thoroughly with water.''
    (iii) ``If condition worsens or does not improve after regular use 
of this product as directed, consult a doctor.''
    (2) For any product containing coal tar identified in 
Sec. 358.710(a), (b), or (c). (i) ``Use caution in exposing skin to 
sunlight after applying this product. It may increase your tendency to 
sunburn for up to 24 hours after application.''
    (ii) ``Do not use for prolonged periods without consulting a 
doctor.''
    (3) For products containing coal tar when formulated to be applied 
and left on the skin (e.g., creams, ointments, lotions). ``Do not use 
this product in or around the rectum or in the genital area or groin 
except on the advice of a doctor.''
    (4) For products containing coal tar identified in Sec. 358.710(c) 
for the control of psoriasis. ``Do not use this product with other forms 
of psoriasis therapy such as ultraviolet radiation or prescription drugs 
unless directed to do so by a doctor.''
    (5) For products containing any ingredient identified in 
Sec. 358.710(b) or (c) for the control of seborrheic dermatitis or 
psoriasis. ``If condition covers a large area of the body, consult your 
doctor before using this product.''
    (d) Directions. The labeling of the product contains the following 
information under the heading ``Directions.'' More detailed directions 
applicable to a particular product formulation may also be included.
    (1) For products containing active ingredients for the control of 
dandruff, seborrheic dermatitis, or psoriasis when formulated to be 
applied and then washed off after brief (a few minutes) exposure (e.g, 
shampoos, preshampoo rinses, postshampoo rinses). ``For best results use 
at least twice a week or as directed by a doctor.''
    (2) For products containing active ingredients for the control of 
dandruff, seborrheic dermatitis, or psoriasis when formulated so as to 
be applied and left on the skin or scalp (e.g., creams, ointments, 
lotions, hairgrooms). ``Apply to affected areas one to four times daily 
or as directed by a doctor.''
    (3) For products containing active ingredients for the control of 
seborrheic dermatitis or psoriasis of the skin when formulated as soaps. 
``Use on affected areas in place of your regular soap.''
    (e) The word ``physician'' may be substituted for the word 
``doctor'' in any of the labeling statements in this section.



PART 361--PRESCRIPTION DRUGS FOR HUMAN USE GENERALLY RECOGNIZED AS SAFE AND EFFECTIVE AND NOT MISBRANDED: DRUGS USED IN RESEARCH--Table of Contents




    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 371; 42 U.S.C. 262.



Sec. 361.1  Radioactive drugs for certain research uses.

    (a) Radioactive drugs (as defined in Sec. 310.3(n) of this chapter) 
are generally recognized as safe and effective when administered, under 
the conditions set forth in paragraph (b) of this section, to human 
research subjects during the course of a research project intended to 
obtain basic information regarding the

[[Page 286]]

metabolism (including kinetics, distribution, and localization) of a 
radioactively labeled drug or regarding human physiology, 
pathophysiology, or biochemistry, but not intended for immediate 
therapeutic, diagnostic, or similar purposes or to determine the safety 
and effectiveness of the drug in humans for such purposes (i.e., to 
carry out a clinical trial). Certain basic research studies, e.g., 
studies to determine whether a drug localizes in a particular organ or 
fluid space and to describe the kinetics of that localization, may have 
eventual therapeutic or diagnostic implications, but the initial studies 
are considered to be basic research within the meaning of this section.
    (b) The conditions under which use of radioactive drugs for research 
are considered safe and effective are:
    (1) Approval by Radioactive Drug Research Committee. A Radioactive 
Drug Research Committee, composed and approved by the Food and Drug 
Administration in accordance with paragraph (c) of this section, has 
determined, in accordance with the standards set forth in paragraph (d) 
of this section, that:
    (i) The pharmacological dose is within the limits set forth in 
paragraph (b)(2) of this section;
    (ii) The radiation dose is within the limits set forth in paragraph 
(b)(3) of this section;
    (iii) The radiation exposure is justified by the quality of the 
study being undertaken and the importance of the information it seeks to 
obtain;
    (iv) The study meets the other requirements set forth in paragraph 
(d) of this section regarding qualifications of the investigator, proper 
licensure for handling radioactive materials, selection and consent of 
research subjects, quality of radioactive drugs used, research protocol 
design, reporting of adverse reactions, and approval by an appropriate 
Institutional Review Committee; and
    (v) The use of the radioactive drug in human subjects has the 
approval of the Radioactive Drug Research Committee.
    (2) Limit on pharmacological dose. The amount of active ingredient 
or combination of active ingredients to be administered shall be known 
not to cause any clinically detectable pharmacological effect in human 
beings. If the same active ingredients (exclusive of the radionuclide) 
are to be administered simultaneously, e.g., under a ``Investigational 
New Drug Application'' or for a therapeutic use in accordance with 
labeling for a drug approved under part 314 of this chapter, the total 
amount of active ingredients including the radionuclide shall be known 
not to exceed the dose limitations applicable to the separate 
administration of the active ingredients excluding the radionuclide.
    (3) Limit on radiation dose. The amount of radioactive material to 
be administered shall be such that the subject receives the smallest 
radiation dose with which it is practical to perform the study without 
jeopardizing the benefits to be obtained from the study.
    (i) Under no circumstances may the radiation dose to an adult 
research subject from a single study or cumulatively from a number of 
studies conducted within 1 year be generally recognized as safe if such 
dose exceeds the following:

Whole body, active blood-forming organs, lens of the eye, and gonads:

                                                                        
------------------------------------------------------------------------
                                                                  Rems  
------------------------------------------------------------------------
  Single dose.................................................         3
  Annual and total dose commitment............................         5
Other organs:                                                           
  Single dose.................................................         5
  Annual and total dose commitment............................        15
------------------------------------------------------------------------

    (ii) For a research subject under 18 years of age at his last 
birthday, the radiation dose shall not exceed 10 percent of that set 
forth in paragraph (b)(3)(i) of this section.
    (iii) All radioactive material included in the drug either as 
essential material or as a significant contaminant or impurity shall be 
included when determining the total radiation doses and dose 
commitments. Radiation doses from x-ray procedures that are part of the 
research study (i.e., would not have occurred but for the study) shall 
also be included. The possibility of followup studies shall be 
considered for inclusion in the dose calculations.
    (iv) Numerical definitions of dose shall be based on an absorbed 
fraction

[[Page 287]]

method of radiation absorbed dose calculation, such as the system set 
forth by the Medical Internal Radiation Dose Committee of the Society of 
Nuclear Medicine, or the system set forth by the International 
Commission on Radiological Protection.
    (c) A Radioactive Drug Research Committee, in order to comply with 
paragraph (b)(1) of this section, shall be composed, shall function, and 
shall obtain and maintain approval of the Food and Drug Administration 
in conformity with the following:
    (1) Membership. A Radioactive Drug Research Committee shall consist 
of at least five individuals. Each committee shall include the following 
three individuals: (i) A physician recognized as a specialist in nuclear 
medicine, (ii) a person qualified by training and experience to 
formulate radioactive drugs, and (iii) a person with special competence 
in radiation safety and radiation dosimetry. The remainder of the 
committee shall consist of individuals qualified in various disciplines 
pertinent to the field of nuclear medicine (e.g., radiology, internal 
medicine, clinical pathology, hematology, endocrinology, radiation 
therapy, radiation physics, radiation biophysics, health physics, and 
radiopharmacy). Membership shall be sufficiently diverse to permit 
expert review of the technical and scientific aspects of proposals 
submitted to the committee. The addition of consultants in other 
pertinent medical disciplines is encouraged. A Radioactive Drug Research 
Committee shall be either associated with a medical institution operated 
for care of patients and with sufficient scientific expertise to allow 
for selection of committee members from its faculty, or with a committee 
established by a State authority to provide advice on radiation health 
matters. Joint committees involving more than one medical institution 
which have been established in order to achieve a high level and 
diversity of experience will be acceptable. The Director of the Center 
for Drug Evaluation and Research may modify any of the foregoing 
requirements in a particular situation where alternative factors provide 
substantially the same composition and association.
    (2) Function. Each Radioactive Drug Research Committee shall select 
a chairman, who shall sign all applications, minutes, and reports of the 
committee. Each committee shall meet at least once each quarter in which 
research activity has been authorized or conducted. A quorum consisting 
of more than 50 percent of the membership must be present with 
appropriate representation of the required fields of specialization. 
Minutes shall be kept and shall include the numerical results of votes 
on protocols involving use in human subjects. No member shall vote on a 
protocol in which he is an investigator.
    (3) Reports. Each Radioactive Drug Research Committee shall submit 
an annual report on or before January 31 of each year to the Food and 
Drug Administration, Center for Drug Evaluation and Research, HFD-160, 
5600 Fishers Lane, Rockville, MD 20857. The annual report shall include 
the names and qualifications of the members of, and of any consultants 
used by, the Radioactive Drug Research Committee, and, for each study 
conducted during the preceding year, a summary of information presented 
in the following format:

               Report on Research Use of Radioactive Drug

    1. Title of the research project.
    2. Brief description of the purpose of the research project.
    3. Name of the investigator responsible.
    4. Pharmacological dose:
    a. Active ingredients.
    b. Maximum amount administered per subject.
    5. Name of the radionuclide(s) used, including any present, as 
significant contaminants or impurities.
    6. Radiation absorbed dose. Provide the maximum dose commitement to 
the whole body and each organ specified in 21 CFR 361.1(b)(3)(i) that 
was received by a representative subject and the calculations or 
references that were used to estimate these maximum dose commitments. 
The report shall include the dose contribution of both the administered 
radionuclide(s) and any X-ray procedures associated with the study. If 
the study elicits data on the uptake or excretion of the radioactive 
drug pertinent to the estimation of dose commitment, report the mean 
value and range of values. For each subject provide:
    (a) Age, sex, and approximate weight.

[[Page 288]]

    (b) Total activity of each radionuclide administered for each 
radioactive drug used in the study. Report each X-ray procedure used in 
conjunction with the study.
    (c) If the subject has participated in other radioactive drug 
research studies, report the name of the radioactive drug used in these 
other studies, the date of administration, and the total activity of 
each radionuclide administered. If any X-ray procedures were used, 
identify the X-ray procedure(s) and include an estimate of the absorbed 
radiation doses.
    (d) If more than one administration of a radioactive drug per 
subject, cumulative radiation dose and dose commitment, expressed as 
whole body, active blood-forming organs, lens of the eye, gonads, and 
other organ doses from the administered radionuclides.
    7. A claim of confidentiality, if any.
    Note: Contents of this report are available for public disclosure 
unless confidentiality is requested by the investigator and it is 
adequately shown by the investigator that the report constitutes a trade 
secret or confidential commercial information as defined in 21 CFR 
20.61.
                       _________________________________________________
                                                        Investigator    
                      __________________________________________________
                                              Chairman, Radioactive Drug
                                                  Research Committee    


At any time a proposal is approved which involves exposure either of 
more than 30 research subjects, or of any research subject under 18 
years of age, the committee shall immediately submit to the Food and 
Drug Administration a special summary of information in the format shown 
in this paragraph. Contents of these reports are available for public 
disclosure, unless confidentiality is requested by the investigator and 
it is adequately shown by the investigator that the report constitutes a 
trade secret or confidential commercial information as defined in 
Sec. 20.61 of this chapter.
    (4) Approval. Each Radioactive Drug Research Committee shall be 
specifically approved by the Center for Drug Evaluation and Research of 
the Food and Drug Administration. Applications shall be submitted to the 
Food and Drug Administration, Center for Drug Evaluation and Research, 
HFD-160, 5600 Fishers Lane, Rockville, MD 20857, and shall contain the 
names and qualifications of the members of the committee, and a 
statement that the committee agrees to comply with the requirements set 
forth in this section. Approval shall be based upon an assessment of the 
qualifications of the members of the committee, and the assurance that 
all necessary fields of expertise are covered. Approval of a committee 
may be withdrawn at any time for failure of the committee to comply with 
any of the requirements of this section. Approval of a committee shall 
remain effective unless and until the FDA withdraws such approval. 
Changes in membership and applications for new members shall be 
submitted to the Food and Drug Administration as soon as, or before, 
vacancies occur on the committee.
    (5) Monitoring. The Food and Drug Administration shall conduct 
periodic reviews of approved committees. Monitoring of the activities of 
the committee shall be conducted through review of its annual report, 
through review of minutes and full protocols for certain studies, and 
through on-site inspections.
    (d) In making the determination required in paragraph (b)(1) of this 
section, a Radioactive Drug Research Committee shall consider the 
following requirements and assure that each is met:
    (1) Radiation dose to subjects. To assure that the radiation dose to 
research subjects is as low as practicable to perform the study and meet 
the criteria of Sec. 361.1(b)(3), the Radioactive Drug Research 
Committee shall require that:
    (i) The investigator provide absorbed dose calculations based on 
biologic distribution data available from published literature or from 
other valid studies.
    (ii) The investigator provide for an acceptable method of radioassay 
of the radioactive drug prior to its use to assure that the dose 
calculations actually reflect the administered dose.
    (iii) The radioactive drug chosen for the study has that combination 
of half-life, types of radiations, radiation energy, metabolism, 
chemical properties, etc., which results in the lowest dose to the whole 
body or specific organs with which it is possible to obtain the 
necessary information.
    (iv) The investigator utilize adequate and appropriate 
instrumentation for the detection and measurement of the specific 
radionuclide.

[[Page 289]]

    (2) Pharmacological dosage. To determine that the amount of active 
ingredients to be administered does not exceed the limitations set forth 
in paragraph (b)(2) of this section, the committee shall require that 
the investigator provide pharmacological dose calculations based on data 
available from published literature or from other valid human studies.
    (3) Qualifications of investigators. Each investigator shall be 
qualified by training and experience to conduct the proposed research 
studies.
    (4) License to handle radioactive materials. The responsible 
investigator or institutions shall, in the case of reactor-produced 
isotopes, be licensed by the Nuclear Regulatory Commission or Agreement 
State to possess and use the specific radionuclides for research use or 
be a listed investigator under a broad license, or in the case of non-
reactor-produced isotopes, be licensed by other appropriate State or 
local authorities, when required by State or local law, to possess and 
use the specific radionuclides for research use.
    (5) Human research subjects. Each investigator shall select 
appropriate human subjects and shall obtain the review and approval of 
an institutional review committee that conforms to the requirements of 
part 56 of this chapter, and shall obtain the consent of the subjects or 
their legal representatives in accordance with part 50 of this chapter. 
The research subjects shall be at least 18 years of age and legally 
competent. Exceptions are permitted only in those special situations 
when it can be demonstrated to the committee that the study presents a 
unique opportunity to gain information not currently available, requires 
the use of research subjects less than 18 years of age, and is without 
significant risk to the subject. Studies involving minors shall be 
supported with review by qualified pediatric consultants to the 
Radioactive Drug Research Committee. Each female research subject of 
childbearing potential shall state in writing that she is not pregnant, 
or, on the basis of a pregnancy test be confirmed as not pregnant, 
before she may participate in any study.
    (6) Quality of radioactive drug. The radioactive drug used in the 
research study shall meet appropriate chemical, pharmaceutical, 
radiochemical, and radionuclidic standards of identity, strength, 
quality, and purity as needed for safety and be of such uniform and 
reproducible quality as to give significance to the research study 
conducted. The Radioactive Drug Research Committee shall determine that 
radioactive materials for parenteral use are prepared in sterile and 
pyrogen-free form.
    (7) Research protocol. No matter how small the amount of 
radioactivity, no study involving administration of a radioactive drug, 
as defined in Sec. 310.3(n) of this chapter, to research subjects under 
this section, shall be permitted unless the Radioactive Drug Research 
Committee concludes, in its judgment, that scientific knowledge and 
benefit is likely to result from that study. Therefore, the protocol 
shall be based upon a sound rationale derived from appropriate animal 
studies or published literature and shall be of sound design such that 
information of scientific value may result. The radiation dose shall be 
both sufficient and no greater than necessary to obtain valid 
measurement. The projected number of subjects shall be sufficient but no 
greater than necessary for the purpose of the study. The number of 
subjects shall also reflect the fact that the study is intended to 
obtain basic research information referred to in paragraph (a) of this 
section and not intended for immediate therapeutic, diagnostic or 
similar purposes or to determine the safety and effectiveness of the 
drug in humans for such purposes (i.e., to carry out a clinical trial).
    (8) Adverse reactions. The investigator shall immediately report to 
the Radioactive Drug Research Committee all adverse effects associated 
with the use of the radioactive drug in the research study. All adverse 
reactions probably attributable to the use of the radioactive drug in 
the research study shall be immediately reported by the Radioactive Drug 
Research Committee to the Food and Drug Administration, Center for Drug 
Evaluation and Research, HFD-160, 5600 Fishers Lane, Rockville, MD 
20857.
    (9) Approval by an institutional review board. The investigator 
shall obtain the

[[Page 290]]

review and approval of an institutional review board that conforms to 
the requirements of part 56 of this chapter.
    (e) The results of any research conducted pursuant to this section 
as part of the evaluation of a drug pursuant to part 312 of this chapter 
shall be included in the submissions required under part 312 of this 
chapter.
    (f) A radioactive drug prepared, packaged, distributed, and 
primarily intended for use in accordance with the requirements of this 
section shall be exempt from section 502(f)(1) of the act and 
Secs. 201.5 and 201.100 of this chapter if the packaging, label, and 
labeling are in compliance with Federal, State, and local law regarding 
radioactive materials and if the label of the immediate container and 
shielded container, if any, either separate from or as part of any label 
and labeling required for radioactive materials by the Nuclear 
Regulatory Commission or by State or local radiological health 
authorities bear the following:
    (1) The statement ``Caution: Federal law prohibits dispensing 
without prescription'';
    (2) The statement ``To be administered in compliance with the 
requirements of Federal regulations regarding radioactive drugs for 
research use (21 CFR 361.1)'';
    (3) The established name of the drug, if any;
    (4) The established name and quantity of each active ingredient;
    (5) The name and half-life of the radionuclide, total quantity of 
radioactivity in the drug product's immediate container, and amount of 
radioactivity per unit volume or unit mass at a designated referenced 
time;
    (6) The route of administration, if it is for the other than oral 
use;
    (7) The net quantity of contents;
    (8) An identifying lot or control number from which it is possible 
to determine the complete manufacturing history of the package of the 
drug;
    (9) The name and address of the manufacturer, packer, or 
distributor;
    (10) The expiration date, if any;
    (11) If the drug is intended for parenteral use, a statement as to 
whether the contents are sterile;
    (12) If the drug is for other than oral use, the names of all 
inactive ingredients, except that:
    (i) Trace amounts of harmless substances added solely for individual 
product identification need not be named.
    (ii) If the drug is intended for parenteral use, the quantity or 
proportion of all inactive ingredients, except that ingredients added to 
adjust pH or to make the drug isotonic may be declared by name and a 
statement of their effect; if the vehicle is water for injection, it 
need not be named. Provided, however, That in the case of containers too 
small or otherwise unable to accommodate a label with sufficient space 
to bear all such information, the information required by paragraphs (f) 
(1) and (12) of this section may be placed on the shielded container 
only.
[40 FR 31308, July 25, 1975, as amended at 40 FR 44543, Sept. 29, 1975; 
42 FR 15674, Mar. 22, 1977; 43 FR 14646, Apr. 7, 1978; 46 FR 8955, Jan. 
27, 1981; 49 FR 44460, Nov. 7, 1984; 50 FR 8996, Mar. 6, 1985; 55 FR 
11582, Mar. 29, 1990; 56 FR 10806, Mar. 14, 1991]



PART 369--INTERPRETATIVE STATEMENTS RE WARNINGS ON DRUGS AND DEVICES FOR OVER-THE-COUNTER SALE--Table of Contents




               Subpart A--Definitions and Interpretations

Sec.
369.1  Purpose of issuance.
369.2  Definitions.
369.3  Warnings required on drugs exempted from prescription-dispensing 
          requirements of section 503(b)(1)(C).
369.4  Warnings suggested for drugs by formal or informal statements of 
          policy.
369.5  Warnings required on insulin intended for over-the-counter sale.
369.6  [Reserved]
369.7  Warnings required by official compendia.
369.8  Warning statements in relation to conditions for use.
369.9  General warnings re accidental ingestion by children.
369.10  Conspicuousness of warning statements.

           Subpart B--Warning and Caution Statements for Drugs

369.20  Drugs; recommended warning and caution statements.

[[Page 291]]

369.21  Drugs; warning and caution statements required by regulations.
369.22  Drugs; warning and caution statements specifically required by 
          law.

    Authority:  21 U.S.C. 321, 331, 351, 352, 353, 355, 356, 357, 371.

    Source:  39 FR 11745, Mar. 29, 1974, unless otherwise noted.



               Subpart A--Definitions and Interpretations



Sec. 369.1  Purpose of issuance.

    The warning and caution statements suggested in subparts B and C of 
this part, for inclusion in the label or labeling of drugs and devices 
subject to section 502(d) and (f)(2) and other relevant provisions of 
the Federal Food, Drug, and Cosmetic Act are issued for the purpose of 
assisting industry in preparing proper labeling for these articles for 
over-the-counter sale and in meeting the legal requirements of the act 
that the label or labeling of drugs and devices bear adequate warnings, 
in such manner and form as are necessary for the protection of users. 
Only section 502(d) of the act requires use of the specific language 
included in these suggested warning and caution statements. These 
suggested warning or caution statements are illustrative of those that 
may be necessary or desirable. It is the responsibility of the 
manufacturer, packer, shipper, or distributor in interstate commerce to 
see that such statements are adequate for compliance with the provisions 
of the law. Omission of any article from this suggested list does not 
relieve drugs and devices subject to provisions of the act from bearing 
adequate warning or caution statements where such statements are 
necessary or desirable for the protection of the user.



Sec. 369.2  Definitions.

    (a) As used in this part, the term act means the Federal Food, Drug, 
and Cosmetic Act.
    (b) The terms drugs and devices are defined in section 201(g) and 
(k) of the act.
    (c) Official compendia are defined in section 201(j) of the act.



Sec. 369.3  Warnings required on drugs exempted from prescription-dispensing requirements of section 503(b)(1)(C).

    Drugs exempted from prescription-dispensing requirements under 
section 503(b)(1)(C) of the act are subject to the labeling requirements 
prescribed in Sec. 310.201(a) of this chapter. Although, for 
convenience, warning and caution statements for a number of the drugs 
named in Sec. 310.201 of this chapter (cross-referenced in the text of 
this part) are included in subpart B of this part, the inclusion of such 
drugs in Secs. 369.20, 369.21, 369.22 in no way affects the requirements 
for compliance with Sec. 310.201(a) of this chapter, or the provisions 
of an effective application pursuant to section 505(b) of the act.



Sec. 369.4  Warnings suggested for drugs by formal or informal statements of policy.

    The warning and caution statements included in subpart B of this 
part in no way affect any warning statement suggested for such drugs or 
devices by any statement of policy or interpretation in subchapter C of 
this chapter.
[39 FR 11745, Mar. 29, 1974, as amended at 40 FR 13496, Mar. 27, 1975]



Sec. 369.5  Warnings required on insulin intended for over-the-counter sale.

    Warning and caution statements for insulin products sold over the 
counter must comply with the specific labeling provisions of the act and 
Sec. 429.11 of this chapter.



Sec. 369.6  [Reserved]



Sec. 369.7  Warnings required by official compendia.

    Any drug included in the official compendia defined by the act shall 
bear such warning or caution statement as may be required by such 
compendia, and no statement in subpart B or subpart C of this part is 
intended to alter, modify, or permit the omission of any such statement 
required by such compendia.



Sec. 369.8  Warning statements in relation to conditions for use.

    The mention in any warning or caution statement included in subparts 
A,

[[Page 292]]

B, and C of this part, of a disease condition does not imply a finding 
on the part of the Food and Drug Administration that any drug or device 
is efficacious in such condition; nor is any drug or device bearing 
labeling referring to such disease condition precluded from regulatory 
action under the applicable provisions of the act if such claim is 
considered to be misbranding.



Sec. 369.9  General warnings re accidental ingestion by children.

    Section 369.20 includes under certain items, but not all medicines, 
the statement: ``Warning--Keep this and all medicines out of children's 
reach. In case of accidental overdose, contact a physician 
immediately'', or ``Warning--Keep out of the reach of children''. 
However, in view of the possibility of accidental ingestion of drugs, it 
is not only suggested but is recommended that one of these statements be 
used on the label of all drug products.



Sec. 369.10  Conspicuousness of warning statements.

    Necessary warning statements should appear in the labeling 
prominently and conspicuously as compared to other words, statements, 
designs, and devices, and in bold type on clearly contrasting 
background, in order to comply with the provisions of section 502(c) and 
(f)(2) of the act. The warning statements should be placed in the 
labeling in juxtaposition with the directions for use and, in any case, 
should appear on the label when there is sufficient label space in 
addition to mandatory label information.



           Subpart B--Warning and Caution Statements for Drugs



Sec. 369.20  Drugs; recommended warning and caution statements.

ACETANILID.

    Warning--Do not exceed recommended dosage. Overdosage or continued 
use may result in serious blood disturbances.

ACETOPHENETIDIN CONTAINING PREPARATIONS. (See Sec. 201.309 of this 
chapter.)

    Warning--This medication may damage the kidneys when used in large 
amounts or for a long period of time. Do not take more than the 
recommended dosage, nor take regularly for longer than 10 days without 
consulting your physician.

ANESTHETICS FOR EXTERNAL USE (LOCAL ANESTHETICS). (See also 
Sec. 310.201(a)(19) and (23) of this chapter.)

    Caution--Do not use in the eyes. Not for prolonged use. If the 
condition for which this preparation is used persists or if a rash or 
irritation develops, discontinue use and consult physician.

ANTIHISTAMINICS FOR EXTERNAL USE (EXCEPT PREPARATIONS FOR OPHTHALMIC 
USE).

    Caution--Do not use in the eyes. If the condition for which this 
preparation is used persists or if a rash or irritation develops, 
discontinue use and consult physician.


ANTIHISTAMINICS, ORAL. (See also Sec. 310.201(a)(4) and (a)(24) of this 
chapter.)

    Caution--This preparation may cause drowsiness. Do not drive or 
operate machinery while taking this medication. Do not give to children 
under 6 years of age or exceed the recommended dosage unless directed by 
physician.
    The reference to drowsiness is not required on preparations for the 
promotion of sleep or on preparations that are shown not to produce 
drowsiness.

ANTIPERSPIRANTS.

    Do not apply to broken skin. If a rash develops, discontinue use.

ANTIPYRINE.

    Warning--Do not exceed recommended dosage. If skin rash appears, 
discontinue use and consult physician.

ANTISEPTICS FOR EXTERNAL USE.

    Caution--In case of deep or puncture wounds or serious burns, 
consult physician. If redness, irritation, swelling, or pain persists or 
increases or if infection occurs discontinue use and consult physician.
    The reference to wounds and burns is not required on preparations 
intended solely for diaper rash.

ARSENIC PREPARATIONS.


[[Page 293]]


    Warning--Frequent or prolonged use may cause serious injury. Do not 
exceed recommended dosage. Keep out of the reach of children.

BELLADONNA PREPARATIONS AND PREPARATIONS OF ITS ALKALOIDS (ATROPINE, 
HYOSCYAMINE, AND SCOPOLAMINE (HYOSCINE); HYOSCYAMUS, STRAMONIUM, THEIR 
DERIVATIVES, AND RELATED DRUG PREPARATIONS.

    Warning--Not to be used by persons having glaucoma or excessive 
pressure within the eye, by elderly persons (where undiagnosed glaucoma 
or excessive pressure within the eye occurs most frequently), or by 
children under 6 years of age, unless directed by a physician. 
Discontinue use if blurring of vision, rapid pulse, or dizziness occurs. 
Do not exceed recommended dosage. Not for frequent or prolonged use. If 
dryness of the mouth occurs, decrease dosage. If eye pain occurs, 
discontinue use and see your physician immediately as this may indicate 
undiagnosed glaucoma.
    In the case of scopolamine or scopolamine aminoxide preparations 
indicated for insomnia, the portion of the above warning that reads 
``children under 6 years of age'' should read instead ``children under 
12 years of age''.

BORIC ACID (POWDERED, CRYSTALLINE, OR GRANULAR).

    Warning--Do not use as a dusting powder, especially on infants, or 
take internally. Use only as a solution. Do not apply to badly broken or 
raw skin, or to large areas of the body.

BROMIDES.

    Caution--Use only as directed. Do not give to children or use in the 
presence of kidney disease. If skin rash appears or if nervous symptoms 
persist, recur frequently, or are unusual, discontinue use and consult 
physician.

CARBOLIC ACID (PHENOL) PREPARATIONS (MORE THAN 0.5 PERCENT) FOR EXTERNAL 
USE.

    Warning--Use according to directions. Do not apply to large areas of 
the body. If applied to fingers or toes, do not bandage.

CATHARTICS AND LAXATIVES--IRRITANTS AND OTHER PERISTALTIC STIMULANTS.

    Warning--Do not use when abdominal pain, nausea, or vomiting are 
present. Frequent or prolonged use of this preparation may result in 
dependence on laxatives.
    Mercury preparations should have added to the ``frequent use'' 
statement, the words ``and serious mercury poisoning''.
    Phenolphthalein preparations should bear, in addition to the general 
warning, the following statement:
    Caution--If skin rash appears, do not use this or any other 
preparation containing phenolphthalein.
    See also Mineral Oil Laxatives.

CHLORATES: MOUTH WASH OR GARGLE.

    Avoid swallowing.

COBALT PREPARATIONS (See also Sec. 250.106 of this chapter.)

    Warning--Do not exceed the recommended dosage. Do not administer to 
children under 12 years of age unless directed by physician. Do not use 
for more than 2 months unless directed by physician.
    This warning is not required on articles containing not more than 
0.5 milligram of cobalt as a cobalt salt per dosage unit and which 
recommend administration of not more than 0.5 milligram per dose and not 
more than 2 milligrams per 24-hour period.

``COUGH-DUE-TO-COLD'' PREPARATIONS. (See also Sec. 310.201(a)(20) of 
this chapter.)

    Warning--Persons with a high fever or persistent cough should not 
use this preparation unless directed by physician.

COUNTERIRRITANTS AND RUBEFACIENTS.

    Caution--Do not apply to irritated skin or if excessive irritation 
develops. Avoid getting into the eyes or on mucous membranes.
    If offered for use in arthritis or rheumatism, in juxtaposition 
therewith, the statement:
    Caution--If pain persists for more than 10 days, or redness is 
present, or in conditions affecting children under 12 years of age 
consult a physician immediately.
    See also ``Salicylates'' in this section for additional warnings for 
preparations containing methyl salicylate.

[[Page 294]]


CREOSOTE, CRESOLS, GUAIACOL, AND SIMILAR SUBSTANCES IN PREPARATIONS FOR 
EXTERNAL USE.

    Caution--Do not apply to large areas of the body.

CREOSOTE, CRESOLS, GUAIACOL, AND SIMILAR SUBSTANCES IN DOUCHE 
PREPARATIONS.

    Warning--The use of solutions stronger than those recommended may 
result in severe local irritation, burns, or serious poisoning. Mix as 
directed before pouring into douche bag. Do not use more often than 
twice weekly unless directed by physician.

DENTURE RELINERS, PADS, AND CUSHIONS.

    Warning--For temporary use only. Long-term use of this product may 
lead to faster bone loss, continuing irritation, sores, and tumors. For 
Use Only Until a Dentist Can Be Seen.

DENTURE REPAIR KITS.

    Warning--For emergency repairs only. Long-term use of home-repaired 
dentures may cause faster bone loss, continuing irritation, sores, and 
tumors. This kit for emergency use only. See Dentist Without Delay.

DIARRHEA PREPARATIONS.

    Warning--Do not use for more than 2 days or in the presence of high 
fever or in infants or children under 3 years of age unless directed by 
a physician.

DOUCHE PREPARATIONS.

    Warning--Do not use more often than twice weekly unless directed by 
physician.
    See also Creosote * * * Douche for additional warning.

DRESSINGS, PROTECTIVE SPRAY-ON TYPE. (See also Sec. 310.201(a) (11) and 
(18) of this chapter.)

    Warning--In case of deep or puncture wounds or serious burns consult 
physician. If redness, irritation, swelling or pain persists or 
increases or if infection occurs consult physician. Keep away from eyes 
or other mucous membranes. Avoid inhaling.
    See also Dispensers Pressurized by Gaseous Propellants * * * for 
additional warnings to be included for products under pressure.

IODINE AND IODIDES (ORAL).

    Caution--If a skin rash appears, discontinue use and consult 
physician.

MERCURY PREPARATIONS FOR EXTERNAL USE.

    Warning--Discontinue use if rash or irritation develops or if 
condition for which used persists. Frequent or prolonged use, or 
application to large areas may cause serious mercury poisoning.

MINERAL OIL LAXATIVES. (See also Sec. 201.302 of this chapter.)

    Caution--Take only at bedtime. Avoid prolonged use. Do not 
administer to infants or young children, in pregnancy, or to bedridden 
or aged patients unless directed by physician.

NASAL PREPARATIONS: VASOCONSTRICTORS (PHENYL- PROPANOLAMINE).

    Caution--Do not exceed recommended dosage.

NUX VOMICA AND STRYCHNINE PREPARATIONS.

    Warning--Do not exceed the recommended dosage. Keep out of the reach 
of children.

OPHTHALMIC PREPARATIONS. (See also Sec. 200.50 of this chapter.)

    Boric acid offered for use in the preparation of ophthalmic 
solutions should bear the statement: Prepare solution by boiling in 
water. Store in a sterile container. Prepare sufficient for one day's 
use and discard unused portion.

PHENACETIN-CONTAINING PREPARATION. (See acetophenetidin.)

PHENYLPROPANOLAMINE HY- DROCHLORIDE PREPARATIONS, ORAL.

    Caution--Individuals with high blood pressure, heart disease, 
diabetes, or thyroid disease should use only as directed by physician.

POTASSIUM PERMANGANATE AQUEOUS SOLUTIONS (CONTAINING NOT MORE THAN 0.04 
PERCENT POTASSIUM PERMANGANATE). (See Sec. 250.108 of this chapter.)

    Warning--For external use on the skin only. Severe injury may result 
from use internally or as a douche. Avoid contact with mucous membranes.

[[Page 295]]


QUININE AND OTHER CINCHONA DERIVATIVES (EXCEPT FOR USE IN MALARIA).

    Caution--Discontinue use if ringing in the ears, deafness, skin 
rash, or visual disturbances occur.

RESINS, OLEORESINS, AND VOLATILE OILS.

    Caution--If nausea, vomiting, abdominal discomfort, diarrhea, or 
skin rash occurs, discontinue use and consult physician.

RESORCINOL (NOT THE MONOACETATE) HAIR PREPARATIONS.

    Caution--Excessive use of this preparation may temporarily discolor 
blond, white, or red hair.

SALICYLATES, INCLUDING ASPIRIN AND SALICYLAMIDE (EXCEPT METHYL 
SALICYLATE, EFFERVESCENT SALICYLATE PREPARATIONS, AND PREPARATIONS OF 
AMINOSALICYLIC ACID AND ITS SALTS). (See also Sec. 201.314 of this 
chapter.)

    Warning--Keep this and all medicines out of children's reach. In 
case of accidental overdose, contact a physician immediately; or
    Warning--Keep out of the reach of children.
    If the article is an aspirin preparation, it should bear the first 
of the above two warning statements. In either case, the above 
information should appear on the label.
    Caution--For children under 3 years of age, consult your physician; 
or
    Caution--For younger children, consult your physician.
    One of the two immediately preceding caution statements is required 
on the label of all aspirin tablets, but such a statement is not 
required on the labels of other salicylates clearly offered for 
administration to adults only.
    If offered for use in arthritis or rheumatism, in juxtaposition 
therewith, the statement:
    Caution--If pain persists for more than 10 days, or redness is 
present, or in conditions affecting children under 12 years of age, 
consult a physician immediately.

SALICYLATES: METHYL SALICYLATE (WINTERGREEN OIL). See also Secs. 201.303 
and 201.314 of this chapter.

    Warning--Do not use otherwise than as directed. Keep out of the 
reach of children to avoid accidental poisoning.
    If the preparation is a counterirritant or rubefacient the 
statement:
    Caution--Discontinue use if excessive irritation of the skin 
develops. Avoid getting into the eyes or on mucous membranes.
    If offered for use in arthritis or rheumatism, in juxtaposition 
therewith, the statement:
    Caution--If pain persists for more than 10 days, or redness is 
present, or in conditions affecting children under 12 years of age 
consult a physician immediately.

SILVER.

    Caution--Frequent or prolonged use of this preparation may result in 
permanent discoloration of skin and mucous membranes.

SODIUM PERBORATE MOUTHWASH AND GARGLE AND TOOTHPASTE.

    Caution--Discontinue use if irritation or inflammation develops, or 
increases. Avoid swallowing.

SULFONAMIDE NOSE DROPS.

    Caution--Do not use if a known allergy to sulfonamide drugs exists.

SULFUR PREPARATION FOR EXTERNAL USE.

    Caution--If undue skin irritation develops or increases, discontinue 
use and consult physician.

THROAT PREPARATIONS FOR TEMPORARY RELIEF OF MINOR SORE THROAT: LOZENGES, 
TROCHES, WASHES, GARGLES, ETC. (See also Sec. 201.315 of this chapter.)

    Warning--Severe or persistent sore throat or sore throat accompanied 
by high fever, headache, nausea, and vomiting may be serious. Consult 
physician promptly. Do not use more than 2 days or administer to 
children under 3 years of age unless directed by physician.

TOOTHACHE PREPARATIONS.

    For temporary use only until a dentist can be consulted.

ZINC STEARATE DUSTING POWDERS.


[[Page 296]]


    Warning--Keep out of the reach of infants and children; avoid 
inhaling.
[39 FR 11745, Mar. 29, 1974, as amended at 40 FR 8917, Mar. 3, 1975; 40 
FR 13496, Mar. 27, 1975; 41 FR 10885, Mar. 15, 1976; 51 FR 27760, Aug. 
1, 1986; 51 FR 35340, Oct. 2, 1986; 52 FR 15893, Apr. 30, 1987; 52 FR 
30057, Aug. 12, 1987; 52 FR 47324, Dec. 11, 1987; 53 FR 7093, Mar. 4, 
1988; 55 FR 31783, Aug. 3, 1990; 57 FR 58376, Dec. 9, 1992; 59 FR 43412, 
Aug. 23, 1994]



Sec. 369.21  Drugs; warning and caution statements required by regulations.

ACETAMINOPHEN (N-ACETYL-p-AMINOPHENOL) (See Sec. 310.201(a)(1) of this 
chapter.)

    Warning--Do not give to children under 3 years of age or use for 
more than 10 days unless directed by a physician.
    If offered for use in arthritis, or rheumatism, in juxtaposition 
therewith, the statement:
    Caution--If pain persists for more than 10 days, or redness is 
present, or in conditions affecting children under 12 years of age 
consult a physician immediately.

ALCOHOL RUBBING COMPOUND. (See 26 CFR 182.855(a)(5); The National 
Formulary, Tenth Edition 1955, pp. 27-28; and section 502(g) of the 
act).

    Warning--For external use only. If taken internally serious gastric 
distrubances will result.

ANTIHISTAMINICS, ORAL (PHENYLTOLOXAMINE DIHYDROGEN CITRATE AND 
CHLOROTHEN CITRATE PREPARATIONS). (See Sec. 310.201(a)(4) and (a)(24) of 
this chapter.)

    Caution--This preparation may cause drowsiness. Do not drive or 
operate machinery while taking this medication. Do not give to children 
under 6 years of age or exceed the recommended dosage unless directed by 
physician.
    If offered for symptoms of colds, the statement:
    Caution--If relief does not occur within 3 days, discontinue use and 
consult physician.

CARBETAPENTANE CITRATE PREPARATIONS. (See Cough-Due-to-Cold 
Preparations.)

``COUGH-DUE-TO-COLD'' PREPARATIONS (CARBETAPENTANE CITRATE). (See 
Sec. 310.201(a)(20) of this chapter.)

    Warning--Keep out of the reach of children. Do not administer to 
children under 2 years of age unless directed by physician. Persistent 
cough may indicate the presence of a serious condition. Persons with a 
high fever or persistent cough should not use this preparation unless 
directed by physician.

DICYCLOMINE HYDROCHLORIDE WITH AN ANTACID. (See Sec. 310.201(a)(8) of 
this chapter.)

    Warning--Do not exceed the recommended dosage. Do not administer to 
children under 12 years of age or use for a prolonged period unless 
directed by physician, since persistent or recurring symptoms may 
indicate a serious disease requiring medical attention.

DIPHEMANIL METHYLSULFATE FOR EXTERNAL USE. (See Sec. 310.201(a)(22) of 
this chapter.)

    Caution--If redness, irritation, swelling, or pain persists or 
increases, discontinue use and consult physician.

DRUGS IN DISPENSERS PRESSURIZED BY GASEOUS PROPELLANTS. (See also 
Sec. 310.201(a) (11) and (18) of this chapter.)

    The warnings herein shall appear prominently and conspicuously, but 
in no case may the letters be less than \1/16\ inch in height.
    If the label of any package is too small to accommodate the 
warnings, the Commissioner may establish by regulation an acceptable 
alternative method, e.g., a type size smaller than \1/16\ inch in 
height. A petition requesting such a regulation, as an amendment to this 
paragraph, shall be submitted to the Dockets Management Branch in the 
form established in part 10 of this chapter.
    Warning--Avoid spraying in eyes. Contents under pressure. Do not 
puncture or incinerate. Do not store at temperature above 120 deg. F. 
Keep out of reach of children.
    In the case of products packaged in glass containers, the word 
``break'' may be substituted for the word ``puncture.''
    The words ``Avoid spraying in eyes'' may be deleted from the warning 
in the case of a product not expelled as a

[[Page 297]]

spray, or that is intended to be used in the eyes.
    In addition to the above warning, the label of a drug packaged in a 
self-pressurized container in which the propellant consists in whole or 
in part of a halocarbon or hydrocarbon shall bear the following warning:
    Warning--Use only as directed. Intentional misuse by deliberately 
concentrating and inhaling the contents can be harmful or fatal.
    The warning is not required for the following products:
    (a) Products expelled in the form of a foam or cream, which contain 
less than ten percent propellant in the container;
    (b) Products in a container with a physical barrier that prevents 
escape of the propellant at the time of use;
    (c) Products of a net quantity of contents of less than 2 ozs. that 
are designed to release a measured amount of product with each valve 
actuation;
    (d) Products of a net quantity of contents of less than \1/2\ oz.

DYCLONINE HYDROCHLORIDE. (See Sec. 310.201(a)(23) of this chapter.)

    Caution--Do not use in the eyes. Not for prolonged use. Do not apply 
to large areas of the body. If redness, irritation, swelling, or pain 
persists or increases, discontinue use unless directed by physician. Do 
not use, but consult physician for deep or puncture wounds or serious 
burns. Do not use in case of rectal bleeding, as this may indicate 
serious disease.

HEXADENOL. (See Sec. 310.201(a)(11) of this chapter.)

    Caution--Do not use for treatment of serious burns or skin 
conditions or for conditions which persist for prolonged periods. In 
such cases, consult your physician. Do not spray in vicinity of eyes, 
mouth, nose, or ears. Do not store above 120 deg. F.

INSULIN. (See Sec. 429.11(c) of this chapter.)

Insulin (40 or 100 U.S.P. units per milliliter):

    Caution--Do not remove stopper. Not for intravenous nor 
intramuscular use. Do not use after expiration date shown on outside 
wrapper or container. Do not use if drug has become viscous or if its 
color has become other than water clear.
    In addition to the above warnings, the following statements should 
be included in the labeling: ``Keep in a cold place, avoid freezing. 
Failure to follow directions for use may lead to infection.'' Potamine 
zinc insulin, isophane insulin, lente insulin, semilente insulin, or 
ultralente insulin:
    Caution--Do not remove stopper. Not for intravenous nor 
intramuscular use. Do not use after expiration date shown on outside 
wrapper or container. Do not substitute for any other insulin-containing 
drug unless directed by physician. Do not use when precipitate has 
become lumped or granular in appearance or has formed a deposit of solid 
particles on the wall of the container.
    In addition to the above warnings for protamine zinc insulin * * *, 
the following statements should be included in the labeling of these 
preparations: ``Keep in a cold place, avoid freezing''; ``Shake 
carefully'' or ``Shake well before using'' or ``Shake well'' or ``Shake 
carefully to suspend all particles''; ``Failure to follow directions for 
use may lead to infection''.

Globin zinc insulin:
    Caution--Do not remove stopper. Not for intravenous nor 
intramuscular use. Do not use after expiration date shown on outside 
wrapper or container. Do not use if any turbidity or precipitate has 
developed in the solution. Do not substitute for any other insulin-
containing drug unless directed by physician.
    In addition to the above warnings for globin zinc insulin, the 
following statements should be included in the labeling: ``Keep in a 
cold place, avoid freezing. Failure to follow directions for use may 
lead to infection''.

IPECAC SYRUP IN ONE-FLUID OUNCE CONTAINERS FOR EMERGENCY TREATMENT OF 
POISONING, TO INDUCE VOMITING. (See Sec. 201.308 of this chapter.)

    Ipecac syrup packaged for over-the-counter sale must bear statements 
to the following effect, in a prominent and conspicuous manner:
    The following statement (boxed and in red letters):
    ``For emergency use to cause vomiting in poisoning. Before using, 
call

[[Page 298]]

physician, the Poison Control Center, or hospital emergency room 
immediately for advice.''
    The following warning: Warning--Keep out of reach of children. Do 
not use in unconscious persons. Ordinarily, this drug should not be used 
if strychnine, corrosives such as alkalies (lye) and strong acids, or 
petroleum distillates such as kerosene, gasoline, coal oil, fuel oil, 
paint thinner, or cleaning fluid have been ingested.

ISOAMYLHYRDOCUPREINE AND ZOLAMINE HYDROCHLORIDE RECTAL PREPARATIONS FOR 
EXTERNAL USE (See Sec. 310.201(a)(3) of this chapter.)

    Warning--Do not use this preparation in case of rectal bleeding, as 
this may indicate serious disease.

NEOMYCIN SULFATE WITH A VASOCONSTRICTOR, IN NASAL PREPARATIONS (SPRAY OR 
DROPS).

    Caution--Do not exceed recommended dosage. Do not administer to 
children under 3 years of age unless directed by physician.

PRAMOXINE HYDROCHLORIDE FOR EXTERNAL USE. (See Sec. 310.201(a)(19) of 
this chapter.)

    Caution--Do not use in the eyes or nose. Not for prolonged use. Do 
not apply to large areas of the body. If redness, irritation, swelling, 
or pain persists or increases, discontinue use unless directed by a 
physician.

SODIUM GENTISATE. (See Secs. 201.314, 310.201(a)(2) of this chapter.)

    Warning--Do not give to children under 6 years of age or use for 
prolonged period unless directed by physician.
    Warning--Keep this and all medications out of the reach of children; 
or
    Warning--Keep out of the reach of children.
    If offered for use in arthritis or rheumatism, in juxtaposition 
therewith, the statement:
    Caution--If pain persists for more than 10 days, or redness is 
present, or in conditions affecting children under 12 years of age, 
consult a physician immediately.

TUAMINOHEPTANE SULFATE NASAL PREPARATIONS. (See Sec. 310.201(a)(16) of 
this chapter.)

    Caution--Do not exceed recommended dosage. Overdosage may cause 
nervousness, restlessness, or sleeplessness. Individuals with high blood 
pressure, heart disease, diabetes, or thyroid disease should use only as 
directed by physician. Do not use for more than 3 or 4 consecutive days 
unless directed by physician.

VIBESATE PREPARATIONS. (See Sec. 310.201(a)(18) of this chapter.)

    Caution--Do not use but consult physician for deep or puncture 
wounds or serious burns. If redness, irritation, swelling, or pain 
persists or increases, discontinue use and consult physician.
    Warning--Contents under pressure. Do not puncture. Do not use or 
store near heat or open flame. Exposure to temperatures above 130 deg. 
Fahrenheit may cause bursting. Never throw container into fire or 
incinerator.
[39 FR 11745, Mar. 29, 1974, as amended at 40 FR 8917, Mar. 3, 1975; 40 
FR 13496, Mar. 27, 1975; 41 FR 10885, Mar. 15, 1976; 42 FR 22033, Apr. 
29, 1977; 42 FR 36994, July 19, 1977; 44 FR 22053, Apr. 13, 1979; 44 FR 
55170, Sept. 25, 1979; 52 FR 15893, Apr. 30, 1987; 52 FR 30057, Aug. 12, 
1987; 52 FR 47324, Dec. 11, 1987; 55 FR 11582, Mar. 29, 1990; 57 FR 
58376, Dec. 9, 1992; 59 FR 4218, Jan. 28, 1994; 61 FR 20101, May 3, 
1996]



Sec. 369.22  Drugs; warning and caution statements specifically required by law.

PREPARATIONS CONTAINING HABIT-FORMING DERIVATIVES OF SUBSTANCES NAMED IN 
SECTION 502(d) OF THE ACT. (See Secs. 329.1, 329.10, and 329.20 of this 
chapter.)

    The statement ``Warning--May be habit forming'' is required to 
appear on the labels of all drugs containing derivatives designated in 
Sec. 329.1 of this chapter as habit forming, including exempt narcotic 
preparations described in Sec. 329.20(a) of this chapter and 
preparations containing one or more derivatives of barbituric acid, 
unless such drug is not suitable for internal use and is distributed and 
sold exclusively for such external use as involves no possibility of 
habit formation.

[[Page 299]]



PART 429--DRUGS COMPOSED WHOLLY OR PARTLY OF INSULIN--Table of Contents




                      Subpart A--General Provisions

Sec.
429.3  Definitions and interpretations.

                    Subpart B--Packaging and Labeling

429.10  Packaging.
429.11  Labeling.
429.12  Distinguishing colors on packages.

                      Subpart C--Product Standards

429.25  Standards of quality and purity for protamine.
429.26  Standards of quality and purity for globin hydrochloride.

                      Subpart D--Tests and Methods

429.30  Tests and methods of assay.

                        Subpart E--Certification

429.40  Requests for certification; samples; storage; approvals 
          preliminary to certification.
429.41  Certifications.
429.45  Conditions on the effectiveness of certificates.
429.47  Authority to refuse certification service.

                  Subpart F--Administrative Procedures

429.50  Hearing procedure.
429.55  Fees.

                           Subpart G--Records

429.60  Records of distribution.

    Authority:  21 U.S.C. 352, 356, 371.

    Cross References: For other regulations in this chapter concerning 
insulin drugs, see also Sec. 200.15.

    Source:  39 FR 11750, Mar. 29, 1974, as amended at 40 FR 13497, Mar. 
27, 1975, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 429.3  Definitions and interpretations.

    For the purpose of the regulations in this part:
    (a) The term act means the Federal Food, Drug, and Cosmetic Act, as 
amended.
    (b) The term Secretary means the Secretary of Health and Human 
Services.
    (c) The term Commissioner means the Commissioner of Food and Drugs.
    (d) The term U.S.P. means the official United States Pharmacopeia, 
including supplements thereto.
    (e) The term N.F. means the official National Formulary, including 
supplements thereto.
    (f) The definitions and interpretations of terms contained in 
section 201 of the act shall be applicable to such terms when used in 
the regulations in this part.
    (g) The term insulin means the active principle of pancreas which 
affects the metabolism of carbohydrate in the animal body and which is 
of value in the treatment of diabetes mellitus.
    (h) The term insulin injection means the insulin injection 
recognized in the U.S.P.
    (i) The term protamine zinc insulin suspension means the protamine 
zinc insulin suspension recognized in the U.S.P.
    (j) The term globin zinc insulin injection means the globin zinc 
insulin injection recognized in the U.S.P.
    (k) The term isophane insulin suspension means the isophane insulin 
suspension recognized in the U.S.P.
    (l) The term insulin zinc suspension means the insulin zinc 
suspension recognized in the U.S.P.
    (m) The term prompt insulin zinc suspension means the prompt insulin 
zinc suspension recognized in the U.S.P.
    (n) The term extended insulin zinc suspension means the extended 
insulin zinc suspension recognized in the U.S.P.
    (o) The term master lot means a quantity (which is purified and 
which has been mixed in one container so as to be homogeneous) of:
    (1) A concentrated solution of insulin; or
    (2) The insulin-containing solids, in amorphous or crystalline form, 
derived from one or more such solutions.
    (p) Except as provided in Sec. 429.41(c), the term batch means a 
quantity of a drug, in labeled packages, of uniform composition and 
intended for administration without further change, in which the sole 
insulin-containing ingredient is a single dilution (which has been mixed 
in one container so as to be homogeneous) of:
    (1) A single master lot or part thereof; or

[[Page 300]]

    (2) A mixture of two or more master lots or parts thereof; except 
that such term means a portion of such quantity when certification of 
such portion is requested.
    (q) The term master lot mark means an identifying mark or other 
identifying device assigned to a master lot by the manufacturer thereof.
    (r) The term batch mark means an identifying mark or other 
identifying device assigned to a batch by the manufacturer thereof.
[39 FR 11750, Mar. 29, 1974, as amended at 39 FR 40285, Nov. 15, 1974]



                    Subpart B--Packaging and Labeling



Sec. 429.10  Packaging.

    Each batch shall be packaged in immediate containers of colorless 
transparent glass. Such containers shall be closed with a substance 
through which successive doses may be withdrawn by hypodermic needle 
without removing the closure or destroying its effectiveness. The 
containers and closures shall be sterile at the time the containers are 
filled and closed. The composition of the containers and closures shall 
be such as will not cause any change in the strength, quality, or purity 
of the contents beyond any limit therefor prescribed in applicable 
standards of strength, quality, and purity. The shape of the containers 
shall be cylindrical, except that the cross-section of the containers 
for isophane insulin suspension containing less than 100 U.S.P. Units of 
insulin per milliliter shall be a rounded square, and the shoulder of 
the containers for insulin zinc suspension, prompt insulin zinc 
suspension, or extended insulin zinc suspension containing less than 100 
U.S.P. Units of insulin per milliliter shall be hexagonal.
[39 FR 11750, Mar. 29, 1974, as amended at 39 FR 40285, Nov. 15, 1974]



Sec. 429.11  Labeling.

    Each package from a batch that has been certified in accordance with 
the regulations in this part shall bear, on its label or labeling as 
hereinafter indicated, the following:
    (a) On the outside wrapper or container and the immediate container 
of the retail package:
    (1) The batch mark of such batch;
    (2) The potency of the drug in terms of the U.S.P. Units of insulin 
per milliliter; and
    (3) The statement ``Expiration date --------,'' the blank being 
filled in with the date on which the certificate applicable to such 
batch expires with respect to such package, as provided in 
Sec. 429.45(b)(1).
    (b) On the outside container or wrapper of the retail package, the 
statement ``Keep in a cold place, avoid freezing.''
    (c) If the batch contains 40 or 100 U.S.P. Units of insulin per 
milliliter, on the circular or other labeling of the retail package:
    (1) A statement that the treatment of diabetes mellitus is an 
individual problem and that the use of the drug, the time of its 
administration, and the number of daily doses and the quantity of each, 
as well as diet and exercise, are problems which require direct and 
continuous medical supervision;
    (2) A statement explaining that the volume of the dose depends on 
the number of units of insulin per milliliter stated on the label, and 
that the patient should understand the meaning of the volume markings on 
the syringe;
    (3) A description of a practicable method for sterilizing the needle 
and syringe before use;
    (4) A description of the technique of withdrawal from the vial and 
the use of an antiseptic on the stopper, and a caution against the 
removal of the stopper;
    (5) A description of the technique for cleansing, and the use of an 
antiseptic on the site of injection;
    (6) A statement that failure to comply with the techniques described 
in paragraphs (c) (3), (4), and (5) of this section may lead to 
infection of the patient;
    (7) A statement that injection should be subcutaneous, at a 
different site from that of the preceding injection, and a caution 
against intravenous or intramuscular use;
    (8) An explanation of hypoglycemia and its relation to overdosage, 
omission of meals, illness, and infection;

[[Page 301]]

    (9) A statement of the significance of sugar in the urine and of the 
necessity of tests therefor; and
    (10) A caution against use after the expiration date shown on the 
outside wrapper or container.
    (d) On the circular or other labeling of the retail package, if the 
batch is insulin injection (in addition to the information required by 
paragraphs (a), (b), and (c) or (i) of this section), a caution against 
use if the drug has become viscous or if its color has become other than 
water clear.
    (e) On the outside wrapper or container and immediate container of 
the retail package, if the batch is protamine zinc insulin suspension, 
isophane insulin suspension, insulin zinc suspension, prompt insulin 
zinc suspension, or extended insulin zinc suspension (in addition to the 
information required by paragraphs (a), (b), and (c) of this section), 
the statement ``Shake carefully,'' or ``Shake well before using,'' or 
``Shake well,'' or ``Shake carefully to suspend all particles.''
    (f) On the circular or other labeling of the retail package, if the 
batch is protamine zinc insulin suspension, isophane insulin suspension, 
insulin zinc suspension, prompt insulin zinc suspension, or extended 
insulin zinc suspension (in addition to the information required by 
paragraphs (a), (b), (c), and (e) of this section):
    (1) An explanation of the difference, as compared with other 
insulin-containing drugs, in onset of action, duration, and the time and 
frequency of administration;
    (2) A caution that it is not to be substituted for any other 
insulin-containing drug except on the advice and direction of a 
physician;
    (3) A statement that a uniform suspension of the preparation is 
necessary and is brought about by careful shaking before use; and
    (4) A caution against use when the precipitate has become lumped or 
granular in appearance or has formed a deposit of solid particles on the 
wall of the container.
    (g) On the circular or other labeling of the retail package, if the 
batch is globin zinc insulin injection (in addition to the information 
required by paragraphs (a), (b), and (c) of this section):
    (1) An explanation of the difference, as compared with other 
insulin-containing drugs, in onset of action, duration, and the time and 
frequency of administration;
    (2) A caution that it is not to be substituted for any other 
insulin-containing drug, except on the advice and direction of a 
physician; and
    (3) A caution against use if any turbidity or precipitate has 
developed in the solution.
    (h) If the batch contains 500 U.S.P. Units of insulin per 
milliliter, on the outside container or wrapper and the immediate 
container of the retail package:
    (1) The statement ``Caution: Federal law prohibits dispensing 
without prescription''; 2 and
---------------------------------------------------------------------------

    2 For the Spanish-language version of the required 
labeling statement, see Sec. 201.16(a), Sec. 801.16 and Sec. 290.6 of 
this chapter.
---------------------------------------------------------------------------

    (2) The statement ``Warning--High potency--Not for ordinary use''.
    (i) If the batch contains 500 U.S.P. Units of insulin per 
milliliter, on the circular or other labeling of the retail package:
    (1) Information adequate for the safe and effective use of the drug, 
by practitioners licensed by law to administer it, in insulin shock 
therapy and for the treatment of diabetic patients with high insulin 
resistance (daily requirement more than 200 units);
    (2) A prominently placed and conspicuous statement: ``Warning--This 
insulin preparation contains 500 units of insulin in each milliliter. 
Extreme caution must be observed in measurement of dosage because 
inadvertent overdose may result in irreversible insulin shock. Serious 
consequences may result if it is used other than under constant medical 
supervision'';
    (3) A caution against intravenous use; and
    (4) A caution against use after the expiration date shown on the 
outside wrapper or container.
[39 FR 11750, Mar. 29, 1974, as amended at 40 FR 13497, Mar. 27, 1975; 
41 FR 6912, Feb. 13, 1976; 44 FR 55170, Sept. 25, 1979]

[[Page 302]]



Sec. 429.12  Distinguishing colors on packages.

    (a) The outside containers or wrappers of the packages, and the 
labels on the immediate containers of each potency of insulin injection 
shall be distinguished by the following colors:

    Red, if it contains 40 U.S.P. Units of insulin per milliliter.
    White, if it contains 100 U.S.P. Units of insulin per milliliter.
    Narrow (at least 5 but not more than 20 to each inch) brown and 
white diagonal stripes, if it contains 500 U.S.P. Units of insulin per 
milliliter.


But if the master lot used was in crystalline form, the distinguishing 
colors, instead of those prescribed above, may be the following:

    Red and gray, if it contains 40 U.S.P. Units of insulin per 
milliliter.

    (b) The outside containers or wrappers of the packages, and the 
labels on the immediate containers of each potency of protamine zinc 
insulin suspension shall be distinguished by the following colors:

    Red and white, if it contains 40 U.S.P. Units of insulin per 
milliliter.
    Black and white, if it contains 100 U.S.P. Units of insulin per 
milliliter.

    (c) The outside containers or wrappers of the packages, and the 
labels of the immediate containers of each potency of globin zinc 
insulin injection shall be distinguished by the following colors:

    Red and brown, if it contains 40 U.S.P. Units of insulin per 
milliliter.
    Black and white, if it contains 100 U.S.P. Units of insulin per 
milliliter.

    (d) The outside containers or wrappers of the packages, and the 
labels of the immediate containers of each potency of isophane insulin 
suspension shall be distinguished by the following colors:

    Red and blue, if it contains 40 U.S.P. Units of insulin per 
milliliter.
    Black and white, if it contains 100 U.S.P. Units of insulin per 
milliliter.

    (e) The outside containers or wrappers of the packages, and the 
labels of the immediate containers, of insulin zinc suspension, prompt 
insulin zinc suspension, and extended insulin zinc suspension shall bear 
a mark or design to distinguish each drug, and each potency of these 
drugs shall be distinguished by the following colors:

    Red and lavender, if it contains 40 U.S.P. Units of insulin per 
milliliter.
    Black and white, if it contains 100 U.S.P. Units of insulin per 
milliliter.
[39 FR 11750, Mar. 29, 1974, as amended at 39 FR 40286, Nov. 15, 1974; 
44 FR 55170, Sept. 25, 1979]



                      Subpart C--Product Standards



Sec. 429.25  Standards of quality and purity for protamine.

    When protamine is dried to constant weight at 100 deg. C., its total 
nitrogen content is not less than 22.5 percent and not more than 25.5 
percent, and its sulfate content, calculated as SO4, is not 
less than 16 percent and not more than 19 percent.



Sec. 429.26  Standards of quality and purity for globin hydrochloride.

    The ash content of globin hydrochloride is not more than 0.3 
percent; its nitrogen content, calculated to moisture, ash, and 
hydrochloric acid free basis, is not less than 16.0 percent and not more 
than 17.5 percent.



                      Subpart D--Tests and Methods



Sec. 429.30  Tests and methods of assay.

    The following tests and methods of assay are prescribed for the 
purposes of the regulations in this part 429. (All reagents specified in 
this section shall be of U.S.P. quality or better.)
    (a) Tests and methods of assay for insulin injection, protamine zinc 
insulin suspension, globin zinc insulin injection, isophane insulin 
suspension, insulin zinc suspension, prompt insulin zinc suspension, and 
extended insulin zinc suspension. The tests and methods of assay for 
insulin injection, protamine zinc insulin suspension, globin zinc 
insulin injection, isophane insulin suspension, insulin zinc suspension, 
prompt insulin zinc suspension, and extended insulin zinc suspension 
shall be those set forth therefor in the U.S.P. or N.F., except that 
alternative test procedures may be employed when such have been 
authorized by the Commissioner.
    (b) [Reserved]

[[Page 303]]

    (c) Isophane ratio. The isophane ratio shall be expressed as 
milligrams of protamine per 100 U.S.P. Units of insulin.
    (1) Reagents--(i) The stock buffer solution. Dissolve in water the 
quantities of metacresol, phenol, glycerin, and disodium phosphate 
required to make 10 liters of the batch of isophane insulin and dilute 
to 1,000 milliliters.
    (ii) The insulin solution. From a sample of the zinc-insulin 
crystals to be used in making the batch weigh a quantity which contains 
10,000 U.S.P. Units of insulin. Dissolve the crystals in 15 milliliters 
of 0.1 percent hydrochloric acid. The resulting solution must be clear. 
Add it to 25 milliliters of the stock buffer solution (paragraph 
(c)(1)(i) of this section). Dilute with water to approximately 200 
milliliters. Adjust the pH to 7.2 using hydrochloric acid or sodium 
hydroxide. The solution must be clear at this stage. If sodium chloride 
is to be used in preparing the batch add 25 milliliters of 4.2 percent 
(w/v) sodium chloride solution. Dilute to 250 milliliters with water. 
The pH must be between 7.1 and 7.4.
    (iii) The protamine solution. Weigh 500 milligrams of the protamine 
to be used in making the batch and dissolve in 10 milliliters of the 
stock buffer solution (paragraph (c)(1)(i) of this section). If sodium 
chloride is to be used in preparing the batch add 10 milliliters of 4.2 
percent (w/v) sodium chloride solution. Dilute with water to 
approximately 80 milliliters. Adjust the pH to 7.2 using hydrochloric 
acid or sodium hydroxide. Dilute with water to 100 milliliters. The pH 
must be between 7.2 and 7.4 and the solution must be clear.
    (2) Conduct of the test. Measure six 25-milliliter samples of the 
insulin solution (paragraph (c)(1)(ii) of this section) into six tubes. 
To the first tube add 0.60 milliliter of the protamine solution 
(paragraph (c)(1)(iii) of this section), to the second add 0.72 
milliliter, to the third add 0.84 milliliter, to the fourth add 0.96 
milliliter, to the fifth add 1.08 milliliters, and to the sixth add 1.20 
milliliters. Mix the contents of each tube and let stand for at least 30 
minutes. Centrifuge. (Do not filter.) From each supernatant fluid remove 
two 10-milliliter samples, thus creating two series of samples. To each 
of one series add 1 milliliter of the insulin solution (paragraph 
(c)(1)(ii) of this section). To each of the other series add 1 
milliliter of the protamine solution (paragraph (c)(1)(iii) of this 
section). Mix each sample and let stand 10 minutes. Measure the 
turbidity of each sample by means of a photometer or nephelometer. Plot 
the readings of the two series of samples, using the amount of protamine 
originally added in milligrams per 100 U.S.P. Units of insulin as 
abscissas, and the photometer or nephelometer readings as ordinates. The 
abscissa of the intersection of the two curves indicates the isophane 
ratio of the protamine to the zinc-insulin crystals. In order to 
increase the precision of the test, when the approximate isophane ratio 
is known, the quantities of protamine solution to be added to the six 
tubes may be so chosen that the range (0.60 to 1.20 milliliters) is 
reduced, and the approximate isophane ratio is near the middle of the 
range.

The isophane ratio found is not more than 100 percent nor less than 90 
percent of the ratio of protamine to insulin used in the trial mixture 
referred to in Sec. 429.40(d)(7).
    (d)-(e) [Reserved]
    (f) Chloride in globin hydrochloride--(1) Conduct of the test. Weigh 
accurately approximately 0.5 gram of globin hydrochloride into a small 
beaker and dissolve in 10-15 milliliters of distilled water. Add 10 
milliliters of tenth-normal silver nitrate, 5 milliliters of nitric 
acid, and 5 milliliters of a saturated solution of potassium 
permanganate. Stir and place on a steam bath for approximately 1 hour. 
If any brown color remains, stir again, rinse the sides of the beaker 
with distilled water and place on the steam bath until the brown color 
disappears. Transfer quantitatively to a 50-milliliter volumetric flask 
and fill the flask to the mark with distilled water. Mix and filter 
through a dry filter paper into a dry vessel. Transfer exactly 40 
milliliters of the filtrate to a flask, add 2 milliliters of ferric 
ammonium sulfate test solution and titrate with tenth-normal ammonium 
thiocyanate. To obtain the percent chloride as HCl, subtract 1.25 times 
the number of milliliters of ammonium thiocyanate used from 10;

[[Page 304]]

multiply this difference by 0.365 and divide by the weight of the sample 
in grams.
    (2) Reagents. The reagents used are those described in the U.S.P.
    (g) Sulfate in protamine--(1) Conduct of the test. Weigh accurately 
about 250 milligrams of protamine and dissolve it in about 100 
milliliters of approximately tenth-normal hydrochloric acid. Heat to 
boiling and add 5 milliliters of barium chloride test solution. Digest 
on a steam bath for 1 hour; allow to cool. Filter through an ignited and 
weighed Gooch crucible; wash free of chlorides. Dry, ignite, and weight. 
The weight of barium sulfate thus obtained multiplied by 41.15 and 
divided by the weight of sample is the percent sulfate (SO4) 
in the sample. Calculate the results to a moisture-free basis.
    (2) Reagents. The reagents used are those described in the U.S.P.
    (h) Nitrogen. Determine total nitrogen by the method described in 
the U.S.P., for insulin U.S.P.
    (i) Zinc in insulin-containing solutions or suspensions. Use the 
method described in the U.S.P. for insulin injection.
    (j) Zinc in insulin-containing solids. Dissolve 10 to 20 milligrams, 
accurately weighed, of insulin-containing solids in 5 to 10 milliliters 
of distilled water containing one drop of 5N hydrochloric acid, and 
proceed as directed in the U.S.P. under the test for zinc in insulin 
injection.
[39 FR 11750, Mar. 29, 1974, as amended at 39 FR 40286, Nov. 15, 1974]



                        Subpart E--Certification



Sec. 429.40  Requests for certification; samples; storage; approvals preliminary to certification.

    (a) A request for certification of a batch is to be addressed to the 
Food and Drug Administration, Division of Research and Testing (HFD-
470), 200 C St. SW., Washington, DC 20204.
    (b) The initial request for certification submitted by any person 
shall be preceded or accompanied by a full statement of the facilities 
and controls used to maintain the identity, strength, quality, and 
purity of each batch, including a description of:
    (1) The equipment, methods, and processes used in diluting master 
lots and parts thereof, and in maintaining the identity, strength, 
quality, and purity of master lots and dilutions therefrom;
    (2) The tests and assays made on master lots and mixtures thereof, 
on dilutions and batches therefrom, and on ingredients used in such 
dilutions and batches; and
    (3) The laboratory facilities used in such controls.

Such initial request shall also be preceded or accompanied by the keys 
to the master lot marks and batch marks used by such person. When any 
change is made in any of such facilities or controls, or in any such 
key, the next request for certification thereafter shall be accompanied 
by a full statement of such change.
    (c) A person who requests certification of a batch shall submit in 
connection with his request statements showing:
    (1) The master lot mark of each master lot used or to be used wholly 
or partly as an ingredient or component of an ingredient of the batch;
    (2) The quantity of each such master lot so used;
    (3) The original quantity of each such master lot (unless such 
information has been previously submitted);
    (4) The quantity of the batch; and
    (5) The batch mark.
    (d) Except as otherwise provided in paragraphs (g) and (h) of this 
section, a person who requests certification of a batch shall submit in 
connection with his request and in the quantities hereinafter indicated, 
accurately representative samples of the following:
    (1) The single master lot or the mixture of two or more master lots 
or parts thereof, to be used as ingredients of the batch; in a quantity 
containing approximately 10,000 U.S.P. Units of insulin, except that, if 
the batch is to be isophane insulin suspension, the quantity shall 
contain not less than 20,000 U.S.P. Units of insulin.
    (2) If the batch is to be insulin injection, a trial dilution made 
from such master lot or mixture, glycerin, phenol or cresol, and 
hydrochloric acid, which dilution conforms to the standard of identity, 
strength, quality, and purity

[[Page 305]]

for insulin injection, except that it may contain approximately 40, 80, 
or 100 units of insulin per milliliter in a quantity containing 
approximately 5,000 U.S.P. units of insulin.
    (3) If the batch is to be protamine zinc insulin suspension, a trial 
mixture which is intended to be accurately representative of the mixture 
which will constitute the finished batch; in a quantity containing 
approximately 10,000 U.S.P. units of insulin.
    (4) If the batch is to be protamine zinc insulin suspension or 
isophane insulin suspension, the lot of protamine used as an ingredient 
of the trial mixture referred to in paragraph (d)(3) or (7) of this 
section; in a quantity of approximately 2 grams.
    (5) If the batch is to be globin zinc insulin injection, a trial 
mixture made from the master lot or mixture referred to in paragraph 
(d)(1) of this section, globin, zinc chloride, hydrochloric acid, 
glycerin, and phenol or cresol, which mixture is intended to be 
accurately representative of the mixture which will constitute the 
finished batch; in a quantity containing approximately 10,000 U.S.P. 
units of insulin.
    (6) If the batch is to be globin zinc insulin injection, the lot of 
globin hydrochloride from which the globin is to be prepared for use as 
an ingredient of the trial mixture referred to in paragraph (d)(5) of 
this section; in a quantity of approximately 5 grams.
    (7) If the batch is to be isophane insulin suspension, a trial 
mixture which is intended to be accurately representative of the 
finished batch; in a quantity of approximately 10,000 U.S.P. units of 
insulin.
    (8) If the batch is to be insulin zinc suspension, prompt insulin 
zinc suspension, or extended insulin zinc suspension, a trial mixture 
which is intended to be accurately representative of the finished batch; 
in a quantity of approximately 50 milliliters.
    (9) The finished batch; for all tests except sterility, not less 
than 10 retail packages.
    (10) The finished batch for sterility testing, 20 retail packages, 
collected at approximately equal intervals throughout each filling 
operation (as defined by the U.S.P.), except that if it is insulin 
injection containing 500 U.S.P. Units of insulin per milliliter, in lieu 
of the volume contained in the retail package each such container may 
contain an amount of drug that is less than that contained in the retail 
package but in no case less than 5 milliliters.
    (e) Except as otherwise provided by paragraphs (g) and (h) of this 
section, a person who requests certification shall submit in connection 
with his request results of the tests and assays listed after each of 
the following materials, made by him on a sample of such material:
    (1) The master lot or mixture, referred to in paragraph (d)(1) of 
this section: Ash, nitrogen, potency, pH, sterility, and zinc, if such 
master lot or mixture is a solution; ash, moisture, nitrogen, potency, 
and zinc, if such master lot or mixture is a solid.
    (2) A trial dilution of such master lot or mixture, of the potency 
of the trial dilution referred to in paragraph (d)(2) of this section: 
Nitrogen, pH, and potency.
    (3) If the batch is to be protamine zinc insulin suspension, the 
trial mixture referred to in paragraph (d)(3) of this section: Nitrogen, 
pH, zinc, and biological reaction (by the test prescribed in the 
U.S.P.).
    (4) If the batch is to be protamine zinc insulin suspension or 
isophane insulin suspension, the protamine referred to in paragraph 
(d)(4) of this section: Moisture, nitrogen, and sulfate.
    (5) If the batch is to be globin zinc insulin injection the trial 
mixture referred to in paragraph (d)(5) of this section: Nitrogen, pH, 
zinc, and biological reaction (by the test prescribed in the U.S.P.).
    (6) If the batch is to be globin zinc insulin injection, the globin 
hydrochloride referred to in paragraph (d)(6) of this section: Moisture, 
nitrogen, chloride, and ash.
    (7) If the batch is to be isophane insulin suspension, the trial 
mixture referred to in paragraph (d)(7) of this section: Nitrogen, pH, 
zinc, isophane ratio of the protamine to the master lot or mixture (by 
the test prescribed in Sec. 429.30(c)), and biological activity of the 
supernatant liquid (by the test prescribed in the U.S.P.).

[[Page 306]]

    (8) If the batch is to be insulin zinc suspension, prompt insulin 
zinc suspension, or extended insulin zinc suspension, the trial mixture 
referred to in paragraph (d)(8) of this section: Nitrogen, pH, zinc, 
zinc in the supernatant liquid and insulin not extracted by buffered 
acetone solution.
    (9) The finished batch: Nitrogen, pH, sterility; and if the batch is 
protamine zinc insulin suspension, globin zinc insulin injection, 
isophane insulin suspension, insulin zinc suspension, prompt insulin 
zinc suspension, or extended insulin zinc suspension, zinc.
    (f) The results of tests and assays for the following shall be 
reported in the terms indicated:
    (1) Ash (except globin hydrochloride)--milligrams per 1,000 U.S.P. 
Units of insulin.
    (2) Ash in globin hydrochloride--percent by weight.
    (3) Chloride--percent by weight as HCl.
    (4) Insulin not extracted by buffered acetone solution--percent of 
total nitrogen of the preparation not extracted by buffered acetone 
solution.
    (5) Isophane ratio--milligrams of protamine per 100 U.S.P. Units of 
insulin.
    (6) Moisture--percent by weight.
    (7) Nitrogen (except in globin hydrochloride and protamine)--
milligrams per milliliter in the cases of solutions and suspensions, and 
percent by weight in the case of solids.
    (8) Nitrogen in globin hydrochloride--percent by weight, calculated 
to a moisture-free, ash-free, chloride-free basis.
    (9) Nitrogen in protamine--percent by weight, calculated to a 
moisture-free basis.
    (10) Potency--U.S.P. Units of insulin per milliliter in the case of 
solutions, and U.S.P. Units of insulin per milligram in the case of 
solids.
    (11) pH.
    (12) Sulfate--percent by weight as SO4, calculated to a 
moisture-free basis.
    (13) Zinc--milligrams per milliliter in the cases of solutions and 
suspensions, and percent by weight in the case of solids.
    (g)(1) No sample referred to in paragraphs (d) (1) to (3), 
inclusive, of this section, and no result referred to in paragraphs (c) 
(1) to (8), inclusive of this section, is required if such sample or 
result has been submitted in connection with a previous request for 
certification. Except for paragraphs (d) (9), (10), and (e)(9), the 
samples referred to in paragraph (d) of this section and the results 
referred to in paragraph (e) of this section for insulin injection, 
protamine zinc insulin suspension, globin zinc insulin injection, or 
isophane insulin suspension are not required if the Commissioner has 
previously approved a trial mixture containing 40, 100 units of insulin 
per milliliter or trial dilution containing approximately 40, 100 units 
of insulin per milliliter and the mixture or dilution was prepared from 
the same materials and in the same manner, except for adjustment of pH 
of the buffer solution.
    (2) Each sample submitted pursuant to this section shall be so 
packaged as to maintain its representative character, and in the case of 
any solution or suspension, shall be collected and packaged under 
aseptic conditions. Each package shall be clearly identified as to its 
contents and shall bear the name and post office address of the person 
submitting the request.
    (3) The packages constituting the samples submitted pursuant to 
paragraph (d)(9) of this section shall be collected at such intervals 
that the quantities packaged between collections are approximately 
equal; in no case shall any such quantity be more than 10,000 packages. 
The collections shall cover the entire period of packaging.
    (4) Each sample submitted pursuant to paragraphs (d) (2), (3), (5), 
(7), and (8) of this section shall be accompanied by a statement showing 
the identity, quality, and quantity of each substance used as an 
ingredient or as a component of an ingredient in the material from which 
the sample was taken.
    (5) If the tests and assays, results of which are submitted pursuant 
to paragraph (e)(2) of this section, were not made on the same trial 
dilution as that from which the sample submitted pursuant to paragraph 
(d)(2) of this section was taken, such sample shall be accompanied by a 
statement showing the identity, quality, and quantity of each substance 
used as an ingredient or as a

[[Page 307]]

component of an ingredient of the trial dilution on which such tests and 
assays were made.
    (6) The value for nitrogen submitted pursuant to paragraphs (e) (1) 
and (2) of this section may be calculated from the result of a test 
therefor submitted pursuant to either paragraph (e) (1) or (2) of this 
section. The result on potency required under paragraph (e)(1) of this 
section may be calculated from an assay therefor submitted pursuant to 
paragraph (e)(2) of this section. The value of each of the components 
nitrogen and zinc, to the extent required under paragraph (e)(9) of this 
section, may be calculated from the result of a test therefor submitted 
pursuant to paragraph (e) (3), or (5), or (7) or (8) of this section or 
from the result of a test of the bulk dilution from which the batch was 
prepared. The value for nitrogen required under paragraph (e)(9) of this 
section may, if the batch is insulin injection, insulin zinc suspension, 
prompt insulin zinc suspension, or extended insulin zinc suspension, be 
calculated from a test therefor submitted pursuant to either paragraph 
(e) (1) or (2) of this section. Each calculated value shall be indicated 
as such.
    (7) The information required under paragraphs (c) (1), (2), and (3) 
of this section, and the samples and results of tests and assays 
required under paragraphs (d) (1) and (2) and (e) (1) and (2) of this 
section, should be submitted before submission of the samples and 
results required in paragraphs (d) (3) to (8), inclusive, of this 
section and (e) (3) to (8), inclusive, of this section; and the samples 
and results required under paragraphs (d) (3) to (8), inclusive, and (e) 
(3) to (8), inclusive, should be submitted before submission of the 
information, samples, and results required under paragraphs (c) (4) and 
(5), (d) (9) and (10), and (e)(9) of this section. All information, 
including results of tests and assays (except results of tests for 
sterility), required under this section should be submitted at the same 
time as the samples to which they relate are submitted.
    (h) The person who requests certifications shall submit such 
information additional to that submitted pursuant to paragraphs (b), 
(c), (e), and (g) of this section, such additional samples of any 
substance referred to in paragraph (d) of this section, and such samples 
of any other substance used or to be used as an ingredient or as a 
component of an ingredient in the batch, as the Commissioner may require 
for the purpose of investigations to determine whether or not such batch 
complies with the requirements set forth by Sec. 429.41 for the issuance 
of a certificate.
    (i) After a sample required by paragraph (d) of this section is 
taken from any master lot or mixture of part of two or more master lots, 
such master lot or master lots and all parts thereof, and all dilutions 
and batches and all parts thereof in which any such master lot is used 
as an ingredient or as a component of an ingredient, shall be stored at 
the establishment where manufactured until used up or shipped or 
otherwise delivered, at a temperature above freezing but not above 
15 deg. C. (59 deg. F.), and under such other conditions as prevent, so 
far as practicable, any change in composition; except that master lots 
and parts thereof which are solids may be stored at ordinary room 
temperatures.
    (j) As promptly as practicable after the samples submitted pursuant 
to paragraphs (d) (1) and (2) of this section, and any other material or 
information relative thereto that may be required under this section, 
are received by the Commissioner, he shall notify the person who 
submitted such samples of his approval or refusal to approve the use of 
the master lot or mixture for the making of bulk dilutions. In case of a 
refusal to approve, the Commissioner shall state his reasons therefor.
    (k) In like manner, the Commissioner shall notify the person who 
submits samples pursuant to paragraphs (d) (3) to (8), inclusive, of 
this section of his approval or refusal to approve the use of the 
materials represented by such samples in completing the manufacture of 
the batch. In case of a refusal to approve, the Commissioner shall state 
his reasons therefor.
    (l) If, under the provisions of paragraph (j) or (k) of this 
section, the Commissioner has refused to approve any material for use in 
a subsequent operation, he shall examine no other

[[Page 308]]

sample required hereunder which includes such material as an ingredient 
or component of an ingredient, unless and until the person requesting 
certification makes an adequate showing that the cause for such refusal 
no longer exists.
[39 FR 11750, Mar. 29, 1974, as amended at 39 FR 40286, Nov. 15, 1974; 
44 FR 48968, Aug. 21, 1979; 44 FR 55170, Sept. 25, 1979; 45 FR 40111, 
June 13, 1980; 50 FR 8996, Mar. 6, 1985; 55 FR 11582, Mar. 29, 1990]



Sec. 429.41  Certifications.

    (a) If it appears to the Commissioner, after such investigation as 
he considers necessary, that:
    (1) The information (including results of tests and assays) and the 
samples required by or pursuant to Sec. 429.40 have been submitted, and 
such information contains no untrue statement of a material fact;
    (2) The batch complies with the regulations in this part 429 and 
conforms to the standards of identity, quality, strength, and purity for 
insulin injection, protamine zinc insulin suspension, globin zinc 
insulin injection, isophane insulin suspension, insulin zinc suspension, 
prompt insulin zinc suspension, or extended insulin zinc suspension;

the Commissioner shall certify that such batch is safe and efficacious 
for use, subject to such conditions on the effectiveness of such 
certifications as are set forth in Sec. 429.45, and shall issue to the 
person who requested it a certificate to that effect.
    (b) If the Commissioner determines, after such investigation as he 
considers to be necessary, that the information submitted pursuant to 
Sec. 429.40 or the batch covered by such request, does not comply with 
the requirements set forth in paragraph (a) of this section for the 
issuance of a certificate, the Commissioner shall refuse to certify such 
batch and shall give notice thereof to the person who requested 
certification, stating his reasons for refusal.
    (c) Upon the request of the manufacturer, the Commissioner shall 
certify as a ``batch'' a master lot, which has been approved in 
accordance with Sec. 429.40(j) as safe and efficacious for use in 
preparation of an insulin-containing drug, subject to the conditions on 
the effectiveness of such certifications as are set forth in 
Sec. 429.45(a) (1) and (b) (4).
    (d) For the purposes of his investigations under the authority of 
this section, the Commissioner may accept, when he is satisfied as to 
the completeness and accuracy thereof, the results of any tests or 
assays made by the control laboratory of the Insulin Committee of the 
University of Toronto.



Sec. 429.45  Conditions on the effectiveness of certificates.

    (a) A certificate shall not become effective:
    (1) If it is obtained through fraud, or through misrepresentation or 
concealment of a material fact.
    (2) With respect to any package, unless its immediate container 
complies with the requirements of Sec. 429.10 and such package or such 
immediate container has been so sealed that its contents cannot be used 
without destroying such package or seal.
    (3) With respect to any package, unless its label and labeling bear 
all words, statements, and other information, and are distinguished by 
the color or colors, required by Secs. 429.11 and 429.12.
    (b) A certificate shall cease to be effective: (1) With respect to 
any package of insulin injection, protamine zinc insulin suspension, 
globin zinc insulin injection, isophane insulin suspension, insulin zinc 
suspension, prompt insulin zinc suspension, or extended insulin zinc 
suspension on the expiration date specified in the U.S.P.
    (2) With respect to any package, when such package or the seal 
thereof or the immediate container therein or the seal of the immediate 
container is broken, or when its label or labeling ceases to conform to 
any requirement of Sec. 429.11 or Sec. 429.12.
    (3) With respect to any package, when the drug therein so changes 
that it fails to meet the standards of identity, strength, quality, and 
purity upon the basis of which the batch was certified; except that 
those minor changes in potency (not exceeding 10 percent from the 
potency stated on the label, in the case of insulin injection) which 
occur before the expiration date, and which are normal and unavoidable 
in

[[Page 309]]

good storage and distribution practice, shall be disregarded.
    (4) With respect to a master lot of insulin, 5 years after date of 
issue if the master lot is a solution, or 10 years after date of issue 
if the master lot is a solid.
[39 FR 11750, Mar. 29, 1974, as amended at 39 FR 40286, Nov. 15, 1974]



Sec. 429.47  Authority to refuse certification service.

    When the Commissioner finds, after giving notice and opportunity for 
hearing, that a person has:
    (a) Obtained or attempted to obtain a certificate through fraud, or 
through misrepresentation or concealment of a material fact;
    (b) Falsified the records required to be kept by Sec. 429.60; or
    (c) Failed to keep such records or to make them available, or to 
accord full opportunity to make an inventory of stocks on hand or 
otherwise to check the correctness of such records, as required by such 
section;

the Commissioner may immediately suspend service to such person under 
the regulations in this part, and may continue such suspension unless 
and until such person shows adequate cause why such suspension should be 
terminated.



                  Subpart F--Administrative Procedures



Sec. 429.50  Hearing procedure.

    Hearings pursuant to Sec. 429.47 shall be governed by part 16 of 
this chapter.
[41 FR 48267, Nov. 2, 1976, as amended at 42 FR 15674, Mar. 22, 1977]



Sec. 429.55  Fees.

    (a)(1) Fees for the services rendered under the regulations in this 
part shall be such as are necessary to provide, equip, and maintain an 
adequate certification service.
    (2) Whenever in the judgment of the Commissioner the ratio between 
fees collected (which are based upon experience and the best estimate of 
costs and the best estimate of earnings) and the costs of providing the 
service during an elapsed period of time, in the light of all 
circumstances and contingencies, warrants a refund from the fund 
collected during such period, he shall make ratable refunds to those 
persons to whom the services were rendered and charged.
    (b) The fees for requests for certification submitted under 
Sec. 429.40 are as follows:
    (1) $2,400 for each master lot or mixture of two or more master lots 
or parts thereof.
    (2) $1,700 for each dosage form batch.
    (3) The fees established in this paragraph may increase as Federal 
salary costs increase. The rate of increase will be no higher than 
Federal salary increases, commencing with pay raises on or after January 
1, 1997. Notification of the exact fees established and adjustments will 
be communicated directly to the manufacturers of insulin products.
    (c) A person requiring continuing certification services may 
maintain an advance deposit of the estimated costs of such services for 
a period of 2 months or more. Such deposits shall be debited with fees 
for services rendered, but shall not be debited for any fee the amount 
of which is not definitely specified in these regulations unless the 
depositor has previously requested the performance of the services to be 
covered by such fee. A monthly statement for each such advance deposit 
shall be rendered.
    (d) The unearned portion of any advance deposit made pursuant to 
paragraph (b) or (c) of this section shall be refunded to the depositor 
upon his application.
    (e) All advance deposits required by the regulations in this part 
429 shall be paid by money order, bank draft, or certified check drawn 
to the order of the Food and Drug Administration, collectible at par at 
Washington, DC. All deposits shall be forwarded to the Food and Drug 
Administration, Department of Health and Human Services, Washington, DC 
20204, whereupon after making appropriate record thereof they will be 
transmitted to the Chief Disbursing Officer, Division of Disbursement, 
Treasurer of the United

[[Page 310]]

States, for deposit to the special account ``Salaries and Expenses, 
Certification, Inspection and Other Services, Food and Drug 
Administration.''
[39 FR 11750, Mar. 29, 1974, as amended at 42 FR 27227, May 27, 1977; 48 
FR 788, Jan. 7, 1983; 60 FR 56516, Nov. 9, 1995]



                           Subpart G--Records



Sec. 429.60  Records of distribution.

    (a) The person to whom a certificate is issued shall keep complete 
records showing each shipment and other delivery (including exports) of 
each batch or part thereof, by the person requesting certification, and 
showing each such shipment and delivery into, or from any place in, any 
State or Territory, made by any person subject to his control, including 
records showing the date and quantity of each such shipment and delivery 
and the name and post office address of the person to whom such shipment 
or delivery was made.
    (b) Upon the request of any officer or employee of the Food and Drug 
Administration or of any other officer or employee of the United States, 
acting on behalf of the Secretary, the person to whom a certificate is 
issued, at all reasonable hours within 2 years after disposal of all the 
batch covered by such certificate, shall make such records available to 
any such officer or employee, and shall accord to such officer or 
employee full opportunity to make inventory of stocks of such batch on 
hand and otherwise to check the correctness of such records.



PART 430--ANTIBIOTIC DRUGS; GENERAL--Table of Contents




                      Subpart A--General Provisions

Sec.
430.3  Definitions applicable to all certifiable antibiotic drugs.
430.4  Definitions of antibiotic substances.
430.5  Definitions of master and working standards.
430.6  Definitions of the terms ``unit'' and ``microgram'' as applied to 
          antibiotic substances.

   Subpart B--Antibiotic Drugs Affected by the Drug Amendments of 1962

430.10  Certification or release of antibiotic drugs affected by the 
          drug amendments of 1962.

    Authority:  21 U.S.C. 321, 351, 352, 353, 355, 357, 371; 42 U.S.C. 
216, 241, 262.



                      Subpart A--General Provisions



Sec. 430.3  Definitions applicable to all certifiable antibiotic drugs.

    (a) The definitions and interpretations contained in section 201 of 
the Federal Food, Drug, and Cosmetic Act shall be applicable to such 
terms when used in the regulations in this chapter covering the 
certification of antibiotic and antibiotic-containing drugs.
    (b) The term Commissioner means the Commissioner of Food and Drugs 
and any other officer of the Food and Drug Administration whom he may 
designate to act in his behalf for the purpose of the regulations for 
the certification of antibiotic and antibiotic-containing drugs.
    (c) The term act means the Federal Food, Drug, and Cosmetic Act and 
amendments thereto. (52 Stat. 1040 et seq.; 21 U.S.C. 301-392).
    (d) The term U.S.P. means the official Pharmacopeia of the United 
States, including supplements thereto. The term N.F. means the official 
National Formulary, including supplements thereto.
    (e) The term batch means a specific homogeneous quantity of a drug.
    (f) The term batch mark means an identifying mark or other 
identifying device assigned to a batch by the manufacturer or packer 
thereof.
    (g) The term manufacture does not include the use of a drug as an 
ingredient in compounding any prescription issued by a practitioner 
licensed by law to administer such drug.
[39 FR 18925, May 30, 1974]



Sec. 430.4  Definitions of antibiotic substances.

    (a) The following are definitions of antibiotic substances:
    (1) Penicillin. Each of the several antibiotic substances (e.g., 
penicillin F, penicillin G, penicillin X) produced by the growth of 
Penicillium notatum or Penicillium chrysogenum, and each of

[[Page 311]]

the same substances produced by any other means, is a kind of 
penicillin.
    (2) Streptomycin. Each of the several antibiotic substances produced 
by the growth of Streptomyces griseus, and each of the same substances 
produced by any other means, is a kind of streptomycin.
    (3) Dihydrostreptomycin. Each of the antibiotic substances produced 
by hydrogenation of streptomycin, and each of the same substances 
produced by any other means, is a kind of dihydrostreptomycin.
    (4) Chlortetracycline. Each of the several antibiotic substances 
produced by the growth of Streptomyces aureofaciens, and each of the 
same substances produced by any other means is a kind of 
chlortetracycline.
    (5) Tetracycline. Each of the several antibiotic substances produced 
by the hydrogenation of chlortetracycline, and each of the same 
substances produced by any other means, is a kind of tetracycline.
    (6) Chloramphenicol. Each of the several antibiotic substances 
produced by the growth of Streptomyces venezuelae, and each of the same 
substances produced by any other means, is a kind of chloramphenicol.
    (7) Bacitracin. Each of the several antibiotic substances produced 
by the growth of Bacillus subtilis var. Tracy, and each of the same 
substances produced by any other means, is a kind of bacitracin.
    (8) [Reserved]
    (9) Amphotericin. Each of the antibiotic substances produced by the 
growth of Streptomyces nodosus, and each of the same substances produced 
by any other means, is a kind of amphortericin.
    (10) Colistin. Each of the antibiotic substances produced by the 
growth of Bacillus polymyxa var. colistinus, and each of the same 
substances produced by any other means, is a kind of colistin.
    (11) Cycloserine. Each of the antibiotic substances produced by the 
growth of Streptomyces orchidaceus, and each of the same substances 
produced by any other means, is a kind of cycloserine.
    (12) Erythromycin. Each of the antibiotic substances produced by the 
growth of Streptomyces erythreus, and each of the same substances 
produced by any other means, is a kind of erythromycin.
    (13) Gramicidin. Each of the antibiotic substances produced by the 
growth of Bacillus brevis, and each of the same substances produced by 
any other means, is a kind of gramicidin.
    (14) Griseofulvin. Each of the antibiotic substances produced by the 
growth of Penicillium patulum or Penicillium griseofulvum, and each of 
the same substances produced by any other means, is a kind of 
griseofulvin.
    (15) Kanamycin. Each of the antibiotic substances produced by the 
growth of Streptomyces kanamyceticus, and each of the same substances 
produced by any other means, is a kind of kanamycin.
    (16) Neomycin. Each of the antibiotic substances produced by the 
growth of Streptomyces fradiae, and each of the same substances produced 
by any other means, is a kind of neomycin.
    (17) Novobiocin. Each of the antibiotic substances produced by the 
growth of Streptomyces niveus (known also as Streptomyces spheroides), 
and each of the same substances produced by any other means, is a kind 
of novobiocin.
    (18) Nystatin. Each of the antibiotic substances produced by the 
growth of Streptomyces noursei, and each of the same substances produced 
by any other means, is a kind of nystatin.
    (19) Oleandomycin. Each of the antibiotic substances produced by the 
growth of Streptomyces antibioticus, and each of the same substances 
produced by any other means, is a kind of oleandomycin.
    (20) Troleandomycin. Each of the antibiotic substances produced by 
the triacetylation of oleandomycin, and each of the same substances 
produced by any other means, is a kind of troleandomycin.
    (21) Oxytetracycline. Each of the antibiotic substances produced by 
the growth of Streptomyces rimosus, and each of the same substances 
produced by any other means, is a kind of oxytetracycline.
    (22) Paromomycin. Each of the antibiotic substances produced by the 
growth of Streptomyces rimosus var. paromomycinus, and each of the same

[[Page 312]]

substances produced by any other means, is a kind of paromomycin.
    (23) Polymyxin. Each of the antibiotic substances produced by the 
growth of Bacillus polymyxa, and each of the same substances produced by 
any other means, is a kind of polymyxin.
    (24) Plicamycin. Each of the antibiotic substances produced by the 
growth of a variant of Streptomyces plicatus, and each of the same 
substances produced by any other means, is a kind of plicamycin.
    (25) Tyrothricin. Each of the mixtures of antibiotic substances 
produced by the growth of Bacillus brevis, and each of the same mixtures 
of substances produced by any other means, is a kind of tyrothricin.
    (26) Vancomycin. Each of the antibiotic substances produced by the 
growth of Streptomyces orientalis, and each of the same substances 
produced by any other means, is a kind of vancomycin.
    (27) [Reserved]
    (28) Gentamicin. Each of the antibiotic substances produced by the 
growth of Micromonospora purpurea, and each of the same substances 
produced by any other means, is a kind of gentamicin.
    (29) Dactinomycin. Dactinomycin is a specific kind of actinomycin 
produced by the growth of Streptomyces parvullus or the same antibiotic 
produced by any other means.
    (30) Candicidin. Each of the heptaene antibiotic substances produced 
by the growth of Streptomyces griseus and each of the same substances 
produced by any other means is a kind of candicidin.
    (31) Cephalosporin. Each of the antibiotic substances produced by 
the growth of Cephalosporium acremonium, and each of the same substances 
produced by any other means, is a kind of cephalosporin.
    (32) Lincomycin. Each of the antibiotic substances produced by the 
growth of Streptomyces lincolnensis var. lincolnensis, and each of the 
same substances produced by any other means, is a kind of lincomycin.
    (33) Demeclocycline. Each of the antibiotic substances produced by 
removal of the 6-methyl group from chlortetracycline, and each of the 
same substances produced by any other means, is a kind of 
demeclocycline.
    (34) Clindamycin. Each of the antibiotic substances produced by the 
7-chloro-substitution of the 7(R)hydroxyl group of lincomycin, and each 
of the same substances produced by any other means, is a kind of 
clindamycin.
    (35)  [Reserved]
    (36) Capreomycin. Each of the antibiotic substances produced by the 
growth of Streptomyces capreolus, and each of the same substances 
produced by any other means, is a kind of capreomycin.
    (37) Rifamycin. Each of the several antibiotic substances (e.g., 
rifamycin A, rifamycin B, rifamycin SV) produced by the growth of 
Streptomyces mediterranei, and each of the same substances produced by 
any other means, is a kind of rifamycin.
    (38) Spectinomycin. Each of the antibiotic substances produced by 
the growth of Streptomyces spectabilis, and each of the same substances 
produced by any other means, is a kind of spectinomycin.
    (39) Mitomycin. Mitomycin is the antibiotic substance produced by 
the growth of Streptomyces caespitosus, and each of the same substances 
produced by any other means is a kind of mitomycin.
    (40) Doxorubicin. Each of the antibiotic substances produced by the 
growth of Streptomyces peucetius var. caesius, and each of the same 
substances produced by any other means, is a kind of doxorubicin.
    (41) Bleomycin. Each of the antibiotic substances produced by the 
growth of Streptomyces verticillus and each of the same substances 
produced by any other means is a kind of bleomycin.
    (42) Tobramycin. A specific one of the antibiotic substances 
produced by the growth of Streptomyces tenebrarius, and the same 
substance produced by any other means, is tobramycin.
    (43) Amikacin. Each of the antibiotic substances produced by the 
acylation of the 1-amino group of the 2-deoxy-streptamine moiety of 
kanamycin A with L-(-)-)amino--hydroxybuyric acid, and 
each of the same substances produced by any other means is a kind of 
amikacin.

[[Page 313]]

    (44) Vidarabine. Vidarabine is a purine glycoside antibiotic 
substance produced by the growth of Streptomyces antibioticus, and each 
of the same substances produced by any other means is a kind of 
vidarabine.
    (45) Natamycin. Each of the antibiotic substances produced by the 
growth of Streptomyces natalensis, and each of the same substances 
produced by any other means, is a kind of natamycin.
    (46) Daunorubicin. Each of the antibiotic substances produced by the 
growth of Streptomyces coeruleorubidus and each of the same substances 
produced by any other means is a kind of daunorubicin.
    (47) Sisomicin. A specific one of the antibiotic substances produced 
by the growth of Micromonospora inyoensis, and the same substance 
produced by any other means, is a kind of sisomicin.
    (48) Moxalactam. 5-oxa-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylic 
acid, 7-[[carboxy(4-hydroxyphenyl)acetyl]-amino]-7-methoxy-3-[[(1-
methyl-1H-tetrazol-5-yl)thio]-methyl]-8-oxo-, disodium salt.
    (49) Cefoperazone. Cefoperazone is a semi-synthetic antibiotic 
substance produced by the acylation of the amino group at the 7 position 
of 7-aminocephalosporanic acid with -(4-ethyl-2,3-dioxo-1-
piperazinecarboxamido)--(4-hydroxyphenyl) acetic acid and 
introduction of a methylthiotetrazol group at the 3 position.
    (50) Netilmicin. Netilmicin is a semi-synthetic antibiotic of the 
aminoglycoside group derived from sisomicin, and each of the same 
substances produced by any other means is a kind of netilmicin. It is d-
Streptamine, 4-O-[3-amino-6-(aminomethyl)-3,4-dihydro-2H-pyran-2-yl]-2-
deoxy-6-O-[3-deoxy-4-C-methyl-3-(methylamino)--l-
arabinopyranosyl]-N\1\-ethyl-, (2S-cis)-.
    (51) Cyclosporine. Cyclosporine is a specific cyclic polypeptide 
consisting of 11 amino acids produced by the growth of Cylindrocarpon 
lucidum Booth or Tolypocladium inflatum Gams.
    (52) Cefonicid. 5-Thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylic 
acid, 7-[(hydroxyphenylacetyl)amino]-8-oxo-3[[[1-(sulfomethyl)-1H-
tetrazol-5yl]-thio]methyl]-, disodium salt, [6R-
[67(R*)]].
    (53) Clavulanic acid. Clavulanic acid is an antibiotic substance 
produced by the growth of Streptomyces clavuligerus having the structure 
described as follows: Z-(2R, 5R)-3-(2-hydroxyethylidene)-7-oxo-4-oxa-1-
azabicyclo[3.2.0]heptane-2-carboxylic acid, and each of the same 
substances produced by any other means, is a kind of clavulanic acid.
    (54) Ceftriaxone. Ceftriaxone is a semi-synthetic antibiotic 
substance produced by the addition of S-2-benzothiazolyl-2-(2-
aminothiazol-4-yl)-2-methoxyiminothioacetate to the 7 amino group of 7-
amino-3-(2,5-dihydro-2 methyl-5,6-dioxo-1,2,4-triazin-3-yl)-thiomethyl-
3-cephem-4-carboxylic acid.
    (55) Imipenem monohydrate. Imipenem monohydrate is an antibiotic 
substance having the chemical structure described by the following name: 
[5R-[5,6,(R*)]]-6-(1-hydroxyethyl)-3-[[2-
[(iminomethyl)amino]ethyl]thio]-7-oxo-1-azabicyclo[3.2.0]-hept-2-ene-2-
carboxylic acid monohydrate.
    (56) Aztreonam. [2S[2alpha,3beta(Z)]]-2-[[[1-(2-amino-4-thiazolyl)-
2-[(2-methyl-4-oxo-1-sulfo-3-azetidinyl)amino]-2-
oxoethylidene]amino]oxy]-2-methylpropanoic acid.
    (57) Sulbactam. Sulbactam is a semi-synthetic antibiotic substance 
produced by the oxidation of the sulfur atom at the 4 position to its 
dioxide and the deamination at the 6 position of (2S,5R)-6-amino-3,3-
dimethyl-7-oxo-4 thia-1-azabicyclo[3.2.0]heptane-2-carboxylic acid (6-
APA).
    (58) Cefmenoxime. Cefmenoxime is 5-thia-1-azabicyclo[4.2.0]oct-2-
ene-2-carboxylic acid, 7-[[(2-amino-4-thiazolyl) 
(methoxyimino)acetyl]amino]-3-[[(1-methyl-1H-tetrazol-5-yl)thio]methyl]-
8-oxo-, [6R-[6-,7-(Z)]]-.
    (59) Cefixime. Cefixime is a semisynthetic antibiotic substance 
produced by the acylation of the amino group at the 7 position of 7-
aminocephalosporanic acid with a -[(2-amino-4-thiazolyl) 
(carboxymethoxy)imino] acetyl group and the introduction of a vinyl 
group at the 3 position.
    (60) Cefotiam. Cefotiam is an antibiotic substance having the 
chemical structure described by the following

[[Page 314]]

name: 7-(R)-[2-(2-amino-4-thiazol)acetamido]-3-[[[1-(2-
dimethylamino)ethyl]-1H-tetrazol-5-y1] thio] methyl]-3-cephem-4-
carboxylic acid.
    (61) Mupirocin. Each of the antibiotic substances produced by the 
growth of Pseudomonas fluorescens, and each of the same substances 
produced by any other means, is a kind of mupirocin.
    (62) Cefmetazole. Cefmetazole is an antibiotic substance having the 
chemical structure described by the following name: (6R-cis)-7- 
[[[cyanomethyl)thio]acetyl]amino]-7- methoxy-3-[[(1-methyl-1H- tetrazol-
5-yl)thio]methyl]-8-oxo-5- thia-1-azabicyclo[4.2.0]oct-2- ene-2-
carboxylic acid.
    (63) Cefpiramide. Cefpiramide is an antibiotic substance having the 
chemical structure described by the following name: (6R, 7R)-7-[(R)-2-
(4-hydroxy-6- methyl-nicotinamido)-2-(p- hydroxyphenyl)acetamido]-3-
[[(1- methyl-1H-tetrazol-5-yl)thio]methyl]-8- oxo-5-thia-1-
azabicyclo[4.2.0]oct-2-ene- 2-carboxylic acid.
    (64) Clarithromycin. Clarithromycin is 6-O-methylerythromycin A.
    (65) Azithromycin. Azithromycin is an antibiotic substance having 
the chemical structure described by the following name: 
(2R,3S,4R,5R,8R,10R,11R,12S,13S,14R)-13-[(2,6-dideoxy-3-C-methyl-3-O-
methyl--L-ribo-hexopyranosyl)oxy]-2-ethyl-3,4,10-trihydroxy-
3,5,6,8,10,12,14-heptamethyl-11-[[3,4,6-trideoxy-3-(dimethylamino)-
-D-xylo-hexopyranosyl]oxy]-1-oxa-6-azacyclopentadecan-15-one.
    (66) Cefprozil. Cefprozil is an antibiotic substance having the 
chemical structure described by the following name: (6R,7R)-7-[(R)-2-
amino-2-(p-hydroxyphenyl)acetamido]8-oxo-3-propenyl-5-thia-1-
azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid. It is a mixture of the Z 
(cis) and E (trans) isomers in an approximate ratio of 9:1, 
respectively.
    (67) Idarubicin. Idarubicin is an anthracycline antibiotic substance 
having the chemical structure described by the following name: 5,12-
Naphthacenedione, 9-acetyl-7-[(3-amino-2,3,6-trideoxy--L-lyxo- 
hexopyranosyl)oxy]-7,8,9,10-tetrahydro-6,9,11-trihydroxy-(7S-cis).
    (68) Loracarbef. Loracarbef is an antibiotic substance having the 
chemical structure described by the following name: (6R,7S)-7-[(R)-2-
amino-2-phenylacetamido]-3-chloro-8-oxo-1-azabicyclo[4.2.0]oct-2-ene-2-
carboxylic acid.
    (69) Rifabutin. Rifabutin is an antibiotic substance having the 
chemical structure described by the following name: 
(9S,12E,14S,15R,16S,17R,18R,19R, 20S,21S,22E, 24Z)-6,16, 18,20-
tetrahydroxy-1'-isobutyl-14-methoxy-7,9,15,17,19,21,25-
heptamethylspiro[9,4-(epoxypentadeca[1,11,13]trienimino)-2H-
furo[2',3':7,8]naphth[1,2-d]imidazole-2,4'-piperidine]-5,10,26-(3H,9H)-
trione-16-acetate.
    (70) Cefpodoxime proxetil. Cefpodoxime proxetil is an antibiotic 
substance having the chemical structure described by the following name: 
()-1-Hydroxyethyl(+)-(6R,7R)-7-[2-(2-amino-4-
thiazolyl)glyoxylamido]-3-(methoxymethyl)-8-oxo-5-thia-1-
azabicyclo[4.2.0]oct-2-ene-2-carboxylate,72-(Z)-(O-
methyloxime), isopropyl carbonate (ester).
    (b) [Reserved]
[39 FR 18925, May 30, 1974]

    Editorial Note: For Federal Register citations affecting Sec. 430.4, 
see the List of CFR Sections Affected in the Finding Aids section of 
this volume.



Sec. 430.5  Definitions of master and working standards.

    (a) Master standards--(1) Penicillinand salts of penicillin--(i) 
Penicillin G The term ``penicillin G master standard' means a specific 
lot of crystalline penicillin G that is designated by the Commissioner 
as the standard of comparison in determining the potency of the 
penicillin G working standard.
    (ii) [Reserved]
    (iii) Penicillin V. The term ``penicillin V master standard'' means 
a specific lot of crystalline penicillin V that is designated by the 
Commissioner as the standard of comparison in determining the potency of 
the penicillin V working standard.
    (iv)-(v) [Reserved]
    (vi) Methicillin. The term ``methicillin master standard'' means a 
specific lot of crystalline methicillin that is designated by the 
Commissioner

[[Page 315]]

as the standard of comparison in determining the potency of the 
methicillin working standard.
    (vii) Oxacillin. The term ``oxacillin master standard'' means a 
specific lot of crystalline oxacillin that is designated by the 
Commissioner as the standard of comparison in determining the potency of 
the oxacillin working standard.
    (viii) Ampicillin. The term ``ampicillin master standard'' means a 
specific lot of crystalline ampicillin that is designated by the 
Commissioner as the standard of comparison in determining the potency of 
the ampicillin working standard.
    (ix) Nafcillin. The term ``nafcillin master standard'' means a 
specific lot of crystalline nafcillin that is designated by the 
Commissioner as the standard of comparison in determining the potency of 
the nafcillin working standard.
    (x) Cloxacillin. The term ``cloxacillin master standard'' means a 
specific lot of crystalline cloxacillin that is designated as the 
standard of comparison in determining the potency of the cloxacillin 
working standard.
    (xi) Dicloxacillin. The term ``dicloxacillin master standard'' means 
a specific lot of dicloxacillin that is designated by the Commissioner 
as the standard of comparison in determining the potency of the 
dicloxacillin working standard.
    (xii) The term ``hetacillin working standard'' means a specific lot 
of homogenous preparation of hetacillin.
    (2) Streptomycin. The term ``streptomycin master standard'' means a 
specific lot of streptomycin that is designated by the Commissioner as 
the standard of comparison in determining the potency of the 
streptomycin working standard.
    (3) Dihydrostreptomycin. The term ``dihydrostreptomycin master 
standard'' means a specific lot of crystalline dihydrostreptomycin that 
is designated by the Commissioner as the standard of comparison in 
determining the potency of the dihydrostreptomycin working standard.
    (4) Chlortetracycline. The term ``chlortetracyclin master standard'' 
means a specific lot of crystalline chlortetracycline hydrochloride that 
is designated by the Commissioner as the standard of comparison in 
determining the potency of the chlortetracycline working standard.
    (5) Demeclocycline. The term ``demeclocycline master standard'' 
means a specific lot of crystalline demeclocycline hydrochloride that is 
designated by the Commissioner as the standard of comparison in 
determining the potency of the demeclocycline working standard.
    (6) Tetracycline. The term ``tetracycline master standard'' means a 
specific lot of crystalline tetracycline hydrochloride that is 
designated by the Commissioner as the standard of comparison in 
determining the potency of the tetracycline working standard.
    (7) Rolitetracycline. The term ``rolitetracycline master standard'' 
means a specific lot of crystalline rolitetracycline that is designated 
by the Commissioner as the standard of comparison in determining the 
potency of the rolitetracycline working standard.
    (8) Chloramphenicol. The term ``chloramphenicol master standard'' 
means a specific lot of crystalline chloramphenicol that is designated 
by the Commissioner as the standard of comparison in determining the 
potency of the chloramphenicol working standard.
    (9) Bacitracin The term ``bacitracin master standard'' means a 
specific lot of bacitracin that is designated by the Commissioner as the 
standard of comparison in determining the potency of the bacitracin 
working standard.
    (10) [Reserved]
    (11) Amphotericin. The term ``amphotericin A master standard'' means 
a specific lot of amphotericin A designated by the Commissioner as the 
standard of comparison in determining the potency of the amphotericin A 
working standard. The term ``amphotericin B master standard'' means a 
specific lot of amphotericin B designated by the Commissioner as the 
standard of comparison in determining the potency of the amphotericin B 
working standard.
    (12) Colistin. The term ``colistin master standard'' means a 
specific lot of colistin designated by the Commissioner as the standard 
of comparison in

[[Page 316]]

determining the potency of the colistin working standard.
    (13) Colistimethate. The term ``colistimethate master standard'' 
means a specific lot of colistimethate designated by the Commissioner as 
the standard of comparison in determining the potency of the 
colistimethate working standard.
    (14) Cycloserine. The term ``cycloserine master standard'' means a 
specific lot of cycloserine designated by the Commissioner as the 
standard of comparison in determining the potency of the cycloserine 
working standard.
    (15) Erythromycin. The term ``erythromycin master standard'' means a 
specific lot of erythromycin designated by the Commissioner as the 
standard of comparison in determining the potency of the erythromycin 
working standard.
    (16) Gramicidin. The term ``gramicidin master standard'' means a 
specific lot of gramicidin designated by the Commissioner as the 
standard of comparison in determining the potency of the gramicidin 
working standard.
    (17) Griseofulvin. The term ``griseofulvin master standard'' means a 
specific lot of griseofulvin designated by the Commissioner as the 
standard of comparison in determining the potency of the griseofulvin 
working standard.
    (18) Kanamycin. The term ``kanamycin master standard'' means a 
specific lot of kanamycin designated by the Commissioner as the standard 
of comparison in determining the potency of the kanamycin working 
standard.
    (19) Neomycin. The term ``neomycin master standard'' means a 
specific lot of neomycin designated by the Commissioner as the standard 
of comparison in determining the potency of the neomycin working 
standard.
    (20) Novobiocin. The term ``novobiocin master standard'' means a 
specific lot of novobiocin designated by the Commissioner as the 
standard of comparison in determining the potency of the novobiocin 
working standard.
    (21) Nystatin. The term ``nystatin master standard'' means a 
specific lot nystatin designated by the Commissioner as the standard of 
comparison in determining the potency of the nystatin working standard.
    (22) Oleandomycin. The term ``oleandomycin master standard'' means a 
specific lot of oleandomycin designated by the Commissioner as the 
standard of comparison in determining the potency of the oleandomycin 
working standard.
    (23) Oxytetracycline. The term ``oxytetracycline master standard'' 
means a specific lot of oxytetracycline designated by the Commissioner 
as the standard of comparison in determining the potency of the 
oxytetracycline working standard.
    (24) Paromomycin. The term ``paromomycin master standard'' means a 
specific lot of paromomycin designated by the Commissioner as the 
standard of comparison in determining the potency of the paromomycin 
working standard.
    (25) Polymyxin B. The term ``polymyxin B master standard'' means a 
specific lot of polymyxin B designated by the Commissioner as the 
standard of comparison in determining the potency of the polymyxin B 
working standard.
    (26) [Reserved]
    (27) Vancomycin. The term ``vancomycin master standard'' means a 
specific lot of vancomycin designated by the Commissioner as the 
standard of comparison in determining the potency of the vancomycin 
working standard.
    (28) [Reserved]
    (29) Troleandomycin. The term ``troleandomycin master standard'' 
means a specific lot of troleandomycin designated by the Commissioner as 
the standard of comparison in determiing the potency of the 
troleandomycin working standard.
    (30) Gentamicin. The term ``gentamicin master standard'' means a 
specific lot of gentamicin designated by the Commissioner as the 
standard of comparison in determining the potency of the gentamicin 
working standard.
    (31) Dactinomycin. The term ``dactinomycin master standard'' means a 
specific lot of dactinomycin designated by the Commissioner as the 
standard of comparison in determining the potency of the dactinomycin 
working standard.
    (32) Candicidin. The term ``candicidin master standard'' means a 
specific lot of candicidin that is designated by the Commissioner as the 
standard of comparison in determining the potency of the candicidin 
working standard.

[[Page 317]]

    (33) Cephalothin. The term ``cephalothin master standard'' means a 
specific lot of cephalothin designated by the Commissioner as the 
standard of comparison in determining the potency of the cephalothin 
working standard.
    (34) Lincomycin. The term ``lincomycin master standard'' means a 
specific lot of lincomycin designated by the Commissioner as the 
standard of comparison in determining the potency of the lincomycin 
working standard.
    (35) Methacycline. The term ``methacycline master standard'' means a 
specific lot of methacycline designated by the Commissioner as the 
standard of comparison in determining the potency of the methacycline 
working standard.
    (36) Doxycycline. The term ``doxycycline master standard'' means a 
specific lot of -6-deoxyoxytetracycline designated by the 
Commissioner as the standard of comparison in determining the potency of 
the doxycycline working standard.
    (37) Cephaloridine. The term ``cephaloridine master standard'' means 
a specific lot of cephaloridine that is designated by the Commissioner 
as the standard of comparison in determining the potency of the 
cephaloridine working standard.
    (38) Plicamycin. The term ``plicamycin master standard'' means a 
specific lot of plicamycin designated by the Commissioner as the 
standard of comparison in determining the potency of the plicamycin 
working standard.
    (39) Clindamycin. The term ``clindamycin master standard'' means a 
specific lot of clindamycin designated by the Commissioner as the 
standard of comparison in determining the potency of the clindamycin 
working standard.
    (40) Cephaloglycin. The term ``cephaloglycin master standard'' means 
a specific lot of cephaloglycin designated by the Commissioner as the 
standard of comparison in determining the potency of the cephaloglycin 
working standard.
    (41) Carbenicillin. The term ``carbenicillin master standard'' means 
a specific lot of carbenicillin designated by the Commissioner as the 
standard of comparison in determining the potency of the carbenicillin 
working standard.
    (42) Cephalexin. The term ``cephalexin master standard'' means a 
specific lot of cephalexin that is designated by the Commissioner as the 
standard of comparison in determining the potency of the cephalexin 
working standard.
    (43) [Reserved]
    (44) Capreomycin. The term ``capreomycin master standard'' means a 
specific lot of capreomycin designated by the Commissioner as the 
standard of comparison in determining the potency of the capreomycin 
working standard.
    (45) Rifampin. The term ``rifampin master standard'' means a 
specific lot of rifampin designated by the Commissioner as the standard 
of comparison in determining the potency of the rifampin working 
standard.
    (46) Minocycline. The term ``minocycline master standard'' means a 
specific lot of minocycline designated by the Commissioner as the 
standard of comparison in determining the potency of the minocycline 
working standard.
    (47) Spectinomycin. The term ``spectinomycin master standard'' means 
a specific lot of spectinomycin designated by the Commissioner as the 
standard of comparison in determining the potency of the spectinomycin 
working standard.
    (48) Clindamycin palmitate hydrochloride. The term ``clindamycin 
palmitate hydrochloride master standard'' means a specific lot of 
clindamycin palmitate hydrochloride designated by the Commissioner as 
the standard of comparison in determining the potency of the clindamycin 
palmitate hydrochloride working standard.
    (49) Carbenicillin indanyl. The term ``carbenicillin indanyl master 
standard'' means a specific lot of carbenicillin indanyl designated by 
the Commissioner as the standard of comparison in determining the 
potency of the carbenicillin indanyl working standard.
    (50) Cephapirin. The term ``cephapirin master standard'' means a 
specific lot of cephapirin that is designated by the Commissioner as the 
standard of comparison in determining the potency of the cephapirin 
working standard.
    (51) Cefazolin. The term ``cefazolin master standard'' means a 
specific lot

[[Page 318]]

of cefazolin that is designated by the Commissioner as the standard of 
comparison in determining the potency of the cefazolin working standard.
    (52) Mitomycin. The term ``mitomycin master standard'' means a 
specific lot of crystalline mitomycin that is designated by the 
Commissioner as the standard of comparison in determining the potency of 
the mitomycin working standard.
    (53) Amoxicillin. The term ``amoxicillin master standard'' means a 
specific lot of amoxicillin that is designated by the Commissioner as 
the standard of comparison in determining the potency of the amoxicillin 
working standard.
    (54)  [Reserved]
    (55) Cephradine. The term ``cephradine master standard'' means a 
specific lot of cephradine that is designated by the Commissioner as the 
standard of comparison in determining the potency of the cephradine 
working standard.
    (56) Doxorubicin. The term ``doxorubicin master standard'' means a 
specific lot of crystalline doxorubicin that is designated by the 
Commissioner as the standard of comparison in determining the potency of 
the doxorubicin working standard.
    (57) Bleomycin. The term ``bleomycin master standard'' means a 
specific lot of bleomycin designated by the Commissioner as the standard 
of comparison in determining the potency of the bleomycin working 
standard.
    (58) Tobramycin. The term ``tobramycin master standard'' means a 
specific lot of tobramycin designated by the Commissioner as the 
standard of comparison in determining the potency of the tobramycin 
working standard.
    (59) Amikacin. The term ``amikacin master standard'' means a 
specific lot of amikacin designated by the Commissioner as the standard 
of comparison in determining the potency of the amikacin working 
standard.
    (60) Vidarabine. The term ``vidarabine master standard'' means a 
specific lot of vidarabine that is designated by the Commissioner as the 
standard of comparison in determining the potency of the vidarabine 
working standard.
    (61) Ticarcillin. The term ``ticarcillin master standard'' means a 
specific lot of ticarcillin designated by the Commissioner as the 
standard of comparison in determining the potency of the ticarcillin 
working standard.
    (62) Cefadroxil. The term ``cefadroxil master standard'' means a 
specific lot of cefadroxil that is designated by the Commissioner as the 
standard of comparison in determining the potency of the cefadroxil 
working standard.
    (63) Natamycin. The term ``natamycin master standard'' means a 
specific lot of natamycin designated by the Commissioner as the standard 
of comparison in determining the potency of the natamycin working 
standard.
    (64) Cefoxitin. The term ``cefoxitin master standard'' means a 
specific lot of cefoxitin that is designated by the Commissioner as the 
standard of comparison in determining the potency of the cefoxitin 
working standard.
    (65) Cefamandole. The term ``cefamandole master standard'' means a 
specific lot of cefamandole that is designated by the Commissioner as 
the standard of comparison in determining the potency of the cefamandole 
working standard.
    (66) Cefaclor. The term ``cefaclor master standard'' means a 
specific lot of cefaclor that is designated by the Commissioner as the 
standard of comparison in determining the potency of the cefaclor 
working standard.
    (67) Cyclacillin. The term ``cyclacillin master standard'' means a 
specific lot of cyclacillin that is designated by the Commissioner as 
the standard of comparison in determining the potency of the cyclacillin 
working standard.
    (68) Daunorubicin. The term ``daunorubicin master standard'' means a 
specific lot of daunorubicin that is designated by the Commissioner as 
the standard of comparison in determining the potency of the 
daunorubicin working standard.
    (69) Sisomicin. The term ``sisomicin master standard'' means a 
specific lot of sisomicin that is designated by the Commissioner as the 
standard of comparison in determining the potency of the sisomicin 
working standard.
    (70) Meclocycline. The term ``meclocycline master standard'' means a 
specific lot of meclocycline that is designated by the Commissioner as 
the standard of comparison in determining

[[Page 319]]

the potency of the meclocycline working standard.
    (71) Cefotaxime. The term ``cefotaxime master standard'' means a 
specific lot of cefotaxime that is designated by the Commissioner as the 
standard of comparison in determining the potency of the cefotaxime 
working standard.
    (72) Mezlocillin. The term ``mezlocillin master standard'' means a 
specific lot of mezlocillin that is designated by the Commissioner as 
the standard of comparison in determining the potency of the mezlocillin 
working standard.
    (73) Moxalactam. The term ``moxalactam master standard'' means a 
specific lot of moxalactam that is designated by the Commissioner as the 
standard of comparison in determining the potency of the moxalactam 
working standard.
    (74) Piperacillin. The term ``piperacillin master standard'' means a 
specific lot of piperacillin that is designated by the Commissioner as 
the standard of comparison in determining the potency of the 
piperacillin working standard.
    (75) Azlocillin. The term ``azlocillin master standard'' means a 
specific lot of azlocillin that is designated by the Commissioner as the 
standard of comparison in determining the potency of the azlocillin 
working standard.
    (76) Cefoperazone. The term ``cefoperazone master standard'' means a 
specific lot of cefoperazone that is designated by the Commissioner as 
the standard of comparison in determining the potency of the 
cefoperazone working standard.
    (77) Netilmicin. The term ``netilmicin master standard'' means a 
specific lot of netilmicin that is designated by the Commissioner as the 
standard of comparison in determining the potency of the netilmicin 
working standard.
    (78) Cefuroxime. The term ``cefuroxime master standard'' means a 
specific lot of cefuroxime that is designated by the Commissioner as the 
standard of comparison in determining the potency of the cefuroxime 
working standard.
    (79) Ceftizoxime. The term ``ceftizoxime master standard'' means a 
specific lot of ceftizoxime that is designated by the Commissioner as 
the standard of comparison in determining the potency of the ceftizoxime 
working standard.
    (80) Cyclosporine. The term ``cyclosporine master standard'' means a 
specific lot of cyclosporine that is designated by the Commissioner as 
the standard of comparison in determining the potency of the 
cyclosporine working standard.
    (81) Ceforanide. The term ``ceforanide master standard'' means a 
specific lot of ceforanide that is designated by the Commissioner as the 
standard of comparison in determining the potency of the ceforanide 
working standard.
    (82) Cefonicid. The term ``cefonicid master standard'' means a 
specific lot of cefonicid that is designated by the Commissioner as the 
standard of comparison in determining the potency of the cefonicid 
working standard.
    (83) Clavulanic acid. The term ``clavulanic acid master standard'' 
means a specific lot of clavulanic acid or a salt thereof that is 
designated by the Commissioner as the standard of comparison in 
determining the potency of the clavulanic acid working standard.
    (84) Amdinocillin. The term ``amdinocillin master standard'' means a 
specific lot of amdinocillin that is designated by the Commissioner as 
the standard of comparison in determining the potency of the 
amdinocillin working standard.
    (85) Ceftriaxone. The term ``ceftriaxone master standard'' means a 
specific lot of ceftriaxone that is designated by the Commissioner as 
the standard of comparison in determining the potency of the ceftriaxone 
working standard.
    (86) Ceftazidime. The term ``ceftazidime master standard'' means a 
specific lot of ceftazidime that is designated by the Commissioner as 
the standard of comparison in determining the potency of the ceftazidime 
working standard.
    (87) Imipenem. The term ``imipenem master standard'' means a 
specific lot of imipenem that is designated by the Commissioner as the 
standard of comparison in determining the potency of the imipenem 
working standard.
    (88) Cefotetan. The term ``cefotetan master standard'' means a 
specific lot of cefotetan that is designated by the

[[Page 320]]

Commissioner as the standard of comparison in determining the potency of 
the cefotetan working standard.
    (89) Aztreonam. The term ``aztreonam master standard'' means a 
specific lot of aztreonam that is designated by the Commissioner as the 
standard of comparison in determining the potency of the aztreonam 
working standard.
    (90) Sulbactam. The term ``sulbactam master standard'' means a 
specific lot of sulbactam that is designated by the Commissioner as the 
standard of comparison in determining the potency of the sulbactam 
working standard.
    (91) Cefuroxime axetil. The term ``cefuroxime axetil master 
standard'' means a specific lot of cefuroxime axetil that is designated 
by the Commissioner as the standard of comparison in determining the 
potency of the cefuroxime axetil working standard.
    (92) Cefmenoxime. The term ``cefmenoxime master standard'' means a 
specific lot of cefmenoxime that is designated by the Commissioner as 
the standard of comparison in determining the potency of the cefmenoxime 
working standard.
    (93) Cefixime. The term ``cefixime master standard'' means a 
specific lot of cefixime that is designated by the Commissioner as the 
standard of comparison in determining the potency of the cefixime 
working standard.
    (94) Cefotiam. The term ``cefotiam master standard'' means a 
specific lot of cefotiam that is designed by the Commissioner as the 
standard of comparison in determining the potency of the cefotiam 
working standard.
    (95) Clindamycin phosphate. The term ``clindamycin phosphate master 
standard'' means a specific lot of clindamycin phosphate that is 
designed by the Commissioner as the standard for comparison in 
determining the potency of the clindamycin phosphate standard.
    (96) Mupirocin. The term ``mupirocin master standard'' means a 
specific lot of mupirocin or a salt thereof that is designated by the 
Commissioner as the standard of comparison in determining the potency of 
the mupirocin working standard.
    (97) Cefmetazole. The term ``cefmetazole master standard'' means a 
specific lot of cefmetazole that is designated by the Commissioner as 
the standard of comparison in determining the potency of the cefmetazole 
working standard.
    (98) Cefpiramide. The term ``cefpiramide master standard'' means a 
specific lot of cefpiramide that is designated by the Commissioner as 
the standard of comparison in determining the potency of the cefpiramide 
working standard.
    (99) Clarithromycin. The term ``clarithromycin master standard'' 
means a specific lot of clarithromycin that is designated by the 
Commissioner as the standard of comparison in determining the potency of 
the clarithromycin working standard.
    (100) Azithromycin. The term ``azithromycin master standard'' means 
a specific lot of azithromycin that is designated by the Commissioner as 
the standard of comparison in determining the potency of the 
azithromycin working standard.
    (101) Cefprozil. The term ``cefprozil master standard'' means a 
specific lot of the (Z) isomer of cefprozil and a specific lot of the 
(E) isomer of cefprozil that are designated by the Commissioner as the 
standard of comparison in determining the potency of the cefprozil 
working standard.
    (102) Idarubicin. The term ``idarubicin master standard'' means a 
specific lot of idarubicin that is designated by the Commissioner as the 
standard of comparison in determining the potency of the idarubicin 
working standard.
    (103) Loracarbef. The term ``loracarbef master standard'' means a 
specific lot of loracarbef that is designated by the Commissioner as the 
standard of comparison in determining the potency of the loracarbef 
working standard.
    (104) Rifabutin. The term ``rifabutin master standard'' means a 
specific lot of rifabutin that is designated by the Commissioner as the 
standard of comparison in determining the potency of the rifabutin 
working standard.
    (105) Cefpodoxime proxetil. The term ``cefpodoxime proxetil master 
standard'' means a specific lot of the (R) isomer of cefpodoxime 
proxetil that is designated by the Commissioner as the standard of 
comparison in determining the potency of the cefpodoxime proxetil 
working standard.

[[Page 321]]

    (b) Working standards. The potency or purity of each preparation has 
been determined by comparison with its master standard , and each has 
been designated by the Commissioner as working standards for use in 
determining the potency or purity of antibiotic substances subject to 
the regulations in this chapter.Unless otherwise noted, the working 
standard and the U.S.P. reference standard for the antibiotic drug named 
are identical.
    (1) Penicillin. (i) The term ``penicillin G working standard'' means 
a specific lot of a homogeneous preparation of penicillin G.
    (ii) [Reserved]
    (iii) The term ``penicillin V working standard'' means a specific 
lot of a homogeneous preparation of penicillin V.
    (iv) [Reserved]
    (v) The term ``methicillin working standard'' means a specific lot 
of a homogeneous preparation of methicillin.
    (vi) The term ``oxacillin working standard'' means a specific lot of 
a homogeneous preparation of oxacillin.
    (vii) The term ``ampicillin working standard'' means a specific lot 
of a homogeneous preparation of ampicillin.
    (viii) The term ``nafcillin working standard'' means a specific lot 
of a homogeneous preparation of nafcillin.
    (ix) The term ``cloxacillin working standard'' means a specific lot 
of a homogeneous preparation of cloxacillin.
    (x) The term ``penicillin G procaine working standard'' means a 
specific lot of a homogeneous preparation of penicillin G procaine.
    (xi) The term ``dicloxacillin working standard'' means a specific 
lot of a homogeneous preparation of dicloxacillin.
    (xii) The term ``bacampicillin hydrochloride working standard'' 
means a specific lot of a homogeneous preparation of bacampicillin 
hydrochloride.
    (2) Amphotericin A. The term ``amphotericin A working standard'' 
means a specific lot of a homogeneous preparation of amphotericin A.
    (3) Amphotericin B. The term ``amphotericin B working standard'' 
means a specific lot of a homogeneous preparation of amphotericin B.
    (4) Streptomycin. The term ``streptomycin working standard'' means a 
specific lot of a homogeneous preparation of streptomycin.
    (5) Dihydrostreptomycin. The term ``dihydrostreptomycin working 
standard'' means a specific lot of a homogeneous preparation of 
dihydrostreptomycin.
    (6) Chlortetracycline. The term ``chlortetracycline working 
standard'' means a specific lot of a homogeneous preparation of 
chlortetracycline.
    (7) Demeclocycline. The term ``demeclocycline working standard'' 
means a specific lot of a homogeneous preparation of demeclocycline.
    (8) Tetracycline. The term ``tetracycline working standard'' means a 
specific lot of a homogeneous preparation of tetracycline.
    (9) Rolitetracycline. The term ``rolitetracycline working standard'' 
means a specific lot of a homogeneous preparation of rolitetracycline.
    (10) Chloramphenicol. The term ``chloramphenicol working standard'' 
means a specific lot of a homogeneous preparation of chloramphenicol.
    (11) Bacitracin. The term ``bacitracin working standard'' means a 
specific lot of a homogeneous preparation of bacitracin.
    (12) [Reserved]
    (13) Colistin. The term ``colistin working standard'' means a 
specific lot of a homogeneous preparation of colistin.
    (14) Colistimethate. The term ``colistimethate working standard'' 
means a specific lot of a homogeneous preparation of colistimethate.
    (15) Cycloserine. The term ``cycloserine working standard'' means a 
specific lot of a homogeneous preparation of cycloserine.
    (16) Erythromycin. The term ``erythromycin working standard'' means 
a specific lot of a homogeneous preparation of erythromycin.
    (17) Gramicidin. The term ``gramicidin working standard'' means a 
specific lot of a homogeneous preparation of gramicidin.
    (18) Griseofulvin. The term ``griseofulvin working standard'' means 
a specific lot of a homogeneous preparation of griseofulvin.
    (19) Kanamycin. The term ``kanamycin working standard'' means a 
specific lot of a homogeneous preparation of kanamycin.
    (20) Neomycin. The term ``neomycin working standard'' means a 
specific lot

[[Page 322]]

of a homogeneous preparation of neomycin.
    (21) Novobiocin. The term ``novobiocin working standard'' means a 
specific lot of a homogeneous preparation of novobiocin.
    (22) Nystatin. The term ``nystatin working standard'' means a 
specific lot of a homogeneous preparation of nystatin.
    (23) Oleandomycin. The term ``oleandomycin working standard'' means 
a specific lot of a homogeneous preparation of oleandomycin.
    (24) Troleandomycin. The term ``troleandomycin working 
standard''means a specific lot of a homogeneous preparation of 
troleandomycin.
    (25) Oxytetracycline. The term ``oxytetracycline working standard'' 
means a specific lot of a homogeneous preparation of oxytetracycline.
    (26) Paromomycin. The term ``paromomycin working standard'' means a 
specific lot of a homogeneous preparation of paromomycin.
    (27) Polymyxin B. The term ``polymyxin B working standard'' means a 
specific lot of a homogeneous preparation of polymyxin B.
    (28) Vancomycin. The term ``vancomycin working standard'' means a 
specific lot of a homogeneous preparation of vancomycin.
    (29) [Reserved]
    (30) Gentamicin. The term ``gentamicin working standard'' means a 
specific lot of a homogeneous preparation of gentamicin.
    (31) Dactinomycin. The term ``dactinomycin working standard'' means 
a specific lot of a homogeneous preparation of dactinomycin.
    (32) Candicidin. The term ``candicidin working standard'' means a 
specific lot of a homogeneous preparation of candicidin.
    (33) Cephalothin. The term ``cephalothin working standard'' means a 
specific lot of a homogeneous preparation of cephalothin.
    (34) Lincomycin. The term ``lincomycin working standard'' means a 
specific lot of a homogeneous preparation of lincomycin.
    (35) Methacycline. The term ``methacycline working standard'' means 
a specific lot of homogeneous preparation of methacycline.
    (36) Doxycycline. The term ``doxycycline working standard'' means a 
specific lot of homogeneous preparation of -6-
deoxyoxytetracycline.
    (37) Cephaloridine. The term ``cephaloridine working standard'' 
means a specific lot of homogeneous preparation of cephaloridine.
    (38) Plicamycin. The term ``plicamycin working standard'' means a 
specific lot of a homogeneous preparation of plicamycin.
    (39) Clindamycin. The term ``clindamycin working standard'' means a 
specific lot of a homogeneous preparation of clindamycin.
    (40) Cephaloglycin. The term ``cephaloglycin working standard'' 
means a specific lot of homogeneous preparation of cephaloglycin.
    (41) Carbenicillin. The term ``carbenicillin working standard'' 
means a specific lot of homogeneous preparation of carbenicillin.
    (42) Cephalexin. The term ``cephalexin working standard'' means a 
specific lot of a homogeneous preparation of cephalexin.
    (43) [Reserved]
    (44) Capreomycin. The term ``capreomycin working standard'' means a 
specific lot of a homogeneous preparation of capreomycin.
    (45) Rifampin. The term ``rifampin working standard'' means a 
specific lot of a homogeneous preparation of rifampin.
    (46) Minocycline. The term ``minocycline working standard'' means a 
specific lot of a homogeneous preparation of minocycline.
    (47) Spectinomycin. The term ``spectinomycin working standard'' 
means a specific lot of a homogeneous preparation of spectinomycin.
    (48) Clindamycin palmitate hydrochloride. The term ``clindamycin 
palmitate hydrochloride working standard'' means a specific lot of a 
homogeneous preparation of clindamycin palmitate hydrochloride.
    (49) Carbenicillin indanyl. The term ``carbenicillin indanyl working 
standard'' means a specific lot of a homogeneous preparation of 
carbenicillin indanyl.

[[Page 323]]

    (50) Cephapirin. The term ``cephapirin working standard'' means a 
specific lot of a homogeneous preparation of cephapirin.
    (51) Cefazolin. The term ``cefazolin working standard'' means a 
specific lot of a homogeneous preparation of cefazolin.
    (52) Mitomycin. The term ``mitomycin working standard'' means a 
specific lot of a homogeneous preparation of mitomycin.
    (53) Amoxicillin. The term ``amoxicillin working standard'' means a 
specific lot of a homogeneous preparation of amoxicillin.
    (54)  [Reserved]
    (55) Cephradine. The term ``cephradine working standard'' means a 
specific lot of a homogeneous preparation of cephradine.
    (56) Doxorubicin. The term ``doxorubicin working standard'' means a 
specific lot of a homogeneous preparation of doxorubicin.
    (57) Bleomycin. The term ``bleomycin working standard'' means a 
specific lot of a homogeneous preparation of bleomycin.
    (58) Tobramycin. The term ``tobramycin working standard'' means a 
specific lot of a homogeneous preparation of tobramycin.
    (59) Amikacin. The term ``amikacin working standard'' means a 
specific lot of a homogeneous preparation of amikacin.
    (60) Vidarabine. The term ``vidarabine working standard'' means a 
specific lot of a homogeneous preparation of vidarabine.
    (61) Ticarcillin. The term ``ticarcillin working standard'' means a 
specific lot of a homogeneous preparation of ticarcillin.
    (62) Cefadroxil. The term ``cefadroxil working standard'' means a 
specific lot of a homogeneous preparation of cefadroxil.
    (63) Natamycin. The term ``natamycin working standard'' means a 
specific lot of a homogeneous preparation of natamycin.
    (64) Cefoxitin. The term ``cefoxitin working standard'' means a 
specific lot of a homogeneous preparation of cefoxitin.
    (65) Cefamandole. The term ``cefamandole working standard'' means a 
specific lot of a homogeneous preparation of cefamandole.
    (66) Cefaclor. The term ``cefaclor working standard'' means a 
specific lot of a homogeneous preparation of cefaclor.
    (67) Cyclacillin. The term ``cyclacillin working standard'' means a 
specific lot of a homogeneous preparation of cyclacillin.
    (68) Daunorubicin. The term ``daunorubicin working standard'' means 
a specific lot of a homogeneous preparation of daunorubicin.
    (69) Sisomicin. The term ``sisomicin working standard'' means a 
specific lot of a homogeneous preparation of sisomicin.
    (70) Meclocycline. The term ``meclocycline working standard'' means 
a specific lot of a homogeneous preparation of meclocycline.
    (71) Cefotaxime. The term ``cefotaxime working standard'' means a 
specific lot of a homogeneous preparation of cefotaxime.
    (72) Mezlocillin. The term ``mezlocillin working standard'' means a 
specific lot of a homogeneous preparation of mezlocillin.
    (73) Moxalactam. The term ``moxalactam working standard'' means a 
specific lot of a homogeneous preparation of moxalactam.
    (74) Piperacillin. The term ``piperacillin working standard'' means 
a specific lot of a homogeneous preparation of piperacillin.
    (75) Azlocillin. The term ``azlocillin working standard'' means a 
specific lot of a homogeneous preparation of azlocillin.
    (76) Cefoperazone. The term ``cefoperazone working standard'' means 
a specific lot of a homogeneous preparation of cefoperazone.
    (77) Netilmicin. The term ``netimicin working standard'' means a 
specific lot of a homogeneous preparation of netilmicin.
    (78) Cefuroxime. The term ``cefuroxime working standard'' means a 
specific lot of a homogeneous preparation of cefuroxime.
    (79) Ceftizoxe. The term ``ceftizoxime working standard'' means a 
specific lot of a homogeneous preparation of ceftizoxime.

[[Page 324]]

    (80) 4-Epitetracycline. The term ``4-epitetracycline working 
standard'' means a specific lot of a homogeneous preparation of 4-
epitetracycline.
    (81) Chloramphenicol palmitate. The term ``chloramphenicol palmitate 
working standard'' means a specific lot of a homogeneous preparation of 
chloramphenicol palmitate.
    (82) Cyclosporine. The term ``cyclosporine working standard'' means 
a specific lot of a homogeneous preparation of cyclosporine.
    (83) Ceforanide. The term ``ceforanide working standard'' means a 
specific lot of a homogeneous preparation of ceforanide.
    (84) Cefonicid. The term ``cefonicid working standard'' means a 
specific lot of a homogeneous preparation of cefonicid.
    (85) Clavulanic acid. The term ``clavulanic acid working standard'' 
means a specific lot of a homogeneous preparation of clavulanic acid or 
a salt thereof.
    (86) Amdinocillin. The term ``amdinocillin working standard'' means 
a specific lot of a homogeneous preparation of amdinocillin.
    (87) Ceftriaxone. The term ``ceftriaxone working standard'' means a 
specific lot of a homogeneous preparation of ceftriaxone.
    (88) Ceftazidime. The term ``ceftazidime working standard'' means a 
specific lot of a homogeneous preparation of ceftazidime.
    (89) Imipenem. The term ``imipenem working standard'' means a 
specific lot of a homogeneous preparation of imipenem.
    (90) Cefotetan. The term ``cefotetan working standard'' means a 
specific lot of a homogeneous preparation of cefotetan.
    (91) Aztreonam. The term ``aztreonam working standard'' means a 
specific lot of a homogeneous preparation of aztreonam.
    (92) Sulbactam. The term ``sulbactam working standard'' means a 
specific lot of a homogeneous preparation of sulbactam.
    (93) Cefuroxime axetil. The term ``cefuroxime axetil working 
standard'' means a specific lot of a homogeneous preparation of 
cefuroxime axetil.
    (94) Cefmenoxime. The term ``cefmenoxime working standard'' means a 
specific lot of a homogeneous preparation of cefmenoxime.
    (95) Cefixime. The term ``cefixime working standard'' means a 
specific lot of a homogeneous preparation of cefixime.
    (96) Cefotiam. The term ``cefotiam working standard'' means a 
specific lot of a homogenous preparation of cefotiam.
    (97) Clindamycin phosphate. The term ``clindamycin phosphate working 
standard'' means a specific lot of a homogenous preparation of 
clindamycin phosphate.
    (98) Mupirocin. The term ``mupirocin working standard'' means a 
specific lot of a homogeneous preparation of mupirocin or a salt 
thereof.
    (99) Cefmetazole. The term ``cefmetazole working standard'' means a 
specific lot of a homogeneous preparation of cefmetazole.
    (100) Cefpiramide. The term ``cefpiramide working standard'' means a 
specific lot of a homogeneous preparation of cefpiramide.
    (101) Clarithromycin. The term ``clarithromycin working standard'' 
means a specific lot of a homogeneous preparation of clarithromycin.
    (102) Azithromycin. The term ``azithromycin working standard'' means 
a specific lot of a homogeneous preparation of azithromycin.
    (103) Cefprozil. The term ``cefprozil (Z) working standard'' means a 
specific lot of a homogeneous preparation of cefprozil (Z). The term 
``cefprozil (E) working standard'' means a specific lot of a homogeneous 
preparation of cefprozil (E).
    (104) Idarubicin. The term ``idarubicin working standard'' means a 
specific lot of a homogeneous preparation of idarubicin.
    (105) Loracarbef. The term ``loracarbef working standard'' means a 
specific lot of a homogeneous preparation of loracarbef.
    (106) Rifabutin. The term ``rifabutin working standard'' means a 
specific lot of a homogeneous preparation of rifabutin.

[[Page 325]]

    (107) Cefpodoxime proxetil. The term ``cefpodoxime proxetil working 
standard'' means a specific lot of a homogeneous preparation of 
cefpodoxime proxetil.
[39 FR 18925, May 30, 1974]

    Editorial Note: For Federal Register citations affecting Sec. 430.5, 
see the List of CFR Sections Affected appearing in the Finding Aids 
section of this volume.



Sec. 430.6  Definitions of the terms ``unit'' and ``microgram'' as applied to antibiotic substances.

    Unless it has been otherwise specified in the individual definitions 
in this section, the activity assigned to each ``unit'' or ``microgram'' 
is equivalent to an International Unit, if such has been defined by the 
World Health Organization.
    (a) ``Unit''--(1) Penicillin--(i) Penicillin G. The term ``unit'' 
applies to penicillin G means the penicillin activity (potency) 
contained in 0.600 microgram of the penicillin G master standard.
    (ii) [Reserved]
    (iii) Penicillin V. The term ``unit'' applied to penicillin V means 
the penicillin activity (potency) contained in 0.590 microgram of the 
penicillin V master standard.
    (2) Bacitracin. The term ``unit'' applied to bacitracin means a 
bacitracin activity (potency) contained in 13.51 micrograms of the 
bacitracin master standard, except that when the activity (potency) of 
bacitracin is expressed in terms of its weight, as in the feed and 
drinking water of animals, 1 gram of activity is equivalent to 42,000 
units.
    (3) Nystatin. The term ``unit'' applied to nystatin means the 
nystatin activity (potency) contained in 0.2817 microgram of the 
nystatin master standard when dried for 2 hours at 40 deg. C. and a 
pressure of 5 millimeters or less.
    (4) Polymyxin B. The term ``unit'' applied to polymyxin B means the 
polymyxin activity (potency) contained in 0.1274 microgram of the 
polymyxin B master standard when dried for 3 hours at 60 deg. C. and a 
pressure of 5 millimeters or less.
    (5) Bleomycin. The term ``unit'' applied to bleomycin means the 
bleomycin activity (potency) contained in 0.637 milligram of the 
bleomycin master standard.
    (b) ``Microgram''--(1) Streptomycin. The term ``microgram'' applied 
to streptomycin means the streptomycin activity (potency) contained in 
1.250 micrograms of the streptomycin master standard after it is dried 
for 3 hours at 60 deg. C. and a pressure of 5 millimeters or less.
    (2) Dihydrostreptomycin. The term ``microgram'' applied to 
dihydrostreptomycin means the dihydrostreptomycin activity (potency) 
contained in 1.25 micrograms of the dihydrostreptomycin master standard 
after it is dried for 4 hours at 100 deg. C. and a pressure of 50 
microns or less.
    (3) Chlortetracycline. The term ``microgram'' applied to 
chlortetracycline means the chlortetracycline activity (potency) 
contained in 1.0 microgram of the chlortetracycline master standard.
    (4) Demeclocycline. The term ``microgram'' applied to demeclocycline 
means the demeclocycline activity (potency) contained in 1.0 microgram 
of the demeclocycline master standard after it is dried for 3 hours at 
60 deg. C. and a pressure of 5 millimeters or less.
    (5) Tetracycline. The term ``microgram'' applied to tetracycline 
means the tetracycline activity (potency) contained in 1.0 microgram of 
tetracycline master standard.
    (6) Rolitetracycline. The term ``microgram'' applied to 
rolitetracycline means the rolitetracycline activity (potency) contained 
in 1.0 microgram of the rolitetracycline master standard when dried for 
3 hours at 60 deg. C. and a pressure of 5 millimeters or less.
    (7) Chloramphenicol. The term ``microgram'' applied to 
chloramphenicol means the chloramphenicol activity (potency) contained 
in 1.0 microgram of the chloramphenicol master standard.
    (8) Methicillin. The term ``microgram'' applied to methicillin means 
the methicillin activity (potency) contained in 1.105 micrograms of the 
methicillin master standard.
    (9) Oxacillin. The term ``microgram'' applied to oxacillin means the 
oxacillin activity (potency) contained in 1.111

[[Page 326]]

micrograms of the oxacillin master standard.
    (10) [Reserved]
    (11) Amphotericin A. The term ``microgram'' applied to amphotericin 
A means the amphotericin A activity (potency) contained in 1.0 microgram 
of the amphotericin A master standard when dried for 3 hours at 60 deg. 
C. and a pressure of 5 millimeters or less.
    (12) Amphotericin B. The term ``microgram'' applied to amphotericin 
B means the amphotericin B activity (potency) contained in 1.014 
micrograms of the amphotericin B master standard when dried for 3 hours 
at 60 deg. C. and a pressure of 5 millimeters or less.
    (13) Colistin. The term ``microgram'' applied to colistin means the 
colistin base activity (potency) contained in 1.495 micrograms of the 
colistin master standard when dried for 3 hours at 60 deg. C. and a 
pressure of 5 millimeters or less. The numerical value of a microgram of 
colistin is not equivalent to the International Unit.
    (14) Colistimethate. The term ``microgram'' applied to 
colistimethate means the activity (potency) calculated as colistin base 
that is contained in 1.938 micrograms of the colistimethate master 
standard when dried for 3 hours at 60 deg. C. and a pressure of 5 
millimeters or less. The numerical value of a microgram of 
colistimethate is not equivalent to the International Unit.
    (15) Cycloserine. The term ``microgram'' applied to cycloserine 
means the cycloserine activity (potency) contained in 1.0 microgram of 
the cycloserine master standard when dried for 3 hours at 60 deg. C. and 
a pressure of 5 millimeters or less.
    (16) Erythromycin. The term ``microgram'' applied to erythromycin 
means the erythromycin base activity (potency) contained in 1.02 
micrograms of the erythromycin master standard when dried for 3 hours at 
60 deg. C. and a pressure of 5 millimeters or less.
    (17) Gramicidin. The term ``microgram'' applied to gramicidin means 
the gramicidin activity (potency) contained in 1.0 microgram of the 
gramicidin master standard when dried for 3 hours at 60 deg. C. and a 
pressure of 5 millimeters or less.
    (18) Griseofulvin. The term ``microgram'' applied to griseofulvin 
means the griseofulvin activity (potency) contained in 1.0 microgram of 
the griseofulvin master standard.
    (19) Kanamycin. The term ``microgram'' applied to kanamycin means 
the kanamycin base activity (potency) contained in 1.299 micrograms of 
the kanamycin master standard.
    (20) Neomycin. The term ``microgram'' applied to neomycin means the 
neomycin base activity (potency) contained in 1.429 micrograms of the 
neomycin master standard when dried for 3 hours at 60 deg. C. and a 
pressure of 5 millimeters or less.
    (21) Novobiocin. The term ``microgram'' applied to novobiocin means 
the novobiocin acid activity (potency) contained in 1.033 micrograms of 
the novobiocin master standard when dried for 3 hours at 60 deg. C. and 
a pressure of 5 millimeters or less.
    (22) Oleandomycin. The term ``microgram'' applied to oleandomycin 
means the oleandomycin base activity (potency) contained in 1.176 
micrograms of the oleandomycin master standard.
    (23) Troleandomycin. The term ``microgram'' applied to 
troleandomycin means the activity (potency), calculated as the molecular 
equivalent of the oleandomycin base, contained in 1.2315 micrograms of 
the troleandomycin master standard.
    (24) Oxytetracycline. The ``microgram'' applied to oxytetracycline 
means the oxytetracycline base activity (potency) contained in 1.13 
micrograms of the oxytetracycline master standard.
    (25) Paromomycin. The term ``microgram'' applied to paromomycin 
means the paromomycin activity (potency) contained in 1.333 micrograms 
of the paromomycin master standard when dried for 3 hours at 60 deg. C. 
and a pressure of 5 millimeters or less.
    (26) Tyrothricin. The term ``microgram'' applied to tyrothricin 
means the activity (potency) contained in 0.2 microgram of the 
gramicidin master standard when dried for 3 hours at 60 deg. C. and a 
pressure of 5 millimeters or less.

[[Page 327]]

    (27) Vancomycin. The term ``microgram'' applied to vancomycin means 
the vancomycin base activity (potency) contained in 1.25 micrograms of 
the vancomycin master standard.
    (28) [Reserved]
    (29) Ampicillin. The term ``microgram'' applied to ampicillin means 
the ampicillin activity (potency) contained in 1.1764 micrograms of the 
ampicillin master standard.
    (30) Nafcillin. The term ``microgram'' applied to nafcillin means 
the nafcillin activity (potency) contained in 1.0989 micrograms of the 
nafcillin master standard.
    (31) Gentamicin. The term ``microgram'' applied to gentamicin means 
the gentamicin activity (potency) contained in 1.56 micrograms of the 
gentamicin master standard when dried for 3 hours at 110 deg. C. and a 
pressure of 5 millimeters or less.
    (32) Dactinomycin. The term ``microgram'' applied to dactinomycin 
means the dactinomycin activity (potency) contained in 1.000 microgram 
of the dactinomycin master standard when dried for 3 hours at 60 deg. C. 
and a pressure of 5 millimeters or less.
    (33) Candicidin. The term ``microgram'' applied to candicidin means 
the candicidin activity (potency) contained in 1.0 microgram of the 
candicidin master standard when dried for 3 hours at 40 deg. C. and a 
pressure of 5 millimeters or less.
    (34) Cephalothin. The term ``microgram'' applied to cephalothin 
means the cephalothin activity (potency) contained in 1.056 micrograms 
of the cephalothin master standard when dried for 3 hours at 60 deg. C. 
and a pressure of 5 millimeters or less.
    (35) Lincomycin. The term ``microgram'' applied to lincomycin means 
the lincomycin base activity (potency) contained in 1.156 micrograms of 
the lincomycin master standard.
    (36) Cloxacillin. The term ``microgram'' applied to cloxacillin 
means the cloxacillin activity (potency) contained in 1.135 micrograms 
of the cloxacillin master standard.
    (37) Methacycline. The term ``microgram'' applied to methacycline 
means the methacycline activity (potency) contained in 1.082 micrograms 
of the methacycline master standard when dried for 3 hours at 60 deg. C. 
and a pressure of 5 millimeters or less.
    (38) Doxycycline. The term ``microgram'' applied to doxycycline 
means the doxycycline activity (potency) contained in 1.155 micrograms 
of the doxycycline master standard.
    (39) Cephaloridine. The term ``microgram'' applied to cephaloridine 
means the cephaloridine activity (potency) contained in 1.00806 
micrograms of the cephaloridine master standard when dried for 3 hours 
at 60 deg. C. and a pressure of 5 millimeters or less.
    (40) Dicloxacillin. The term ``microgram'' applied to dicloxacillin 
means the dicloxacillin activity (potency) contained in 1.087 micrograms 
of the dicloxacillin master standard.
    (41) Plicamycin. The term ``microgram'' applied to plicamycin means 
the plicamycin activity (potency) contained in 1.000 microgram of the 
plicamycin master standard when dried for 4 hours at 25 deg. C. and a 
pressure of 5 millimeters or less.
    (42) Clindamycin. The term ``microgram'' applied to clindamycin 
means the clindamycin activity (potency) contained in 1.139 micrograms 
of the clindamycin master standard.
    (43) Cephaloglycin. The term ``microgram'' applied to cephaloglycin 
means the cephaloglycin activity (potency) contained in 1.02564 
micrograms of the cephaloglycin master standard.
    (44) Carbenicillin. The term ``microgram'' applied to carbenicillin 
means the carbenicillin activity (potency) contained in 1.135 micrograms 
of the carbenicillin master standard.
    (45) Cephalexin. The term ``microgram'' applied to cephalexin means 
the cephalexin activity (potency) contained in 1.0707 micrograms of the 
cephalexin master standard.
    (46) [Reserved]
    (47) Capreomycin. The term ``microgram'' applied to capreomycin 
means the capreomycin activity (potency) contained in 1.0870 micrograms 
of the capreomycin master standard when dried for 4 hours at 100 deg. C. 
and a pressure of 5 millimeters or less.
    (48) Rifampin. The term ``microgram'' applied to rifampin means the 
rifampin activity (potency) contained in 1.0101

[[Page 328]]

micrograms of the rifampin master standard.
    (49) Minocycline. The term ``microgram'' applied to minocycline 
means the minocycline activity (potency) contained in 1.1588 micrograms 
of the minocycline master standard.
    (50) Spectinomycin. The term ``microgram'' applied to spectinomycin 
means the spectinomycin activity (potency) contained in 1.490 micrograms 
of the spectinomycin master standard.
    (51) Clindamycin palmitate hydrochloride. The term ``microgram'' 
applied to clindamycin palmitate hydrochloride means the clindamycin 
activity (potency) contained in 1.661 micrograms of the clindamycin 
palmitate hydrochloride master standard.
    (52) Carbenicillin indanyl. The term ``microgram'' applied to 
carbenicillin indanyl means the carbenicillin activity (potency) 
contained in 1.4514 micrograms of the carbenicillin indanyl master 
standard.
    (53) Cephapirin. The term ``microgram'' applied to cephapirin means 
the cephapirin activity (potency) contained in 1.0616 micrograms of the 
cephapirin master standard.
    (54) Cefazolin. The term ``microgram'' applied to cefazolin means 
the cefazolin activity (potency) contained in 1.005 micrograms of the 
cefazolin master standard.
    (55) Mitomycin. The term ``microgram'' applied to mitomycin means 
the mitomycin activity (potency) contained in 1.0416 micrograms of the 
mitomycin master standard.
    (56) Amoxicillin. The term ``microgram'' applied to amoxicillin 
means the amoxicillin activity (potency) contained in 1.17647 micrograms 
of the amoxicillin master standard.
    (57)  [Reserved]
    (58) Cephradine. The term ``microgram'' applied to cephradine means 
the cephradine activity (potency) contained in 1.1111 micrograms of the 
cephradine master standard.
    (59) Doxorubicin. The term ``microgram'' applied to doxorubicin 
means the activity (potency) calculated as doxorubicin hydrochloride 
contained in 1.0204 micrograms of the doxorubicin master standard.
    (60) Tobramycin. The term ``microgram'' applied to tobramycin means 
the tobramycin activity (potency) contained in 1.126 micrograms of the 
tobramycin master standard.
    (61) Amikacin. The term ``microgram'' applied to amikacin means the 
amikacin activity (potency) contained in 1.091 micrograms of the 
amikacin master standard.
    (62) Vidarabine. The term ``microgram'' applied to vidarabine means 
the vidarabine activity (potency) contained in 1.0674 micrograms of the 
vidarabine master standard.
    (63) Ticarcillin. The term ``microgram'' applied to ticarcillin 
means the ticarcillin activity (potency) contained in 1.136 micrograms 
of the ticarcillin master standard.
    (64) Cefadroxil. The term ``microgram'' applied to cefadroxil means 
the cefadroxil activity (potency) contained in 1.0537 micrograms of the 
cefadroxil master standard.
    (65) Natamycin. The term ``microgram'' applied to natamycin means 
the natamycin activity (potency) contained in 1.0846 micrograms of the 
natamycin master standard.
    (66) Cefoxitin. The term ``microgram'' applied to cefoxitin means 
the cefoxitin activity (potency) contained in 1.072 micrograms of the 
cefoxitin master standard.
    (67) Cefamandole. The term ``microgram'' applied to cefamandole 
means the cefamandole activity (potency) contained in 1.1364 micrograms 
of cefamandole master standard.
    (68) Cefaclor. The term ``microgram'' applied to cefaclor means the 
cefaclor activity (potency) contained in 1.0493 micrograms of cefaclor 
master standard.
    (69) Cyclacillin. The term ``microgram'' applied to cyclacillin 
means the cyclacillin activity (potency) contained in 1.01 micrograms of 
the cyclacillin master standard.
    (70) Daunorubicin. The term ``microgram'' applied to daunorubicin 
means the daunorubicin activity (potency) contained in 1.0965 micrograms 
of the daunorubicin master standard.
    (71) Sisomicin. The term ``microgram'' applied to sisomicin means 
the sisomicin activity (potency) contained

[[Page 329]]

in 1.00 microgram of the sisomicin master standard expressed on an 
anhydrous basis.
    (72) Meclocycline. The term ``microgram'' applied to meclocycline 
means the meclocycline activity (potency) contained in 1.0493 micrograms 
of the meclocycline master standard.
    (73) Cefotaxime. The term ``microgram'' applied to cefotaxime means 
the cefotaxime activity (potency) contained in 1.089 micrograms of 
cefotaxime master standard.
    (74) Mezlocillin. The term ``microgram'' applied to mezlocillin 
means the mezlocillin activity (potency) contained in 1.1086 micrograms 
of the mezlocillin master standard.
    (75) Moxalactam. The term ``microgram'' applied to moxalactam means 
the moxalactam activity (potency) contained in 1.1173 micrograms of the 
moxalactam master standard.
    (76) Piperacillin. The term ``microgram'' applied to piperacillin 
means the piperacillin activity (potency) contained in 1.0460 micrograms 
of the piperacillin master standard.
    (77) Cefoperazone. The term ``microgram'' applied to cefoperazone 
means the cefoperazone activity (potency) contained in 1.056 micrograms 
of the cefoperazone master standard.
    (78) Azlocillin. The term ``microgram'' applied to azlocillin means 
the azlocillin activity (potency) contained in 1.128 micrograms of the 
azlocillin master standard.
    (79) Netilmicin. The term ``microgram'' applied to netilmicin means 
the netilmicin activity (potency) contained in 1.000 microgram of the 
netilmicin master standard expressed on an anhydrous basis.
    (80) Cefuroxime. The term ``microgram'' applied to cefuroxime means 
the cefuroxime activity (potency) contained in 1.0893 micrograms of the 
cefuroxime master standard.
    (81) Ceftizoxime. The term ``microgram'' applied to ceftizoxime 
means the ceftizoxime activity (potency) contained in 1.011 micrograms 
of the ceftizoxime master standard.
    (82) Cyclosporine. The term ``microgram'' applied to cyclosporine 
means the cyclosporine activity (potency) contained in 1.0173 micrograms 
of cyclosporine master standard.
    (83) Ceforanide. The term ``microgram'' applied to ceforanide means 
the ceforanide activity (potency) contained in 1.005 micrograms of the 
ceforanide master standard.
    (84) Cefonicid. The term ``microgram'' applied to cefonicid means 
the cefonicid activity (potency) contained in 1.150 micrograms of the 
cefonicid master standard.
    (85) Clavulanic acid. The term ``microgram'' applied to clavulanic 
acid means the clavulanic acid activity (potency) contained in 1.053 
micrograms of clavulanic acid master standard.
    (86) Amdinocillin. The term ``microgram'' applied to amdinocillin 
means the amdinocillin activity (potency) contained in 1.004 micrograms 
of the amdinocillin master standard.
    (87) Ceftriaxone. The term ``microgram'' applied to ceftriaxone 
means the ceftriaxone activity (potency) contained in 1.19 micrograms of 
the ceftriaxone master standard.
    (88) Ceftazidime. The term ``microgram'' applied to ceftazidime 
means the ceftazidime activity (potency) contained in 1.1834 micrograms 
of the ceftazidime master standard.
    (89) Imipenem. The term ``microgram'' applied to imipenem 
monohydrate means the imipenem activity (potency) contained in 1.085 
micrograms of the imipenem master standard.
    (90) Cefotetan. The term ``microgram'' applied to cefotetan means 
the cefotetan activity (potency) contained in 1.012 micrograms of the 
cefotetan master standard.
    (91) Aztreonam. The term ``microgram'' applied to aztreonam means 
the aztreonam activity (potency) contained in 1.05 micrograms of the 
aztreonam master standard.
    (92) Sulbactam. The term ``microgram'' applied to sulbactam means 
the sulbactam activity (potency) contained in 1.002 micrograms of the 
sulbactam master standard.
    (93) Cefuroxime axetil. The term ``microgram'' applied to cefuroxime 
axetil means the cefuroxime activity (potency) contained in 1.246 
micrograms of the cefuroxime axetil master standard.
    (94) Cefmenoxime. The term ``microgram'' applied to cefmenoxime

[[Page 330]]

means the cefmenoxime activity (potency) contained in 1.0482 micrograms 
of the cefmenoxime master standard.
    (95) Cefixime. The term ``microgram'' applied to cefixime means the 
cefixime activity (potency) contained in 1.126 micrograms of the 
cefixime master standard.
    (96) Cefotiam. The term ``microgram'' applied to cefotiam means the 
cefotiam (potency) contained in 1.144 micrograms of the cefotiam master 
standard.
    (97) Clindamycin phosphate. The term ``microgram'' applied to 
clindamycin phosphate means the clindamycin phosphate (potency) 
contained in 1.252 micrograms of the clindamycin phosphate master 
standard.
    (98) Mupirocin. The term ``microgram'' applied to mupirocin means 
the activity (potency) calculated as mupirocin activity (potency) 
contained in 1.075 micrograms of the mupirocin master standard.
    (99) Cefmetazole. The term ``microgram'' applied to cefmetazole 
means the cefmetazole (potency) contained in 1.002 micrograms of the 
cefmetazole master standard.
    (100) Cefpiramide. The term ``microgram'' applied to cefpiramide 
means the cefpiramide (potency) contained in 0.994 microgram of the 
cefpiramide master standard.
    (101) Clarithromycin. The term ``microgram'' applied to 
clarithromycin means the clarithromycin (potency) contained in 1.010 
micrograms of the clarithromycin master standard.
    (102) Azithromycin. The term ``microgram'' applied to azithromycin 
means the azithromycin (potency) contained in 1.063 micrograms of the 
azithromycin master standard.
    (103) Cefprozil. The term ``microgram'' applied to cefprozil (Z) 
means the cefprozil (Z) potency contained in 1.060 micrograms of the 
cefprozil (Z) master standard. The term ``microgram'' applied to 
cefprozil (E) means the cefprozil (E) potency contained in 1.106 
micrograms of the cefprozil (E) master standard.
    (104) Idarubicin. The term ``microgram'' applied to idarubicin means 
the idarubicin activity (potency) calculated as idarubicin hydrochloride 
contained in 1.036 micrograms of the idarubicin master standard.
    (105) Loracarbef. The term ``microgram'' applied to loracarbef means 
the loracarbef (potency) contained in 1.059 micrograms of the loracarbef 
master standard.
    (106) Rifabutin. The term ``microgram'' applied to rifabutin means 
the rifabutin (potency) contained in 1.022 micrograms of the rifabutin 
master standard.
    (107) Cefpodoxime proxetil. The term ``microgram'' applied to 
cefpodoxime proxetil means the cefpodoxime (potency) contained in 1.304 
micrograms of the cefpodoxime proxetil master standard when dried.
[39 FR 18925, May 30, 1974]

    Editorial Note: For Federal Register citations affecting Sec. 430.6, 
see the List of CFR Sections Affected appearing in the Finding Aids 
section of this volume.



   Subpart B--Antibiotic Drugs Affected by the Drug Amendments of 1962



Sec. 430.10  Certification or release of antibiotic drugs affected by the drug amendments of 1962.

    (a) Before the 1962 amendments to it, the Federal Food, Drug, and 
Cosmetic Act only permitted the Food and Drug Administration to provide 
for the certification of batches of antibiotic drugs containing 
penicillin, streptomycin, chlortetracycline, chloramphenicol, or 
bacitracin, or any derivative of them. FDA certified those drugs under 
regulations promulgated on the basis of scientific proof of the drugs' 
safety and effectiveness. Most drugs containing an antibiotic other than 
one of those listed were subject to the new drug provisions of the act, 
which required that an applicant show that the drug was safe and obtain 
FDA approval of a new drug application before marketing it. An 
affirmative showing of effectiveness was not then required to obtain 
approval. Some antibiotic drugs that were not subject to certification, 
however, were also not subject to the new drug provisions of the act 
under informal FDA opinions that the drug was ``not a new drug'' or ``no 
longer a new drug.'' FDA

[[Page 331]]

revoked those opinions under Sec. 310.100 of this chapter.
    (b) The 1962 amendments amended section 507 of the act to require 
the certification, release without certification, or exemption from 
certification, of all antibiotic drugs on the basis of scientific proof 
of safety and effectiveness. The amendments provided that FDA implement 
them for antibiotic drugs that were marketed on April 30, 1963 and were 
not subject to the certification provisions on that date. FDA is 
implementing the amendments with respect to antibiotic drugs formerly 
subject to the new drug provisions of the act through its Drug Efficacy 
Study Implementation (DESI) program under which the agency is evaluating 
those antibiotic drugs for efficacy. Until FDA completes that evaluation 
it will permit continued marketing of those antibiotic drugs under 
paragraph (c) of this section. The agency is also implementing the 1962 
amendments with respect to antibiotic drugs formerly not subject to 
either the certification or new drug provisions of the act and the 
agency is evaluating those antibiotic drugs for both safety and 
efficacy. Until FDA completes that evaluation, it will permit continued 
marketing of those antibiotic drugs under paragraph (d) of this section.
    (c) Unless exempted from certification, FDA will certify or release 
antibiotic drugs which on April 30, 1963 were the subject of an approved 
new drug application under section 505 of the act, under regulations 
providing for certification of the drugs. Although the initial 
regulation for each of these drugs established under section 507(h) of 
the act was not conditioned upon an affirmative finding of the 
effectiveness of the drug, FDA is proceeding under its DESI program to 
amend or repeal those regulations to provide for certification of those 
drugs only if they had been shown to be both safe and effective.
    (d) Unless exempted from certification, FDA will release without 
certification an antibiotic drug that was marketed on April 30, 1963, 
but not subject to certification, and not subject to an approved new 
drug application on that date, unless FDA has made a determination that 
the drug has not been shown to be safe or lacks substantial evidence of 
effectiveness under the DESI program. FDA is proceeding under its DESI 
program to establish regulations under section 507 to provide for 
certification of those drugs only if they have been shown to be safe and 
effective.
[50 FR 7516, Feb. 22, 1985]



PART 431--CERTIFICATION OF ANTIBIOTIC DRUGS--Table of Contents




                      Subpart A--General Provisions

Sec.
431.1  Requests for certification, check tests and assays, and working 
          standards; information and samples required.
431.5  Samples for sterility testing.
431.10  Certification.
431.11  Conditions on the effectiveness of certificates.
431.12  Certification of antibiotic drugs after shipment in bulk 
          containers.
431.17  Request to provide for certification of an antibiotic drug.
431.20  Disposition of outdated drugs.

                  Subpart B--Administrative Procedures

431.50  Forms for certification or exemption of antibiotic drugs.
431.51  Suspension of certification service.
431.52  Hearings.
431.53  Fees.

                     Subpart C--Records and Reports

431.61  Records of distribution.
431.62  Records retention.

                Subpart D--Confidentiality of Information

431.70  Confidentiality of data and information in an investigational 
          new drug notice for an antibiotic drug.

    Authority:  21 U.S.C. 351, 352, 353, 355, 357, 379e; 42 U.S.C. 216, 
241, 262; 5 U.S.C. 552.

    Source:  39 FR 18934, May 30, 1974, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 431.1  Requests for certification, check tests and assays, and working standards; information and samples required.

    (a) A request for certification of a batch (antibiotic Form 7/Form 
FDA-1677) is to be addressed to the Food and

[[Page 332]]

Drug Administration, Division of Research and Testing (HFD-470), 200 C 
St. SW., Washington, DC 20204.
    (b) [Reserved]
    (c) A person who requests certification or check tests and assays of 
a batch shall submit with his request the following information and 
samples:
    (1) The batch mark of the drug.
    (2) The quantity of each ingredient used in making the batch and a 
statement that each such ingredient conforms to the requirements or 
standards prescribed therefor, if any, by specific regulations or 
official compendium or otherwise approved by the Commissioner.
    (3) The size of the batch, including the number of containers of 
each size in the batch.
    (4) The date of the latest assay of the batch.
    (5) The results of the latest tests and assays made by or for him on 
the batch as required for the drug by specific regulations.
    (6) The batch mark(s) of the antibiotic(s) used in making the batch.
    (7) Unless previously submitted, the results and dates of the latest 
tests and assays made by or for him on the antibiotic(s) used in making 
the batch as required by specific regulations.
    (8) The number of accurately representative samples that are 
required for the batch by specific regulations:
    (i) In the case of drugs such as dry powders, solutions, ointments, 
and suspensions, the sample shall be collected by taking single 
immediate containers, before or after labeling, at such intervals 
throughout the entire time of packaging the batch that the quantities 
packaged during the intervals are approximately equal. In no case, 
however, shall more than 5,000 immediate containers have been packaged 
during each such interval of sampling, except for a sample collected for 
sterility testing.
    (ii) In the case of drugs in unit dosage forms, such as tablets, 
capsules, or suppositories, samples shall be collected as follows:
    (a) From batches exceeding 500,000 units, a representative sample 
consisting of 100 units shall be collected by taking single units at 
approximately equal intervals throughout the final production of the 
batch. If the person packaging the units into dispensing-size containers 
is not the manufacturer, the representative sample consisting of 100 
units shall be collected by taking single units at approximately equal 
intervals during packaging.
    (b) From batches of 500,000 units or less, a representative sample 
consisting of not more than 100 units shall be collected by taking 
single units at approximately equal intervals throughout the final 
production of the batch. If the person packaging the units into 
dispensing-size containers is not the manufacturer, the samples shall be 
collected by taking single units at approximately equal intervals during 
packaging. In no case shall more than 5,000 units be produced or 
packaged during a sampling interval. The minimum acceptable sample size 
shall be as specified in the appropriate monograph.
    (c) When the manufacturing process is such that it is not feasible 
to collect the samples throughout the final production of the batch 
(e.g., if tablets undergo further processing, such as polishing or 
coating, after being compressed), the samples may be collected from bulk 
containers of the finished product, according to the following 
requirements:
    (1) For batches exceeding 500,000 units: If the batch is in more 
than 100 containers, the sample is 1 unit from each container. If the 
batch is in 100 containers or less, the sample is 100 units, taken in 
approximately equal amounts from each container.
    (2) For batches of 500,000 units or less: If the batch is in more 
than 100 containers, the sample is 1 unit from each container. If the 
batch is in 100 containers or less, the sample is at least 1 unit for 
every 5,000 units in the batch taken in approximately equal amounts from 
each container. The sample shall not be less than the minimum number of 
units specified in the appropriate monograph.
    (iii) In the case of drugs packaged for repacking or for use in the 
manufacture of another drug, the sample must be representative of the 
batch. Such samples may be taken from a composite composed of portions 
taken from a

[[Page 333]]

representative number of bulk containers, the composite consisting of no 
more than 10 times the amount required for conducting the required tests 
and assays. Such samples are not required if they have been previously 
submitted.
    (iv) In the case of a sterile drug packaged in combination with 
containers of a sterile diluent, the sample shall be collected by taking 
20 immediate containers of the diluent collected at regular intervals 
throughout each filling operation, except that if the diluent is 
sterilized after filling into containers, the representative sample 
shall consist of 20 immediate containers collected from each sterilizer 
load and each container shall be taken from a different part of each 
such sterilizer load. In the case of sterile drugs packaged in 
combination with sterile dispensers, the sample shall be collected by 
taking 20 dispensers from each sterilizer load, and each dispenser shall 
be taken from a different part of such sterilizer load.
    (9) In the case of an initial request for certification, each 
ingredient used in making the batch other than ingredients required by 
specific regulations: 1 package of each containing approximately 5 
grams. Results and dates of the latest tests and assays made by or for 
him on such ingredients shall precede or accompany the submission.
    (10) The results and dates of tests and assays made by or for him on 
the nonantibiotic active ingredients in the batch.
    (11) If such batch or any part thereof is to be packaged with a 
sterile diluent or sterile dispenser, such request shall also be 
accompanied by a statement that such diluent or dispenser is sterile and 
conforms to the requirements prescribed therefor by specific 
regulations.
    (d) Each sample submitted pursuant to the regulations in this 
chapter shall be addressed to the Commissioner. Its package shall be 
clearly identified as to its contents and shall bear the name and post-
office address of the person submitting it.
    (e) In addition to the information and samples specifically required 
to be submitted to the Commissioner by the regulations in this chapter, 
the person who requests certification of a batch shall submit such 
further information and samples as the Commissioner may require for the 
purpose of investigations to determine whether or not such batch 
complies with the requirements of Sec. 431.10 for the issuance of a 
certificate.
    (f) Reference standards identical to working standards are available 
from: U.S.P. Reference Standards, 12601 Twinbrook Parkway, Rockville, 
Md. 20857, 301-881-0666.
[39 FR 18934, May 30, 1974, as amended at 41 FR 46852, Oct. 26, 1976; 43 
FR 41195, 41197, Sept. 15, 1978; 45 FR 40111, June 30, 1980; 50 FR 7516, 
Feb. 22, 1985; 50 FR 8997, Mar. 6, 1985; 55 FR 11582, Mar. 29, 1990]



Sec. 431.5  Samples for sterility testing.

    (a) ``Filling operation'' and ``sample'' defined. (1) The term 
``filling operation'' when used in connection with samples of a batch 
required for sterility testing refers to that period of time not longer 
than 24 consecutive hours during which a homogeneous quantity of drug is 
being filled continuously into market-size containers and during which 
no changes are made in the equipment used for filling. (Short rest 
periods for operators of the filling equipment and the time required to 
change operators between consecutive shifts are not considered as a 
break in continuity of the filling operation.) If more than one filling 
device is used during the filling operation, the samples shall include 
immediate containers filled by each device, and each such container 
shall be identified with a mark corresponding to that assigned to the 
filling device. If more than one filling operation is required to fill a 
batch, each container in the sample shall be identified with the number 
of the operation.
    (2) For the purpose of sterility testing, the term ``sample'' means 
the total number of containers taken from each filling operation.
    (b) Packaging requirements for samples. If a batch of a sterile 
antibiotic is packaged for repacking or for use as an ingredient in the 
manufacture of another drug, the sample required for sterility testing 
may be packaged in one container, in lieu of 20 containers, or in two 
containers in lieu of 40 containers, under the following conditions:

[[Page 334]]

    (1) The weight or volume of the sample is equivalent to the 
composite weight or volume required for a multiple container sample;
    (2) The sample is a composite of samples taken from all parts of the 
batch; and
    (3) The sterility test method prescribed for the drug by the 
regulations in this chapter is ``Bacterial membrane filter method'' 
described in Sec. 436.20(e)(1) of this chapter.



Sec. 431.10  Certification.

    (a) If it appears to the Commissioner, after such investigation as 
he considers necessary, that:
    (1) The information (including results of tests and assays) and 
samples required by or pursuant to the regulations in this chapter have 
been submitted, and the request for certification contains no untrue 
statement of a material fact; and
    (2) The batch complies with the regulations in this chapter and 
conforms to the applicable standards of identity, strength, quality, and 
purity prescribed by the regulations in this chapter;

the Commissioner shall certify that such batch is safe and efficacious 
for use, subject to such conditions on the effectiveness of certificates 
as are prescribed by Sec. 431.11 and shall issue to the person who 
requested it a certificate to that effect.
    (b) If the Commissioner determines, after such investigation as he 
considers to be necessary, that the information submitted pursuant to 
the regulations in this chapter, or the batch covered by such request, 
does not comply with the requirements set forth in paragraph (a) of this 
section for the issuance of a certificate, the Commissioner shall refuse 
to certify such batch and shall give notice thereof to the person who 
requested certification, stating his reasons for refusal.
    (c) All statements, samples, and other information and materials 
submitted in connection with a request for certification shall be 
considered to be part of such request.
    (d) Compliance of a drug with the standards of identity, strength, 
quality, and purity prescribed by regulations in this chapter shall be 
determined by the tests and methods of assay prescribed for such drug by 
regulations issued under this chapter.
    (e) The regulations in this chapter, prescribing tests and methods 
of assay for antibiotic and antibiotic-containing drugs, shall not be 
construed as preventing the Commissioner from using any other test or 
method of assay in his investigations to determine whether or not:
    (1) A request for certification contains any untrue statement of a 
material fact; or
    (2) A certification has been obtained through fraud, or through 
misrepresentation or concealment of a material fact.
    (f) Except as specifically provided by the regulations in this 
chapter, no provision of any regulation shall be construed as exempting 
any certifiable antibiotic drug from any applicable provision of the act 
or any regulation thereunder.



Sec. 431.11  Conditions on the effectiveness of certificates.

    (a) A certificate shall not become effective:
    (1) If it is obtained through fraud or through misrepresentation or 
concealment of a material fact;
    (2) With respect to any package unless it complies with the 
packaging requirements, if any, prescribed by the regulations in this 
chapter which were in effect on the date of the certificate;
    (3) With respect to any package unless its label and labeling bear 
all words, statements, and other information required by the regulations 
in this chapter; or
    (4) With respect to any package of a certifiable antibiotic drug 
subject to the regulations in this chapter, when it is included in a 
packaged combination with another drug, unless such other drug complies 
with the requirements of the regulations in this chapter.
    (b) A certificate shall cease to be effective:
    (1) With respect to any immediate container after the expiration 
date, if any, prescribed by the regulations in this chapter;
    (2) With respect to any immediate container when it or its seal (if 
the regulations in this chapter require it to be

[[Page 335]]

sealed) is broken, or when its label or labeling is altered, multilated, 
destroyed, obliterated, or removed in whole or in part, or ceases to 
conform to any labeling requirement prescribed by the regulations in 
this chapter, except that:
    (i) If the drug in such container is repacked or used as an 
ingredient in the manufacture of another drug, and certification of the 
batch thus made is requested, such certificate shall continue to be 
effective for a reasonable time to permit certification or destruction 
of such batch;
    (ii) If the drug is in a container packaged for dispensing and is 
used in compounding a prescription issued by a practitioner licensed by 
law to administer such drug, such certificate shall continue to be 
effective for a reasonable time to permit the delivery of the drug 
compounded on such prescription; or
    (iii) If its label or labeling is removed in whole or in part for 
the purpose of relabeling and supplemental certification of the 
relabeled drug is requested, as provided by Sec. 433.12 of this chapter.
    (3) With respect to any immediate container of penicillin when it is 
included in the packaged combination penicillin with aluminum hydroxide 
gel or penicillin with a vasoconstrictor, or to any immediate container 
of bacitracin when it is included in the packaged combination bacitracin 
with a vasoconstrictor, except that when certification of the batch so 
included is requested, such certificate shall continue to be effective 
for a reasonable time to permit certification of such batch which is 
part of such combination;
    (4) With respect to any package when the drug therein fails to meet 
the standards of identity, strength, quality, and purity which were in 
effect on the date of the certificate; except that those minor changes 
which occur before the expiration date and which are normal and 
unavoidable in good storage and distribution practice shall be 
disregarded.
    (5) With respect to any package of a certifiable antibiotic drug 
subject to the regulations in this chapter, included in a packaged 
combination with another drug, when such other drug fails to meet the 
requirements of the regulations in this chapter; or
    (6) With respect to any immediate container, if such regulations 
require its labeling to bear a caution against dispensing otherwise than 
on prescription, at the beginning of the act of dispensing or offering 
to dispense it otherwise than:
    (i) By a practitioner licensed by law to administer such drug; or
    (ii) On his prescription issued in his professional practice.



Sec. 431.12  Certification of antibiotic drugs after shipment in bulk containers.

    (a) The Food and Drug Administration has received inquiries from 
certain interested manufacturers concerning their shipment of certified 
antibiotics, packaged in bulk containers, to hospitals and pharmacies 
for repacking or for use in the manufacture of another drug on the order 
or prescription of a physician. The regulations promulgated under 
section 507 of the Federal Food, Drug, and Cosmetic Act (21 U.S.C. 357) 
do not prohibit the shipment of certified bulk containers of antibiotics 
to such persons. However, under the provisions of Sec. 431.11(b)(2)(i), 
certification should be requested of each repacked batch and of each 
batch of another drug manufactured from such bulk drug, unless the 
repackaged drug or other drug has been made exempt from the 
certification requirements by regulation. The fact that the drug is to 
be repacked or manufactured on the order or prescription of a physician 
does not exempt it from the certification requirements of the act. Under 
the provisions of Sec. 431.11(b)(2)(ii), it is only when the drug used 
to compound a prescription is in a container packaged for dispensing 
that certification of the drug so compounded is not required.
    (b) In the light of these provisions, unless the manufacturer and 
shipper of bulk containers of antibiotics has, with the consignee, an 
effective permit issued under Sec. 433.16 of this chapter, if the drug 
is to be repacked, or under Sec. 433.13 of this chapter if it is to be 
used in the manufacture of another drug, the shipper has the 
responsibility of seeing that certification is requested of

[[Page 336]]

each repacked batch and of each batch of another drug manufactured from 
such drug.



Sec. 431.17  Request to provide for certification of an antibiotic drug.

    A request under section 507 of the Federal Food, Drug, and Cosmetic 
Act to provide for certification of an antibiotic drug is required to 
comply with the procedures and meet the requirements applicable to the 
submission to the Food and Drug Administration and review by the agency 
of applications and abbreviated applications, and amendments and 
supplements to them, under part 314 of this chapter.
[50 FR 7516, Feb. 22, 1985]



Sec. 431.20  Disposition of outdated drugs.

    When certification becomes invalid because the expiration date is 
passed, such articles should not be disposed of for drug use either 
through commercial or charitable channels unless the articles have been 
assayed to establish potency and recertified.



                  Subpart B--Administrative Procedures



Sec. 431.50  Forms for certification or exemption of antibiotic drugs.

    The following forms which must be supplied in connection with 
certain certification or exemption procedures for antibiotic drugs may 
be obtained from the Product Surveillance Branch (HFD-333), Food and 
Drug Administration, Department of Health and Human Services, 5600 
Fishers Lane, Rockville, MD 20857.

Form
1  Application for exemption for storage.
2  Application for exemption for processing.
3  Application for exemption for labeling.
4  Application for exemption for manufacturing use.
7  Request for check tests and assays or certification of a batch of --
          --------------(the blank to be filled in with the name of the 
          antibiotic drug).
8  Application for exemption for repacking.
9  Request for supplemental certification of a batch of an antibiotic 
          drug.
[39 FR 18934, May 30, 1974, as amended at 40 FR 28052, July 3, 1975; 41 
FR 10886, Mar. 15, 1976; 50 FR 7516, Feb. 22, 1985; 50 FR 8997, Mar. 6, 
1985; 55 FR 11582, Mar. 29, 1990]



Sec. 431.51  Suspension of certification service.

    When the Commissioner finds that a person has:
    (a) Obtained or attempted to obtain a certificate through fraud or 
through misrepresentation or concealment of a material fact; or
    (b) Falsified the records required to be kept by Sec. 431.61; or
    (c) Failed to keep such records or to make them available, or to 
accord full opportunity to take an inventory of stocks on hand, or 
otherwise to check the correctness of such records as required by 
Sec. 431.61; or
    (d) Failed to establish a system for maintaining the records 
required by Sec. 314.81 of this chapter or has repeatedly or 
deliberately failed to maintain such records or to make required reports 
in accordance with the provisions of that section, or has refused to 
permit access to, or copying, or verification of such records or 
reports; or
    (e) Failed to conform to the requirements of good manufacturing 
practice prescribed by parts 210, 211, 225, 226 and 229 of this chapter;

the Commissioner will immediately suspend service to such person under 
the regulations in this chapter. Upon request a hearing will be granted 
to such person to show cause why such service should be resumed.
[39 FR 18934, May 30, 1974, as amended at 40 FR 13497, Mar. 27, 1975; 55 
FR 11582, Mar. 29, 1990]



Sec. 431.52  Hearings.

    Any person who contests the suspension of certification service 
under Sec. 431.51 shall have an opportunity for a regulatory hearing 
before the Food and Drug Administration pursuant to part 16 of this 
chapter.
[41 FR 48267, Nov. 2, 1976, as amended at 42 FR 15675, Mar. 22, 1977]



Sec. 431.53  Fees.

    (a) Fees for the services rendered under the regulations in this 
chapter shall be such as are necessary to provide, equip, and maintain 
an adequate certification service.

[[Page 337]]

    (b) The fee for such services with respect to each batch of a drug, 
certification of which is provided by the regulations in this chapter, 
shall be $114 for each batch submitted, plus the sum of the fees for all 
individual tests required for certification of each batch. The minimum 
tests for each batch shall be those prescribed in the section relating 
specifically to such drug.
    (1) The fee schedule for specific tests required for antibiotic drug 
certification is as follows:

                         Chargeable Fee Per Test                        
                                                                        
                                                                        
                                                                        
Arquad content..................................................     $20
Benzylpenicilloyl content.......................................      32
Bleomycin.......................................................   1,291
Butanol content.................................................      52
Candicidin potency (special turbidimetric)......................      85
Capreomycin 1 content...........................................     121
Color identity..................................................       8
Column chromatography...........................................     130
Column chromatographic isomer content...........................      65
Copper content..................................................      22
Crystallinity...................................................       4
Cycloserine color assay.........................................      27
Daunorubicin potency (special turbidimetric)....................      19
Depressor substance test........................................      40
Disc potency....................................................      52
Dissolution test................................................     107
Doxycycline purity (paper chromatography).......................     130
Free chloride...................................................      54
Frozen antibiotic test panel....................................      32
Gas chromatography..............................................      32
Gentamicin C....................................................     165
Heavy metals test...............................................      14
High pressure liquid chromatography (HPLC)......................      54
Infrared identity...............................................      19
Infrared quantitative...........................................      19
Iodochlorhydroxyquin content....................................      22
Isoniazid content...............................................      22
Karl Fischer moisture...........................................       8
LD50 toxicity...................................................     185
Loss on drying..................................................      12
Lysine content..................................................     161
Melting range...................................................       8
Metal particles (ophthalmic ointments)..........................      22
Microbiological assay, plate....................................      50
Microbiological assay, turbidimetric............................      29
Microorganism count.............................................      68
Nonaqueous titrations (and compleximetric)......................      22
Paper chromatographic identity..................................      43
Penicillenate and penamaldate content...........................      30
Penicillin chemical assay.......................................      15
Penicillin contamination........................................      39
Penicillin G content............................................      32
pH..............................................................       4
Polarographic assay.............................................      33
Potency (special plate).........................................      91
Probenecid content..............................................      32
Procaine colorimetric...........................................       8
Pyrogens test: 3 rabbits........................................      72
Pyrogens test: 8 rabbits........................................     144
Quantitative thin layer chromatography..........................      80
Residual streptomycin...........................................       8
Residue on ignition.............................................      26
Solubility identification.......................................      54
Specific rotation...............................................      22
Specific rotation (potency quantitative)........................      44
Specific surface area...........................................      22
Sterility test..................................................      68
Sulfate content.................................................       8
Tablet disintegration...........................................       5
Thin layer chromatographic identity.............................      43
Total Chlorine..................................................      54
Ultraviolet absorptivity........................................      32
Ultraviolet identity............................................      32
Ultraviolet potency.............................................      32
Vancomycin identity (bioautograph)..............................     117
Zinc titration..................................................      11
                                                                        

    (2) The fee for a supplemental request submitted pursuant to the 
provisions of Sec. 433.12 of this chapter shall be $50.
    (3) [Reserved]
    (4) In the case of persons using the certification services and 
whose manufacturing facilities are not located in the United States or 
the Commonwealth of Puerto Rico, such persons shall be required to 
deposit each year sufficient funds to cover costs encountered when their 
facilities are inspected pursuant to the provisions of section 704 of 
the act.
    (c) When the Commissioner considers it necessary to make 
investigations of a new product containing a certifiable antibiotic drug 
on which a request has been submitted in accordance with Sec. 431.17, 
the fee for such service shall be the cost thereof. In such case the 
request shall be followed by an advance deposit in such amount as the 
Commissioner specifies, and thereafter such additional advance deposits 
shall be made as the Commissioner estimates may be necessary to prevent 
arrears in the payment of such fee.
    (d) A person requiring continuing certification services may 
maintain an advance deposit of the estimated cost of such services for a 
two-month period. Such deposit shall be debited with fees for services 
rendered, but shall not be debited for any fee the amount of which is 
not definitely specified in the regulations in this chapter unless the 
depositor has previously requested the performance of the services to be 
covered by such fee. A monthly statement for each such advance deposit 
shall be rendered.
    (e) The fees for the services rendered with respect to each batch 
certified under the regulations in this chapter shall accompany the 
request for certification, or the request for check tests and assays, 
unless such fee is covered by an advance deposit maintained in 
accordance with paragraph (d) of

[[Page 338]]

this section. Also, if the Commissioner considers that investigations 
other than examination of such samples are necessary to determine 
whether or not such batch complies with the requirements of Sec. 431.10 
for the issuance of a certificate, the fee shall include the cost of 
such investigations.
    (f) The unearned portion of any advance deposit shall be refunded to 
the depositor upon his application.
    (g) Whenever in the judgment of the Commissioner the ratio between 
fees collected (which are based upon experience and the best estimate of 
costs and the best estimate of earnings) and the costs of providing the 
service during an elapsed period of time, in the light of all 
circumstances and contingencies, warrants a refund from the fund 
collected during such period, he shall make ratable refunds to those 
persons to whom the services were rendered and charged, except for those 
services described under Sec. 433.12 of this chapter.
    (h) All deposits and fees required by the regulations in this 
chapter, shall be paid by money order, bank draft, or certified check 
drawn to the order of the Food and Drug Administration, collectible at 
par at Washington, DC. All such deposits and fees shall be forwarded to 
the Food and Drug Administration, Department of Health and Human 
Services, Accounting Branch (HFA-120), 5600 Fishers Lane, Rockville, MD 
20857, whereupon after making appropriate records thereof they will be 
transmitted to the Chief Disbursing Officer, Division of Disbursement, 
Treasurer of the United States, for deposit to the special account 
``Salaries and Expenses, Certification, Inspection, and Other Services, 
Food and Drug Administration.''
[39 FR 18934, May 30, 1974, as amended at 40 FR 13497, Mar. 27, 1975; 40 
FR 28052, July 3, 1975; 41 FR 2384, Jan. 16, 1976; 41 FR 18291, May 3, 
1976; 44 FR 67113, Nov. 23, 1979; 45 FR 16471, Mar. 14, 1980; 46 FR 
16677, Mar. 13, 1981; 46 FR 60578, Dec. 11, 1981; 46 FR 61071, Dec. 15, 
1981; 50 FR 19918, May 13, 1985; 55 FR 11582, Mar. 29, 1990]



                     Subpart C--Records and Reports



Sec. 431.61  Records of distribution.

    (a) The person who requested certification shall keep complete 
records showing each shipment and other delivery (including exports) of 
each certified batch or part thereof by such person or by any person 
subject to his control. Such records shall show the date and quantity of 
each such shipment or delivery and the name and post-office address of 
the person to whom such shipment or delivery was made, and shall be kept 
for not less than 3 years after such date.
    (b) Upon the request of any officer or employee of the Food and Drug 
Administration, or of any other officer or employee of the United States 
acting on behalf of the Secretary, the person to whom a certificate is 
issued shall at all reasonable hours make such records available to any 
such officer or employee and shall accord to him full opportunity to 
make inventory of stocks of such batch on hand and otherwise to check 
the correctness of such records.



Sec. 431.62  Records retention.

    At the option of the person having control of records required to be 
kept by any regulation in this part 431, photostatic or other permanent 
reproductions may be substituted for such records after the first 2 
years of the holding period.



                Subpart D--Confidentiality of Information



Sec. 431.70  Confidentiality of data and information in an investigational new drug notice for an antibiotic drug.

    (a) The existence of an IND notice for an antibiotic drug will not 
be disclosed by the Food and Drug Administration unless it has 
previously been publicly disclosed or acknowledged.
    (b) The availability for public disclosure of all data and 
information in an IND file for an antibiotic drug shall be handled in 
accordance with the provisions established in Sec. 314.430 of this 
chapter.
    (c) Notwithstanding the provisions of Sec. 314.430 of this chapter, 
the Food and Drug Administration shall disclose upon request to an 
individual on whom an investigational antibiotic has been used a copy of 
any adverse reaction report relating to such use.
[39 FR 44655, Dec. 24, 1974, as amended at 50 FR 7517, Feb. 22, 1985]

[[Page 339]]



PART 432--PACKAGING AND LABELING OF ANTIBIOTIC DRUGS--Table of Contents




Sec.
432.1  Packaging requirements.
432.5  Labeling requirements.
432.9  Labeling of antibiotic drugs intended for export.
432.20  Declaration of potency.

    Authority:  21 U.S.C. 321, 331, 352, 353, 357, 371, 381.

    Cross Reference: For other regulations in this chapter concerning 
antibiotic drugs exempted from certain labeling requirements, see also 
Sec. 201.150 of this chapter.



Sec. 432.1  Packaging requirements.

    Each antibiotic drug subject to certification under section 507 or 
512(n) of the act shall be packaged in immediate containers which shall 
be of such composition as not to cause any change in the strength, 
quality, or purity of the contents beyond any limits therefor in 
applicable standards, except that minor changes so caused that are 
normal and unavoidable in good packaging, storage, and distribution 
practice shall be disregarded. The immediate containers shall be tight 
containers as defined by the U.S.P., except that if the antibiotic drug 
is dispensed as an ointment or cream, the immediate containers shall be 
well-closed containers as defined by the U.S.P. If the antibiotic drug 
is packaged for dispensing, it may be packaged in combination with a 
container of a suitable and harmless diluent approved by the 
Commissioner.
    (a) If it is a sterile preparation, the containers shall be sterile 
at the time of filling and closing and shall be so sealed that the 
contents cannot be used without destroying the seal.
    (b) If it is intended for parenteral use and the container is glass, 
it shall be transparent and colorless or light-resistant as defined by 
the U.S.P. The containers are closed either by fusion or by application 
of suitable closures, in such manner as to prevent contamination or loss 
of content. Multiple-dose containers are closed by a substance through 
which a hypodermic needle may be introduced and withdrawn without 
removing the closure or destroying its effectiveness. Each container 
shall be filled with a quantity of a volume in excess of that 
designated, which excess shall be sufficient to permit the withdrawal 
and administration of the labeled quantity or volume, whether 
administered in single or multiple doses.
    (c) If it is dispensed as a tablet, capsule, troche, pellet, or 
suppository, it may be enclosed in a foil or plastic film and such 
enclosure is a tight container as defined by the U.S.P., except for the 
provision that it shall be capable of tight reclosure. The immediate 
container may contain a dessicant separated from the drug by a plug of 
cotton or other like material.
    (d) If it is dispensed as an ointment or cream, it shall be in 
collapsible tubes that shall in no case be larger than the 2-ounce size, 
except:
    (1) If it is labeled for institutional use, it may be packaged in 
immediate containers larger than the 2-ounce size and it may be packaged 
in immediate containers of glass or plastic; or
    (2) If it is an ointment represented for ophthalmic use, it shall be 
in collapsible tubes which shall not be larger than the \1/8\-ounce 
size.
    (e) If it is intended for ophthalmic use, the closure shall be one 
through which a hypodermic needle cannot be introduced.
[39 FR 18938, May 30, 1974, as amended at 42 FR 44225, Sept. 2, 1977; 44 
FR 10377, Feb. 20, 1979]



Sec. 432.5  Labeling requirements.

    (a) If an antibiotic drug is packaged for dispensing:
    (1) It shall be labeled in accordance with the requirements 
prescribed by Sec. 201.100 of this chapter, issued under section 502(f) 
of the act, unless the regulations pertaining to such drug specifically 
exempt it from such requirements.
    (2) Its labeling shall bear any additional information required for 
the drug by specific regulations.
    (3) Each package shall bear on its outside wrapper or container and 
the immediate container an expiration date prescribed for the drug by 
specific regulations; except that in lieu of the expiration date 
prescribed by specific regulations, a date may be used that is 12, 18, 
24, 30, 36, 42, 48, 54, or 60 months after the month during which the

[[Page 340]]

batch was certified if the person who requests certification has 
submitted to the Commissioner results of tests and assays showing that 
such drug as prepared by him is stable for such period of time. If the 
specific regulation does not stipulate an expiration period, it shall be 
as prescribed by this section. If the manufacturer or repacker of the 
drug has been exempted from the certification requirements, such date 
shall be the number of months after the month during which the batch was 
last assayed and released by the manufacturer or repacker. If an 
expiration date is used that is longer than the minimum date provided 
for the drug by specific regulations, it may be used only if the 
manufacturer has submitted information to the Commissioner adequate to 
prove that the drug is stable for such time.
    (b) If it is packaged solely for manufacturing use or for repacking, 
each package shall bear on its outside wrapper or container and the 
immediate container, the following:
    (1) The number of units or micrograms of activity per milligram or 
per gram, and the number of grams or kilograms in the immediate 
container.
    (2) The batch mark.
    (3) The statement ``Caution: Federal law prohibits dispensing 
without prescription.''
    (4) The statement ``For manufacturing use,'' ``For repacking,'' or 
``For manufacturing use or repacking,'' and, if it is not sterile, the 
statement ``nonsterile.''
    (5) The required expiration date.
    (c) The expiration date prescribed for a drug by the regulations in 
this chapter may be omitted from the label of the immediate container if 
such container contains a single dose and it is packaged in an 
individual wrapper or container that bears the date prescribed.
[39 FR 18938, May 30, 1974, as amended at 40 FR 13497, Mar. 27, 1975]



Sec. 432.9  Labeling of antibiotic drugs intended for export.

    (a) Antibiotic drugs subject to certification under section 507 of 
the act and intended for export will be certified notwithstanding 
failure to meet the labeling requirements of the applicable sections if 
the labeling used for such drugs meets the following conditions:
    (1) It has been approved before use by the Government authorities of 
the country to which the drugs are intended for export; and
    (2) Such labeling represents that such drugs are for use only in 
those conditions for which they are certified for domestic distribution.
    (b) The legend ``Caution: Federal law prohibits dispensing without 
prescription'' might be inappropriate on antibiotic drugs exported from 
the United States, since their sale may or may not be so restricted 
under the laws of the country of destination. The Food and Drug 
Administration would not object to a slight modification of the wording 
to read, ``Caution: Federal (U.S.A.) law prohibits dispensing without 
prescription,'' by a manufacturer who wishes to market a drug under the 
same label both in domestic and foreign commerce.
[39 FR 18938, May 30, 1974, as amended at 40 FR 13497, Mar. 27, 1975]



Sec. 432.20  Declaration of potency.

    Wherever the potency of an antibiotic drug included in the 
regulations in this chapter is expressed in terms of weight, such 
potency shall be equivalent to that contained in the same weight of the 
master standard of the drug.
[39 FR 18938, May 30, 1974, as amended at 40 FR 13497, Mar. 27, 1975]



PART 433--EXEMPTIONS FROM ANTIBIOTIC CERTIFICATION AND LABELING REQUIREMENTS--Table of Contents




                      Subpart A--General Provisions

Sec.
433.1  Exemption of antibiotic drugs for human use from batch 
          certification requirements.
433.2  Conditions on the effectiveness of exemptions of antibiotic drugs 
          for human use from batch certification requirements.
433.3  Assay requirements for antibiotic drugs exempted from 
          certification.

[[Page 341]]

  Subpart B--Exemptions for Which an Application or Notice Is Required

433.12  Exemption for labeling.
433.13  Exemption for manufacturing use.
433.14  Exemption for storage.
433.15  Exemption for processing.
433.16  Exemption for repacking.
433.17  Exemption for investigational use.

                   Subpart C--Specific Use Exemptions

433.20  Antibiotic drugs for isolation and differentiation of 
          microorganisms in clinical use.
433.21  Antibiotics for diagnostic use.
433.22  Biologic drugs that contain antibiotics as a preservative.
433.23  Microbiological culture media containing antibiotics.
433.24  Exemption of antibiotic drugs for use in teaching, law 
          enforcement, research and analysis.
433.25  [Reserved]
433.26  Neomycin sulfate ointment intended for hypersensitivity testing.

                     Subpart D--Records and Reports

433.30  Records retention.

    Authority:  21 U.S.C. 352, 355, 357.

    Source:  39 FR 18939, May 30, 1974, unless otherwise noted.



                      Subpart A--General Provisions



Sec. 433.1  Exemption of antibiotic drugs for human use from batch certification requirements.

    (a) Antibiotic drugs for human use are exempt from the batch 
certification requirements of part 431 of this chapter if the conditions 
of paragraph (b) of this section are met; or, in the case of over-the-
counter antibiotic drugs subject to an over-the-counter drug monograph 
in this chapter, if the conditions of paragraph (c) of this section are 
met.
    (b) The conditions are as follows:
    (1) The antibiotic drug is approved for marketing under an 
appropriate antibiotic application or abbreviated antibiotic application 
or is the subject of review under the Drug Efficacy Study Implementation 
Program.
    (2) The antibiotic drug is packaged and labeled for dispensing in 
accordance with the applicable regulation (monograph) in this chapter 
except where other labeling has been approved in an applicable 
antibiotic application or abbreviated antibiotic application.
    (3) The bulk antibiotic drug used in preparing the antibiotic drug 
product meets the standards of identity, strength, quality, and purity 
specified in the applicable regulation (monograph) in this chapter 
except where other standards have been approved in an applicable 
antibiotic application or abbreviated antibiotic application.
    (4) The antibiotic drug product meets the standards of identity, 
strength, quality, and purity specified in the applicable regulation 
(monograph) in this chapter except where other standards have been 
approved in an applicable antibiotic application or abbreviated 
antibiotic application.
    (c) The over-the-counter antibiotic drug product for human use is 
required to meet the general conditions established in Sec. 330.1 of 
this chapter, and the conditions specified in an applicable over-the-
counter drug monograph in this chapter.
    (d) In accordance with the provisions of section 507(e) of the act, 
an antibiotic-containing drug for human use exempt from the requirements 
for batch certification under paragraph (b) of this section is subject 
following its approval to section 505 of the act and applicable 
regulations for new drugs, generally parts 310 through 314 of this 
chapter. For each antibiotic drug subject to an exemption under 
paragraph (b) of this section:
    (1) An approved antibiotic application is regarded to be an approved 
application under Sec. 314.50 of this chapter.
    (2) An approved abbreviated antibiotic application is regarded to be 
an approved abbreviated application under Sec. 314.94 of this chapter.
    (e) Nothing in this section prevents a manufacturer from applying 
for batch certification of an antibiotic drug for human use subject to 
an exemption under this section as provided in section 507(c) of this 
act.
    (f) All exemptions from batch certification requirements for 
antibiotic drugs for human use under this section

[[Page 342]]

are subject to the conditions of effectiveness under Sec. 433.2.

(Approved by the Office of Management and Budget under control number 
0910-0001)

[51 FR 25524, July 15, 1986; 51 FR 30478, Aug. 27, 1986, as amended at 
57 FR 18001, Apr. 28, 1992]



Sec. 433.2  Conditions on the effectiveness of exemptions of antibiotic drugs for human use from batch certification requirements.

    (a) If at any time an exemption from batch certification 
requirements for an antibiotic drug for human use has been granted, the 
Commissioner finds on the basis of new information before the agency 
with respect to such exempted drug, evaluated together with the evidence 
available to the agency when such exemption was granted, that 
certification of each batch is necessary to ensure its safety and 
efficacy of use, the Commissioner shall act immediately to revoke all 
exemptions from batch certification requirements granted for such drug.
    (b) If the Commissioner finds that the person granted an exemption 
from batch certification requirements for an antibiotic drug for human 
use has failed either to comply with the requirements of section 505 of 
the act and the regulations promulgated thereunder or to meet the 
general conditions established in Sec. 330.1 of this chapter and the 
conditions specified in an applicable over-the-counter drug monograph in 
this chapter; or if the Commissioner finds that the requirements of 
Sec. 433.1 have not been met; or if the Commissioner finds that the 
petition for exemption from batch certification contains any false 
statements of fact, the Commissioner may revoke the exemption from batch 
certification requirements immediately and require batch certification 
of the drug until such person shows adequate cause why the exemption 
from batch certification requirements should be reinstated.
    (c) If the Commissioner repeals or suspends an exemption from batch 
certification requirements for an antibiotic drug for human use, a 
notice to that effect and the reasons therefor will be published in the 
Federal Register.
    (d) Any person who contests the revocation or suspension or denial 
of reinstatement of an exemption from batch certification requirements 
for an antibiotic drug for human use shall have an opportunity for a 
regulatory hearing before the Food and Drug Administration under part 16 
of this chapter.
[47 FR 39159, Sept. 7, 1982, as amended at 51 FR 25524, July 15, 1986]



Sec. 433.3  Assay requirements for antibiotic drugs exempted from certification.

    (a) Certain antibiotic drugs are exempted by regulations in this 
chapter from the certification requirements of sections 507 and 512 of 
the act if such drugs comply with standards prescribed by such 
regulations and on condition that the label of each package bears an 
expiration date which is determined from the date during which the batch 
was last assayed and released by the manufacturer.
    (b) It is the position of the Food and Drug Administration that if 
each batch of such exempted drugs is not tested by the manufacturer or 
his agent to determine whether it complies with the standards of 
identity, strength, quality, and purity prescribed for it, the batch is 
not exempt from certification and it may be deemed to be misbranded 
under section 502(l) of the act or be adulterated under section 
501(a)(5) of the act when in interstate commerce.



  Subpart B--Exemptions for Which an Application or Notice Is Required



Sec. 433.12  Exemption for labeling.

    (a) Except as provided by paragraphs (c) and (d) of this section, a 
shipment or other delivery of a certifiable antibiotic drug which is to 
be labeled at an establishment located elsewhere than at the place of 
manufacture shall be exempt, during the time of introduction into and 
movement in interstate commerce and the time of holding in such 
establishment, from the requirement of section 502(l) of the act or the 
certification requirements of section 512(n) of the act if the labeling 
of each shipping container bears the batch mark of the drug, the number 
of units per package

[[Page 343]]

and the expiration date, and if the person who introduced such shipment 
or delivery into interstate commerce holds a permit (Antibiotic Form 3) 
from the Commissioner authorizing shipment for labeling in such 
establishment.
    (b)(1) An application for such a permit shall be in a form specified 
by the Commissioner and shall give the name and location of the 
establishment in which such labeling is to be done.
    (2) In case the applicant is the operator of such establishment, the 
application shall include a written agreement signed by him that he will 
request certification of each batch from which any shipment or delivery 
is made to such establishment unless it is exempt under section 801(d) 
of the act or Sec. 433.17; that he will not remove any of such 
antibiotic drug from such establishment unless it complies with section 
502(l) of the act or the certification requirements of section 512(n) of 
the act or is so exempt, or if certification is refused, unless it is 
returned within a reasonable time to permit reprocessing and 
certification, destruction, or such exemption at the establishment where 
it was manufactured; that he will keep complete records showing the 
date, quantity, and batch mark of each such shipment and delivery and 
the disposition thereof; that he will make such records available to any 
officer or employee of the Food and Drug Administration at any 
reasonable hour within 3 years after the date of such disposition; and 
that he will accord full opportunity to such officer or employee to make 
inventories of stocks on hand and otherwise check the correctness of 
such records.
    (3) In case the applicant is not the operator of such establishment 
such application shall include or be accompanied by:
    (i) A written agreement signed by the applicant that he will request 
certification of each batch from which any shipment or delivery is made 
to such establishment unless it is exempt under section 801(d) of the 
act or Sec. 433.17; that he will keep complete records showing the date, 
quantity, and batch mark of each such shipment and delivery; and that he 
will make such records available to any officer or employee of the Food 
and Drug Administration at any reasonable hour within 3 years after the 
date of such shipment or delivery; and
    (ii) A written agreement signed by the operator of such 
establishment that he will submit a request, supplemental to that of the 
applicant, for the certification of each batch or portion thereof 
comprised in any such shipment or delivery received by him unless it is 
exempt under section 801(d) of the act or Sec. 433.17; that he will 
specify in his request the number of packages of each size in such 
shipment or delivery, the date of delivery, the batch mark thereof, and 
the batch mark he will use therefor; that the batch marks to be used (if 
different from those of the applicant) will be only those of which the 
key is specified in this agreement; that the expiration date used for 
the batch will be only that assigned to the manufacturer by 
certification; that the labeling to be used for such packages will be 
only that of which specimens are attached to this agreement (including 
specimens of all brochures and other printed matter, except readily 
available medical publications, referred to in such labeling); that when 
any change is made in such key or labeling he will promptly submit to 
the Commissioner a full statement of such change or, in the case of 
changed labeling, specimens showing all such changes; that he will not 
remove any of such antibiotic drug from such establishment unless it 
complies with section 502(l) of the act or is exempt under section 
801(d) of the act or Sec. 433.17 or, if certification is refused, unless 
it is returned within a reasonable time to permit reprocessing and 
certification, destruction, or such exemption at the establishment where 
it was manufactured; that he will keep complete records of the 
disposition of each such shipment and delivery; that he will make such 
records available to any officer or employee of the Food and Drug 
Administration at any reasonable hour within 3 years after the date of 
such disposition; and that he will accord full opportunity to such 
officer or employee to make inventories of stocks on hand and otherwise 
check the correctness of such records.

[[Page 344]]

    (4) When the Commissioner finds that such application contains any 
untrue statement of a material fact or that any provision of any such 
agreement has been violated he may revoke such permit.
    (5) Any person who contests the denial or revocation of a permit 
shall have an opportunity for a regulatory hearing before the Food and 
Drug Administration pursuant to part 16 of this chapter.
    (c) An exemption of a shipment or other delivery under paragraph (a) 
of this section, in case the person who introduced such shipment or 
delivery into interstate commerce is the operator of such establishment, 
shall become void at the beginning of the act of removing or offering to 
remove such shipment or delivery or any part thereof, before or after 
labeling, from such establishment unless such batch complies with 
section 502(l) of the act or the certification requirements of section 
512(n) of the act or is exempt under section 801(d) of the act or 
Sec. 433.17 or, if certification is refused, unless such shipment or 
delivery is returned within a reasonable time to permit reprocessing and 
certification, destruction, or such exemption at the establishment where 
it was manufactured.
    (d) An exemption of a shipment or other delivery under paragraph (a) 
of this section, in case the person who inintroduced such shipment or 
delivery into interstate commerce is not the operator of such 
establishment, shall expire at the beginning of the act of removing or 
offering to remove such shipment or delivery of any part thereof, before 
or after labeling from such establishment unless such batch complies 
with section 502(l) of the act or the certification requirements of 
section 512(n) of the act or is exempt under section 801(d) of the act 
or Sec. 433.17 or, if certification is refused, unless such shipment or 
delivery, within a reasonable time, is destroyed or returned to permit 
reprocessing and certification, destruction, or such exemption at the 
establishment where it was manufactured.
[39 FR 18939, May 30, 1974, as amended at 41 FR 48267, Nov. 2, 1976; 42 
FR 15675, Mar. 22, 1977]



Sec. 433.13  Exemption for manufacturing use.

    (a) Except as provided by paragraphs (c) and (d) of this section, a 
shipment or other delivery of any certifiable antibiotic drug subject to 
the regulations in this chapter that is packed in containers of not less 
than 10,000,000 units of penicillin or 10 grams each of one of the other 
antibiotic drugs shall be exempt, during the time of introduction into 
and movement in interstate commerce and the time of holding in the 
establishment where it is so used, from the requirements of section 
502(l) of the act or the certification requirements of section 512(n) of 
the act, if it conforms to the standards prescribed therefor by the 
section of the regulations in this chapter which is specifically 
applicable to such other antibiotic drug, if the label of each container 
bears the batch mark of the drug, the number of units or grams per 
package, and the date on which the latest assay of the drug was 
completed, and if the person who introduced each shipment or delivery 
into interstate commerce holds a permit from the Commissioner 
authorizing shipment for manufacturing use in such establishment.
    (b) An application for such a permit shall be in a form specified by 
the Commissioner, shall give the name and location of the establishment 
in which such drug is to be used and shall be accompanied by;
    (1) A written agreement signed by the applicant that he will keep 
complete records showing the date, quantity, and batch mark of each 
shipment and other delivery of any such drug to such establishment, and 
that he will make such records available to any officer or employee of 
the Food and Drug Administration at any reasonable hour within 3 years 
after the date of such shipment or delivery;
    (2) A written statement signed by the operator of such establishment 
showing that he has adequate facilities for the manufacture of such 
other drug; such statement shall contain an agreement that he will keep 
complete records showing the date of receipt by him and the quantity and 
batch mark of each such shipment and delivery and the disposition 
thereof and showing the quantity and batch mark of each batch

[[Page 345]]

of such other drug manufactured by him and the disposition thereof; that 
he will make such records available to any officer or employee of the 
Food and Drug Administration at any reasonable hour within 3 years after 
the date of such disposition, and that he will accord full opportunity 
to such officer or employee to make inventories of stocks on hand and 
otherwise check the correctness of such records; and
    (3) A written agreement signed by the person who will own the drug 
after its manufacture is completed that he will request certification of 
each batch thereof unless it is exempt under section 801 (d) of the act 
or Sec. 433.12, Sec. 433.14, Sec. 433.16, or Sec. 433.17, and that he 
will not remove any of such drug from such establishment unless it 
complies with section 502(l) of the act or the certification 
requirements of section 512(n) of the act or is so exempt or is returned 
to him for labeling.

When the Commissioner finds that such application contains any untrue 
statement of a material fact or that any provision of any such agreement 
has been violated, he may revoke such permit. Any person who contests 
the denial or revocation of a permit shall have an opportunity for a 
regulatory hearing before the Food and Drug Administration pursuant to 
part 16 of this chapter.
    (c) An exemption of a shipment or other delivery under paragraph (a) 
of this section, in case the person who introduced such shipment or 
delivery into interstate commerce is the operator of such establishment, 
shall become void at the beginning of the act of removing or offering to 
remove such shipment or delivery or any part thereof from such 
establishment, prior to its use in the manufacture of another drug, 
unless it is exempt under section 801(d) of the act.
    (d) An exemption of a shipment or other delivery under paragraph (a) 
of this section, in case the person who introduced such shipment or 
delivery into interstate commerce is not the operator of such 
establishment, shall expire at the beginning of the act of removing or 
offering to remove such shipment or delivery or any part thereof from 
such establishment, prior to its use in the manufacture of another drug, 
unless it is exempt under section 801(d) of the act.
[39 FR 18939, May 30, 1974, as amended at 41 FR 48267, Nov. 2, 1976; 42 
FR 15675, Mar. 22, 1977]



Sec. 433.14  Exemption for storage.

    (a) Except as provided by paragraphs (c) and (d) of this section, a 
shipment or other delivery of a drug which is to be stored at a 
warehouse located elsewhere than at the place of manufacture shall be 
exempt, during the time of introduction into and movement in interstate 
commerce and the time of holding in such warehouse, from the 
requirements of section 502(l) of the act or the certification 
requirements of section 512(n) of the act if the labeling of each 
shipping container bears the batch mark of the drug, and if the person 
who introduced such shipment or delivery into interstate commerce holds 
a permit from the Commissioner authorizing shipment for storage in such 
warehouse.
    (b) An application for such a permit shall be in a form specified by 
the Commissioner, and shall give the name and location of the warehouse 
in which such drug is to be stored. Such application shall be 
accompanied by:
    (1) A written agreement signed by the applicant that he will request 
certification of each batch thereof unless it is exempt under section 
801(d) of the act or Sec. 433.12, Sec. 433.13, or Sec. 433.16, that he 
will not remove any of such drug from such warehouse unless it complies 
with section 502(l) of the act or the certification requirements of 
section 512(n) of the act or is so exempt or, if certification is 
refused unless it is returned within a reasonable time to permit 
reprocessing and certification, destruction, or such exemption at the 
establishment where it was manufactured; that he will keep complete 
records showing the date, quantity, and batch mark of each shipment and 
other delivery of any such drug to such warehouse, and that he will make 
such records available to any officer or employee of the Food and Drug 
Administration at any reasonable hour within 3 years after the date of 
such shipment or delivery; and
    (2) A written statement signed by the operator of such warehouse 
showing

[[Page 346]]

that he has adequate facilities for such storage; such statement shall 
contain an agreement that he will hold each shipment or other delivery 
of such drug intact, under such conditions as will not cause failure of 
the drug to comply with the requirements for certification, that he will 
keep complete records showing the date of receipt by him and the 
quantity and batch mark of each such shipment and delivery and the 
disposition thereof, that he will make such records available to any 
officer or employee of the Food and Drug Administration at any 
reasonable hour within 3 years after the date of such disposition, and 
that he will accord full opportunity to such officer or employee to make 
inventories of stocks on hand and otherwise check the correctness of 
such records.

If the applicant keeps complete records showing the date, quantity, and 
batch mark of each shipment and other delivery of any such drug from 
such warehouse and the name and post-office address of the person to 
whom such shipment or delivery was made, the agreement to keep records 
of such disposals, to make such records available, and to afford 
opportunity for checking their correctness may be included in the 
applicant's agreement and omitted from that of the operator. When the 
Commissioner finds that such application contains any untrue statement 
of a material fact or that any provision of any such agreement has been 
violated he may revoke such permit. Any person who contests the denial 
or revocation of a permit shall have an opportunity for a regulatory 
hearing before the Food and Drug Administration pursuant to part 16 of 
this chapter.
    (c) An exemption of a shipment or other delivery under paragraph (a) 
of this section, in case the person who introduced such shipment or 
delivery into interstate commerce is the operator of such warehouse, 
shall become void at the beginning of the act of removing or offering to 
remove such shipment or delivery or any part thereof from such warehouse 
unless such batch complies with section 502(l) of the act or the 
certification requirements of section 512(n) of the act or is exempt 
under section 801(d) of the act or Sec. 433.12, Sec. 433.13, or 
Sec. 433.16, or, if certification is refused, unless such shipment or 
delivery is returned within a reasonable time to permit reprocessing and 
certification, destruction, or such exemption at the establishment where 
it was manufactured.
    (d) An exemption of a shipment or other delivery under paragraph (a) 
of this section, in case the person who introduced such shipment or 
delivery into interstate commerce is not the operator of such warehouse, 
shall expire at the beginning of the act of removing or offering to 
remove such shipment or delivery or any part thereof from such warehouse 
unless such batch complies with section 502(l) of the act or the 
certification requirements of section 512(n) of the act or is exempt 
under section 801(d) of the act or Sec. 433.12, Sec. 433.13, or 
Sec. 433.16, or, if certification is refused, unless such shipment or 
delivery within a reasonable time, is destroyed, or returned to permit 
reprocessing and certification, destruction, or such exemption at the 
establishment where it was manufactured.
[39 FR 18939, May 30, 1974, as amended at 41 FR 48267, Nov. 2, 1976; 42 
FR 15675, Mar. 22, 1977]



Sec. 433.15  Exemption for processing.

    (a) Except as provided by paragraphs (c) and (d) of this section, a 
shipment or other delivery of any certifiable antibiotic drug subject to 
the regulations in this chapter in concentrated aqueous solution which 
is to be processed at an establishment located elsewhere than at the 
place of manufacture shall be exempt during the time of introduction 
into and movement in interstate commerce and the time of holding in such 
establishment from the requirements of section 502(l) of the act or the 
certification requirements of section 512(n) of the act, if the person 
who introduced such shipment or delivery into interstate commerce holds 
a permit from the Commissioner authorizing shipment for processing in 
such establishment, and each package of such solution bears the batch 
mark of the drug.
    (b) An application for such a permit shall be in a form specified by 
the Commissioner and shall give the name and location of the 
establishment in which

[[Page 347]]

such processing is to be done. Such application shall be accompanied by:
    (1) A written agreement signed by the applicant that he will keep 
complete records showing the date, quantity, potency, and batch mark of 
each shipment and other delivery of any such solution to such 
establishment, and that he will make such records available to any 
officer or employee of the Food and Drug Administration at any 
reasonable hour within 3 years after the date of such shipment or 
delivery;
    (2) A written agreement signed by the operator of such establishment 
showing that he has adequate facilities for such processing; such 
statement shall contain an agreement that he will keep complete records 
showing the date of receipt by him and the quantity and batch mark of 
each such shipment and delivery and the disposition thereof, that he 
will make such records available to any officer or employee of the Food 
and Drug Administration at any reasonable hour within 3 years after the 
date of such disposition, and that he will accord full opportunity to 
such officer or employee to make inventories of stocks on hand and 
otherwise check the correctness of such records; and
    (3) A written agreement signed by the person who will own the drug 
after the processing is completed that he will request certification of 
each batch thereof unless it is exempt under section 801(d) of the act 
or Sec. 433.12, Sec. 433.13, Sec. 433.14, Sec. 433.16, or Sec. 433.17, 
and that he will not remove any of such drug from such establishment 
unless it complies with section 502(l) of the act or the certification 
requirements of section 512(n) of the act or is so exempt.

When the Commissioner finds that such application contains any untrue 
statement of a material fact or that any provision of any such agreement 
has been violated he may revoke such permit. Any person who contests the 
denial or revocation of a permit shall have an opportunity for a 
regulatory hearing before the Food and Drug Administration pursuant to 
part 16 of this chapter.
    (c) An exemption of a shipment or other delivery under paragraph (a) 
of this section, in case the person who introduced such shipment or 
delivery into interstate commerce is the operator of such establishment, 
shall become void at the beginning of the act of removing or offering to 
remove such shipment or delivery or any part thereof, before or after 
processing, from such establishment unless the batch made from such 
shipment or delivery complies with section 502(l) of the certification 
requirements of section 512(n) of the act or is exempt under section 
801(d) of the act or Sec. 433.12, Sec. 433.13, Sec. 433.14, Sec. 433.16, 
or Sec. 433.17, or, if certification is refused, unless such shipment or 
delivery is reprocessed and certified or destroyed within a reasonable 
time.
    (d) An exemption of a shipment or other delivery under paragraph (a) 
of this section, in case the person who introduced such shipment or 
delivery into interstate commerce is not the operator of such 
establishment, shall expire at the beginning of the act of removing or 
offering to remove such shipment or delivery or any part thereof, before 
or after processing, from such establishment unless the batch made from 
such shipment or delivery complies with section 502(l) of the act or is 
exempt under section 801(d) of the act or the certification requirements 
of section 512(n) of the act or Sec. 433.12, Sec. 433.13, Sec. 433.14, 
Sec. 433.16, or Sec. 433.17, or, if certification has been refused, 
unless such shipment or delivery is reprocessed and certified or 
destroyed within a reasonable time.
[39 FR 18939, May 30, 1974, as amended at 41 FR 48267, Nov. 2, 1976; 42 
FR 15675, Mar. 22, 1977]



Sec. 433.16  Exemption for repacking.

    (a) Except as provided by paragraphs (c) and (d) of this section, a 
shipment or other delivery of a drug which is to be repacked at an 
establishment located elsewhere than at the place of manufacture shall 
be exempt, during the time of introduction into and movement in 
interstate commerce and the time of holding such establishment from the 
requirements of section 502(l) of the act or the certification 
requirements of section 512(n) of the act if the labeling of each 
container bears the batch mark of the drug and the number of units per 
package, and if the person

[[Page 348]]

who introduces such shipment or delivery into interstate commerce holds 
a permit from the Commissioner authorizing shipment for repacking in 
such establishment.
    (b) An application for such a permit shall be in a form specified by 
the Commissioner, and shall give the name and location of the 
establishment in which such repacking is to be done. Such application 
shall be accompanied by:
    (1) A written agreement signed by the applicant that he will keep 
complete records showing the date, quantity, and batch mark of each 
shipment and other delivery of any such drug to such establishment, and 
that he will make such records available to any officer or employee of 
the Food and Drug Administration at any reasonable hour within 3 years 
after the date of each shipment or delivery;
    (2) A written statement signed by the operator of such establishment 
showing that he has adequate facilities for such repacking; such 
statement shall contain an agreement that he will keep complete records 
showing the date of receipt by him and the quantity and batch mark of 
each such shipment and delivery and the disposition thereof, that he 
will make such records available to any officer or employee of the Food 
and Drug Administration at any reasonable hour within 3 years after the 
date of such disposition, and that he will accord full opportunity to 
such officer or employee to make inventories of stocks on hand and 
otherwise check the correctness of such records; and
    (3) A written agreement signed by the person who will own the drug 
after the repacking is completed that he will request certification of 
each batch thereof unless it is exempt under section 801(d) of the act 
or Sec. 433.12, Sec. 433.13, Sec. 433.14, or Sec. 433.17, and that he 
will not remove any of such drug from such establishment unless it 
complies with section 502(l) of the act or the certification 
requirements of section 512(n) of the act or is so exempt or is returned 
to him for labeling or, if certification is refused, unless it is 
returned within a reasonable time to permit reprocessing and 
certification, destruction, or such exemption at the establishment where 
it was manufactured.

When the Commissioner finds that such application contains any untrue 
statement of a material fact or that any provision of any such agreement 
has been violated he may revoke such permit. Any person who contests the 
denial or revocation of a permit shall have an opportunity for a 
regulatory hearing before the Food and Drug Administration pursuant to 
part 16 of this chapter.
    (c) An exemption of a shipment or other delivery under paragraph (a) 
of this section, in case the person who introduced such shipment or 
delivery into interstate commerce is the operator of such establishment, 
shall become void at the beginning of the act of removing or offering to 
remove such shipment or delivery or any part thereof, before or after 
repacking, from such establishment unless such batch complies with 
section 502(l) of the act or the certification requirements of section 
512(n) of the act or is exempt under section 801(d) of the act or 
Sec. 433.12, Sec. 433.13, Sec. 433.14, or Sec. 433.17, or is returned to 
such person for labeling or, if certification is refused, unless such 
shipment or delivery is returned within a reasonable time to permit 
reprocessing and certification, destruction, or such exemption at the 
establishment where it was manufactured.
    (d) An exemption of a shipment or other delivery under paragraph (a) 
of this section, in case the person who introduced such shipment or 
delivery into interstate commerce is not the operator of such 
establishment, shall expire at the beginning of the act of removing or 
offering to remove such shipment or delivery or any part thereof, before 
or after repacking, from such establishment unless such batch complies 
with section 502(l) of the act or the certification requirements of 
section 512(n) of the act or is exempt under section 801(d) of the act 
or Sec. 433.12, Sec. 433.13, Sec. 433.14, or Sec. 433.17, or is returned 
to such person for labeling or, if certification is refused, unless such 
shipment or delivery within a reasonable time, is destroyed or returned 
to permit reprocessing and certification, destruction, or such exemption 
at the

[[Page 349]]

establishment where it was manufactured.
[39 FR 18939, May 30, 1974, as amended at 41 FR 48268, Nov. 2, 1976; 42 
FR 15675, Mar. 22, 1977]



Sec. 433.17  Exemption for investigational use.

    A shipment or other delivery of an antibiotic drug shall be exempt 
from section 502(l) of the act or the certification requirements of 
section 512(n) of the act if all the procedures outlined in part 312 or 
Sec. 511.1 of this chapter are complied with. For the purposes of this 
section, the references in part 312 or Sec. 511.1 of this chapter to 
``new drug'' and ``approved new animal drug application'' shall be 
deemed to read ``antibiotic drug'' and ``approval for certification or 
exemption from certification'' respectively.
[39 FR 18939, May 30, 1974, as amended at 40 FR 13497, May 27, 1975; 55 
FR 11582, Mar. 29, 1990]



                   Subpart C--Specific Use Exemptions



Sec. 433.20  Antibiotic drugs for isolation and differentiation of microorganisms in clinical use.

    Antibiotic drugs subject to section 507 of the act shall be exempt 
from section 502(l) if such drugs are:
    (a) Paper discs impregnated with antibiotics in the amounts listed 
in the following table:

                                                                        
------------------------------------------------------------------------
                 Antibiotic                        Content per disc     
------------------------------------------------------------------------
Bacitracin.................................  0.04 unit.                 
Nystatin...................................  100 units.                 
------------------------------------------------------------------------

    (b) Packaged in a container bearing on its label or labeling the 
following:
    (1) On the outside wrapper or container and the immediate container:
    (i) The batch mark.
    (ii) The potency of each disc in the batch.
    (iii) The expiration date as prescribed under Sec. 432.5(a)(3) of 
this chapter.
    (iv) The statement: Not for Susceptibility Testing.
    (2) On the labeling within or attached to the package: Adequate 
directions for use.



Sec. 433.21  Antibiotics for diagnostic use.

    Antibiotics packaged for the withdrawal of individually weighed 
portions and intended for use solely in laboratory procedures in 
connection with the diagnosis or treatment of disease and conspicuously 
so labeled shall be exempt from the certification requirements of 
section 502(l) and 507 of the act and the certification requirements of 
section 512(n) of the act if they comply with all the following 
conditions:
    (a) The potency, moisture content, and identity comply with the 
standards prescribed for the antibiotic by the specific regulations 
issued in this chapter.
    (b) It is packaged in immediate containers that are tight containers 
as defined by the U.S.P. Each such container shall contain not more than 
1 gram.
    (c) Each package bears on the label or labeling of its outside 
wrapper or container and the immediate container the following:
    (1) The statements ``For the withdrawal of individual portions of 
antibiotic. Each portion must be weighed before use. Diagnostic reagent. 
For professional use only.''
    (2) The number of milligrams or grams contained in each immediate 
container and the potency per milligram.
    (3) The batch mark.
    (4) The statement ``Expiration date --------'', the blank being 
filled in with the date that does not exceed the expiration date 
authorized for the antibiotic by this chapter.
    (d) The circular or other labeling within or attached to the package 
bears directions adequate for the use of such drug.

    Cross References: For tests and methods of assay and certification 
of antibiotics susceptibility discs for laboratory diagnosis of disease, 
see Secs. 460.1 and 460.6 of this chapter.



Sec. 433.22  Biologic drugs that contain antibiotics as a preservative.

    Biological drugs that contain any certifiable antibiotic drug 
subject to the regulations in this chapter, and the purpose of the 
antibiotic is for use only as a preservative and the biological drug is 
conspicuously so labeled, shall be exempt from the requirements of 
sections 502(l) and 507 of the act and

[[Page 350]]

the certification requirements of section 512(n) of the act, if such 
drugs are licensed under the Public Health Service Act of July 1, 1944 
(58 Stat. 682; 42 U.S.C. 201 et seq.) or under the Virus-Serum-Toxin Act 
of March 4, 1913 (37 Stat. 832; 21 U.S.C. 151 et seq.).



Sec. 433.23  Microbiological culture media containing antibiotics.

    Microbiological culture media that contain any certifiable 
antibiotic drug subject to the regulations in this chapter shall be 
exempt from the requirements of sections 502(l) and 507 of the act and 
the certification requirements of section 512(n) of the act if:
    (a) They are intended for use in tissue culture and the antibiotic 
drug is added solely for use as an aid in the prevention of microbial 
contamination; or
    (b) They are intended for use in the isolation of selected organisms 
from mixed cultures and the antibiotic drug is added solely for use as 
an aid in such isolation; and
    (c) The certifiable antibiotic drug used in such culture media 
complies with the applicable standards of identity, strength, quality, 
and purity prescribed therefor.



Sec. 433.24  Exemption of antibiotic drugs for use in teaching, law enforcement, research, and analysis.

    Antibiotic drugs subject to section 507 or 512(n) of the act shall 
be exempt from the requirements of section 502(l) and from the 
certification requirements of section 512(n) of the act if shipped or 
sold to, or in the possession of, persons regularly and lawfully engaged 
in instruction in pharmacy, chemistry, or medicine not involving 
clinical use; or in law enforcement; or in research not involving 
clinical use; or in chemical analysis or physical testing, provided they 
are to be used only for such instruction, law enforcement, research, 
analysis, or testing, and provided further that their labels bear the 
statement ``Not for drug use.''



Sec. 433.25  [Reserved]



Sec. 433.26  Neomycin sulfate ointment intended for hypersensitivity testing.

    Neomycin sulfate ointment subject to sections 502(l) and 507 of the 
act and packaged for use as an allergen for skin patch testing of 
hypersensitivity shall be exempt from the certification requirements of 
section 502(l) and 507 of the act if it complies with all the following 
conditions:
    (a) It contains neomycin sulfate equivalent to 200 milligrams of 
neomycin per gram in petrolatum.
    (b) The neomycin sulfate used in preparing the neomycin sulfate 
ointment conforms to the standards prescribed by Sec. 444.42(a)(1) of 
this chapter except Sec. 444.42(a)(1)(ii).
    (c) The shipment of neomycin sulfate is made as a result of a 
specific request made to the manufacturer or distributor by a 
practitioner licensed by law to administer such drug, and the use of 
neomycin sulfate ointment for patch testing is not promoted by the 
manufacturer or distributor.
    (d) Each package shall bear on its outside wrapper or container and 
on the immediate container, in addition to other labeling information 
required by the act and regulations, the following statements in lieu of 
adequate directions for use:
    (1) The statement, ``Caution: Federal law prohibits dispensing 
without prescription''.
    (2) The statement, ``For use only in patch testing''.
    (3) The potency of the ointment.
    (4) The expiration date as prescribed by Sec. 432.5(a)(3) of this 
chapter.
    (e) The quantity shipped is limited to an amount reasonable for the 
purpose of patch testing in the normal course of the practice of 
medicine and is used solely for such patch testing.
    (f) The manufacturer or distributor maintains records of all 
shipments for this purpose for a period of 2 years after shipment and 
will make them available to the Food and Drug Administration upon 
request.
[43 FR 11151, Mar. 17, 1978]



                     Subpart D--Records and Reports



Sec. 433.30  Records retention.

    At the option of the person having control of records required to be 
kept by any regulation in this part 433, photostatic or other permannnt 
reproductions may be substituted for such

[[Page 351]]

records after the first 2 years of the holding period.



PART 436--TESTS AND METHODS OF ASSAY OF ANTIBIOTIC AND ANTIBIOTIC-CONTAINING DRUGS--Table of Contents




Sec.

          Subpart A--Definitions; Interpretations; Requirements

436.1  Sterility requirements of items packaged with sterile antibiotic 
          drugs.
436.2  Alternative assay methods.

                    Subpart B--Sterility Test Methods

436.20  Sterility test methods and procedures.

                   Subpart C--Biological Test Methods

436.31  Equipment and diluents for use in biological testing.
436.32  Pyrogen test.
436.35  Depressor substances test.

                Subpart D--Microbiological Assay Methods

436.100  Laboratory equipment.
436.101  Solutions.
436.102  Culture media.
436.103  Test organisms.
436.104  Penicillin activity.
436.105  Microbiological agar diffusion assay.
436.106  Microbiological turbidimetric assay.

            Subpart E--General Chemical Tests for Antibiotics

436.200  Loss on drying.
436.201  Moisture determination.
436.202  pH.
436.203  Crystallinity.
436.204  Iodometric assay.
436.205  Hydroxylamine colorimetric assay.
436.206  Test for metal particles in ophthalmic ointments.
436.207  Residue on ignition.
436.208  Heavy metals determination.
436.209  Melting range or temperature.
436.210  Specific rotation.
436.211  Identity test by infrared spectrophotometry.
436.212  Disintegration test.
436.213  Nonaqueous titrations.
436.214  Heat stability.
436.215  Dissolution test.
436.216  High-performance liquid chromatographic assay.
436.217  Film-coat rupture test.

           Subpart F--Chemical Tests for Specific Antibiotics

436.300  Polarimetric assay of carbenicillin indanyl sodium.
436.301  Thin layer chromatography identity test for carbenicillin 
          indanyl.
436.302  Clindamycin vapor phase chromatography.
436.303  Clindamycin content of clindamycin palmitate hydrochloride by 
          vapor phase chromatography.
436.304  Clindamycin phosphate vapor phase chromatography.
436.305  Thin layer chromatographic identity test for hetacillin.
436.306  Lincomycin gas liquid chromatography.
436.307  Spectinomycin vapor phase chromatography.
436.308  Paper chromatography identity test for tetracyclines.
436.309  Anhydrotetracyclines and 4-epian-hydrotetracycline.
436.310  Thin layer chromatography identity test for mitomycin.
436.311  Thin layer chromatography identity test for amoxicillin.
436.312  Atomic absorption method for determining the zinc content of 
          zinc bacitracin.
436.316  Determination of penicillin G content.
436.317  Solubility characteristic test for griseofulvin 
          (ultramicrosize) tablets.
436.318  Continuous flow thin layer chromatography identity test.
436.319  Thin layer chromatography identity test for bacitracin and 
          bacitracin zinc.
436.320  Ferric chloride colorimetric assay.
436.321  Griseofulvin gas liquid chromatography.
436.322  High-pressure liquid chromatographic assay for anthracycline 
          antibiotics.
436.323  Continuous flow thin layer chromatography identity test for 
          cefamandole nafate.
436.324  Polarographic analysis of cefamandole.
436.325  High pressure liquid chromatography assay for vidarabine.
436.326  Thin layer chromatographic identity test for cefoxitin sodium.
436.327  Thin layer chromatographic identity test for cyclacillin.
436.328  High pressure liquid chromatographic assay for sulfisoxazole 
          acetyl content.
436.329  High-pressure liquid chromatographic assay for meclocycline.
436.330  Thin layer chromatographic identity test for bacampicillin.
436.331  High-pressure liquid chromatographic assay for dactinomycin.
436.332  High-pressure liquid

[[Page 352]]

          chromatographic assay for moxalactam.
436.333  Thin layer chromatographic identity test for moxalactam.
436.334  High-pressure liquid chromatographic assay for piperacillin.
436.335  High-pressure liquid chromatographic assay for chloramphenicol 
          palmitate.
436.336  Thin layer chromatographic identity test for azlocillin.
436.337  High-pressure liquid chromatographic assay for cephradine.
436.338  High-pressure liquid chromatographic assay for cefoperazone.
436.339  High-pressure liquid chromatographic assay for bleomycin 
          fractions.
436.340  High-pressure liquid chromatographic assay for tetracycline 
          hydrochloride content and 4-epitetracycline hydrochloride 
          content.
436.341  High-pressure liquid chromatographic assay for plicamycin.
436.342  High-pressure liquid chromatographic assay for cefazolin.
436.343  High-pressure liquid chromatographic assay for cefuroxime.
436.344  Thin layer chromatographic identity test for cefuroxime.
436.345  High-pressure liquid chromatographic assay for ceftizoxime.
436.346  High-pressure liquid chromatographic assay for cyclosporine.
436.347  High-pressure liquid chromatographic assay for cefoxitin.
436.348  High-pressure liquid chromatographic assay for ceforanide.
436.349  High-pressure liquid chromatographic assay for L-lysine in 
          ceforanide for injection.
436.350  High-performance liquid chromatographic assay for cefonicid.
436.351  High-performance liquid chromatographic assay for amoxicillin 
          and clavulanic acid.
436.352  High-performance liquid chromatographic assay for determining 
          clavam-2-carboxylate content in clavulanate potassium.
436.353  High-performance liquid chromatographic assay for amdinocillin.
436.354  High-performance liquid chromatographic assay for ceftriaxone.
436.355  High-performance liquid chromatographic assay for ticarcillin-
          clavulanic acid.
436.356  High-performance liquid chromatographic assay for ceftazidime.
436.357  Atomic absorption test for sodium carbonate content.
436.358  High-performance liquid chromatographic assay for pyridine.
436.360  Gel permeation chromatographic assay for high molecular weight 
          polymer.
436.361  High-performance liquid chromatographic assay for aztreonam.
436.362  Thin-layer chromatographic test for free erythromycin content 
          in erythromycin estolate bulk.
436.363  High-performance liquid chromatographic assay for cefmenoxime.
436.364  Atomic absorption test for sodium carbonate content of 
          cefmenoxime hydrochloride for injection.
436.365  Thin layer chromatographic identity test for rifampin.
436.366  High-performance liquid chromatography assay for determining 
          chromatographic purity of vancomycin.
436.367  Thin-layer chromatographic identity test for cephalexin 
          hydrochloride.
436.368  Thin layer chromatographic identity test for cefprozil.
436.369  Thin layer chromatography test for free N-isobutylpiperidone 
          content in rifabutin.
436.370  Spectrophotometric identity test for rifabutin capsules.

     Subpart G--Chemical Tests for Nonantibiotic Active Ingredients

436.400  Thin layer chromatographic identity test for 
          iodochlorhydroxyquin.

          Subpart H--Tests for Specific Antibiotic Dosage Forms

436.500  Penicillin in oil and wax.
436.503  Procaine penicillin and buffered crystalline penicillin for 
          aqueous injection.
436.504  Penicillin-bacitracin ointment.
436.505  Penicillin-streptomycin-bacitracin ointment; penicillin-
          dihydrostreptomycin-bacitracin ointment; penicillin-
          streptomycin-bacitracin methylene disalicylate ointment; 
          penicillin-dihydrostreptomycin-bacitracin methylene 
          disalicylate ointment.
436.506  Benzathine penicillin G and buffered crystalline penicillin for 
          aqueous injection.
436.507  Benzathine-procaine-buffered crystalline penicillins for 
          aqueous injection.
436.508  Penicillin - bacitracin - neomycin ointment; penicillin-
          bacitracin-neomycin in oil.
436.509  Procaine penicillin-streptomycin-polymyxin in oil; procaine 
          penicillin-dihydrostreptomycin-polymyxin in oil; procaine 
          penicillin-streptomycin-polymyxin ointment; procaine 
          penicillin-dihydrostreptomycin-polymyxin ointment.
436.510  Penicillin - streptomycin - erythromycin ointment; penicillin-
          dihydro-streptomycin - erythromycin ointment.
436.511  Penicillin-streptomycin-bacitracin methylene disalicylate-
          neomycin ointment; penicillin-dihydrostreptomycin-bacitracin 
          methylene disalicylate-neomycin ointment.
436.512  Procaine penicillin G-novobiocin-neomycin-dihydrostreptomycin 
          in oil.

[[Page 353]]

436.513  Chlortetracycline troches; tetracycline hydrochloride troches.
436.514  Chlortetracycline hydrochloride powder topical; tetracycline 
          hydrochloride powder topical.
436.515  Capsules tetracycline and oleandomycin phosphate; capsules 
          tetracycline and troleandomycin; capsules tetracycline 
          hydrochloride and oleandomycin phosphate; capsules 
          tetracycline hydrochloride and troleandomycin.
436.516  Tetracycline-neomycin complex powder topical; tetracycline 
          hydrochloride-neomycin sulfate powder topical.
436.517  Bacitracin-neomycin tablets; zinc bacitracin-neomycin tablets; 
          bacitracin methylene disalicylate-neomycin tablets.
436.542  Acid resistance/dissolution test for enteric-coated 
          erythromycin pellets.
436.543  Acid resistance test for pellet-filled doxycycline hyclate 
          capsules.
436.544  Dissolution test for pellet-filled doxycycline hyclate 
          capsules.
436.545  Acid resistance test for erythromycin particles in tablets.

    Authority:  21 U.S.C. 357.

    Source:  39 FR 18944, May 30, 1974, unless otherwise noted.



          Subpart A--Definitions; Interpretations; Requirements



Sec. 436.1  Sterility requirements of items packaged with sterile antibiotic drugs.

    (a) Diluents packaged in combination with sterile antibiotic drugs. 
If a sterile antibiotic drug is packaged in combination with an 
immediate container of a diluent, the immediate container of diluent 
shall be sterile when tested by the method prescribed in 
Sec. 436.20(e)(1).
    (b) Dispensers packaged in combination with sterile antibiotic 
drugs. If a sterile antibiotic drug is packaged in combination with a 
dispenser, such dispenser shall be sterile when tested by the method 
prescribed in Sec. 436.20(e)(1).
[39 FR 18944, May 30, 1974, as amended at 41 FR 46852, Oct. 26, 1976]



Sec. 436.2  Alternative assay methods.

    Alternative assay methods (including automated procedures) employing 
the same basic chemistry or microbiology as the official methods 
described in this part and in the individual monographs of this chapter 
may be used, provided the results obtained are of equivalent accuracy. 
However, only the results obtained from the official methods designated 
in the individual monographs are conclusive.



                    Subpart B--Sterility Test Methods



Sec. 436.20  Sterility test methods and procedures.

    (a) Laboratory facilities. The test must be performed using aseptic 
techniques in an area as free from contamination as is possible to 
achieve. Testing should not be conducted under direct exposure to 
ultraviolet light or in areas under aerosol treatment. Environmental 
tests to assess the suitability of testing conditions should be made 
frequently enough to assure the validity of test results.
    (b) Equipment and reagents--(1) Bacterial membrane filter. The 
filter has a nominal porosity of 0.45 micronplus-minus0.02 
micron, a diameter of approximately 47 millimeters, and a flowrate of 55 
milliliters to 75 milliliters of distilled water passing each square 
centimeter of filter area per minute with a differential pressure of 70 
centimeters of mercury at 25 deg. C.
    (2) Penicillinase solutions. When the amount of penicillinase to be 
used is specified in terms of Levy units, use a penicillinase solution 
standardized in terms of Levy units. One Levy unit of penicillinase 
inactivates 59.3 units of penicillin G in 1 hour at 25 deg. C. and at a 
pH of 7.0 in a phosphate buffered solution of a pure alkali salt of 
penicillin G when the substrate is in sufficient concentration to 
maintain a zero order reaction.
    (c) Culture media. Use ingredients that conform to the standards 
prescribed by the U.S.P. or N.F. In lieu of preparing the media from the 
individual ingredients, they may be made from dehydrated mixtures which, 
when reconstituted with distilled water, have the same or equivalent 
composition as such media and have growth-promoting buffering, and 
oxygen tension-controlling properties equal to or better than such 
media. The pH of each medium should be adjusted with 2N hydrochloric 
acid or sodium hydroxide

[[Page 354]]

before sterilization, so that after sterilization and the addition of 
the penicillinase, if necessary, the pH will fall within the specified 
range. Dispense 90plus-minus10 milliliter quantities of the 
liquid media into individual test tubes (38 millimeters x 200 
millimeters). Close the tubes with suitable closures, and sterilize in 
an autoclave at 121 deg. C. for 20 minutes. The autoclave temperature 
should be reached within 10 minutes. After sterilization, cool the 
medium at once to approximately 25 deg. C. and store at 20 deg. C. to 
30 deg. C. The sterility of each lot of tubes of liquid medium may be 
confirmed by incubating an adequate number of tubes as described in the 
test procedures in paragraph (e) of this section.
    (1) Medium A. Use U.S.P. fluid thioglycolate medium I.
    (2) Medium B. Use U.S.P. fluid thioglycolate medium I, with 
sufficient sterile penicillinase added to inactivate the penicillin 
activity in the sample under test. The penicillinase must be added to 
individual tubes of sterile medium A, using aseptic technique. Prior to 
use, or at the time of the test, a representative number of the tubes 
containing added penicillinase are incubated at 30 deg. -32 deg. C. for 
24 hours to 48 hours, and are examined for sterility. If the sample 
contains penicillin as the only antibiotic, the ability of the 
penicillinase to inactivate all the penicillin in the sample under test 
is checked as follows: Add to one test tube of medium B the proper 
amount of penicillin from one of the individual containers under test. 
Then add 1.0 milliliter of a 1:1.000 dilution of an 18-24 hour culture 
of Staphylococcus aureus (American Type Culture Collection 6538-P) 
1 in medium A. Typical microbial growth must be observable 
after 24 hours incubation at 30 deg.-32 deg. C. If the sample contains a 
mixture of penicillin plus some other antibiotic or antibacterial agent 
the ability of the penicillinase to inactivate all the penicillin in the 
sample is not tested directly on the sample under test, but is 
determined separately, using an amount of penicillin alone equivalent to 
the amount of penicillin in the sample or by any other suitable method 
for standardizing the penicillin-inactivating power of the penicillinase 
preparation.
---------------------------------------------------------------------------

    1 Available from: American Type Culture Collection, 12301 
Parklawn Drive, Rockville, MD 20852.
---------------------------------------------------------------------------

    (3) Medium C. To each liter of medium A add 5.0 milliliters of 
polysorbate 80 before sterilization. To each tube of sterilized medium 
add sufficient sterile penicillinase, and proceed as directed for medium 
B.
    (4) Medium D. To each liter of medium A add 5.0 milliliters of 
polysorbate 80 and sufficient 2N sodium hydroxide so that the pH will be 
7.9plus-minus0.1 after sterilization. Then add sufficient 
sterile penicillinase to each tube and proceed as directed for medium B.
    (5) Medium E. Use U.S.P. XVIII soybean-casein digest medium.
    (6) Medium F. To each liter of medium E add 5.0 milliliters of 
polysorbate 80 before sterilization. To each tube of sterilized medium 
add sufficient sterile penicillinase to solubilize the penicillin in the 
sample to be tested.
    (7) Medium G. Prepare as follows:

Peptic digest of animal tissue...................................6.0 gm.
Pancreatic digest of casein......................................4.0 gm.
Yeast extract....................................................3.0 gm.
Beef extract.....................................................1.5 gm.
Dextrose.........................................................1.0 gm.
Agar............................................................15.0 gm.
Distilled water, q.s.........................................1,000.0 ml.
pH 6.6plus-minus0.1

Suspend the powder in a liter of distilled water. Allow to stand for 5 
minutes, then mix thoroughly. Boil for 1 or 2 minutes or until solution 
is complete. Dispense in suitable flasks and sterilize at 121 deg. C. 
for 15 minutes. Aseptically pour approximately 25-milliliter quantities 
into sterile Petri dish bottoms measuring 20 millimeters x 100 
millimeters. Cover plates with sterile porcelain tops, glazed on the 
outside. Allow plates to stand at room temperature for 48 hours prior to 
use as a control on the sterility of the plates.
    (8) Medium H. Prepare, sterilize, and dispense as described for 
medium G, except as follows:

Dextrose........................................................40.0 gm.
Peptic digest of animal tissue..................................10.0 gm.
Agar............................................................15.0 gm.
Distilled water q.s..........................................1,000.0 ml.
pH 5.6plus-minus0.1 after sterilization


[[Page 355]]


    (9) Medium I. To each liter of Medium A add 1 milliliter of p-tert- 
octylphenoxy polyethoxyethanol.
    (10) Medium J. To each liter of Medium E add 1 milliliter of p-tert- 
octylphenoxy polyethoxyethanol.
    (11) Medium K. (Rinse medium). Prepare as follows:

Peptic digest of animal tissue...................................5.0 gm.
Beef extract.....................................................3.0 gm.
p-tert-octylphenoxy polyethoxyethanol...........................10.0 gm.
Distilled water, q.s.........................................1,000.0 ml.
pH 6.9plus-minus0.2 after sterilization

    (12) Medium L. To each liter of Medium A add 1 milliliter of p-tert- 
octylphenoxy polyethoxyethanol and approximately 10,000 Levy units of 
penicillinase.
    (13) Medium M. To each liter of Medium E add 1 milliliter of p-tert- 
octylphenoxy polyethoxyethanol and approximately 10,000 Levy units of 
penicillinase.
    (14) Medium N:

Pancreatic digest of casein.....................................15.0 gm.
Peptic digest of soybean meal....................................5.0 gm.
Sodium chloride..................................................5.0 gm.
Agar............................................................15.0 gm.
Water........................................................1,000.0 ml.
pH 7.3plus-minus0.2 after sterilization

    (d) Diluting fluids--(1) Diluting fluid A. Dissolve 1 gram of U.S.P. 
peptic digest of animal tissue or equivalent in sufficient distilled 
water to make 1,000 milliliters. Dispense in flasks and sterilize as 
described in paragraph (c) of this section. Final 
pH=7.1plus-minus0.1.
    (2) Diluting fluid B. To each liter of diluting fluid A add 5.0 
milliliters of polysorbate 80 before sterilization.
    (3) Diluting fluid C. To each liter of diluting fluid A add 0.5 gram 
of sodium thioglycollate, and adjust with NaOH so that after 
sterilization the final pH will be pH 6.6plus-minus0.6. 
Dispense in flasks and sterilize as described in paragraph (c) of this 
section.
    (4) Diluting fluid D. To each liter of diluting fluid A add 1 
milliliter of p-tert- octylphenoxy polyethoxyethanol. Dispense in flasks 
and sterilize as described in paragraph (c) of this section. Final 
pH=7.1plus-minus10.1.
    (5) Diluting fluid E. Use isopropyl myristate that is sterile and 
that has a water-extract pH of 5.5 or greater. Determine the water-
extract pH of a portion of the isopropyl myristate as follows: Place 100 
milliliters of the isopropyl myristate sample and 10 milliliters of 
distilled water into a centrifuge bottle of approximately 250 
milliliters capacity and seal the bottle tightly. Place the centrifuge 
bottle on a shaker so that its longest dimension is oriented in the 
direction of shaker movement and shake at 250 cycles per minute for 1 
hour. Centrifuge the bottle at 1,800 revolutions per minute for 20 
minutes. With a suitable vacuum system, remove and discard the upper 
layer; then pipet 5 milliliters of the lower water layer into a beaker 
and determine the pH using a standardized pH meter. If the water-extract 
pH is less than 5.5, pass the isopropyl myristate through a glass column 
packed with basic aluminum oxide, activity grade No. 1. Determine the 
water-extract pH of a portion of the isopropyl myristate that has been 
passed through the aluminum oxide column. Sterilize isopropyl myristate 
by filtration through a 0.22-micron membrane filter and aseptically 
dispense 100-milliliter portions into sterile 250-milliliter flasks.
    (6) Diluting fluid F. To each liter of diluting fluid A add 20 grams 
of disodium edetate, and adjust with NaOH so that after sterilization 
the final pH will be 7.1plus-minus0.1. Dispense in flasks and 
sterilize as described in paragraph (c) of this section.
    (7) Diluting fluid G. To each liter of sterile diluting fluid A add 
10 grams of sterile L-lysine.
    (8) Diluting fluid H. To each liter of diluting fluid A add 10 grams 
of sodium bicarbonate before sterilization.
    (9) Diluting fluid I. To each liter of diluting fluid A add 23.4 
grams of sterile L-arginine base.
    (10) Diluting fluid J. Sterilize 2.0 grams of anhydrous sodium 
carbonate by dry-heating at 180 deg. C for 2 hours. Dissolve in 100 
milliliters of diluting fluid A just prior to use.
    (e) Conduct of test--(1) Bacterial membrane filter method--(i) 
Sample preparation--(a) Antibiotic drug. From each of 20 immediate 
containers, aseptically transfer approximately 300 milligrams of solids 
if it is not a liquid drug, or 1 milliliter by volume if it is a liquid 
drug, or the entire contents if the container contains less than these 
amounts; except that if it is a liquid

[[Page 356]]

drug containing penicillin in a concentration greater than 300,000 units 
per milliliter, use the volume that contains 300,000 units, into a 
sterile 500-milliliter Erlenmeyer flask containing approximately 200 
milliliters of diluting fluid A. (If it is a composite sample packaged 
in one immediate container in accordance with the requirements of 
Sec. 431.5(b) of this chapter, transfer the entire contents, or 
approximately 6 grams, into the Erlenmeyer flask.) Stopper the flask and 
swirl to dissolve the drug. As soon as the sample has completely 
dissolved, proceed as directed in paragraph (e)(1)(ii) of this section. 
If the pooled portions from 20 containers will not dissolve completely 
in 200 milliliters of diluting fluid or will not filter rapidly, 400 
milliliters of diluting fluid may be used or two separate tests may be 
performed using a pool of 10 containers for each test.
    (b) Diluent packaged in combination with a sterile drug. Using the 
entire contents from each of 20 immediate containers, proceed as 
directed in paragraph (e)(1)(ii) of this section.
    (c) Sterile dispensers packaged in combination with a sterile drug. 
Prepare 20 clean, empty containers of approximately the same size as 
those in which the sterile antibiotic drug is packaged. To each 
container add diluting fluid A in a volume approximately the same as 
that of the sterile drug when it is prepared for dispensing. Cap the 
containers, sterilize by autoclaving at 121 deg. C. for 20 minutes, and 
then allow to cool to room temperature. Aseptically open each dispenser 
package and remove each dispenser in turn. Use each aseptically to 
remove 1 milliliter of the fluid from a separate sterile container 
prepared as described above. Aseptically transfer the fluid to a 500-
milliliter Erlenmeyer flask containing approximately 200 milliliters of 
diluting fluid A. Stopper the flask and proceed as directed in paragraph 
(e)(1)(ii) of this section.
    (ii) Test procedure. Aseptically filter the solution through a 
bacteriological membrane filter. All air entering the filtering system 
is filtered through air filters capable of removing microorganisms. 
Filter three 100-milliliter quantities of diluting fluid A through the 
membrane. For the penicillin and cephalosporin classes of antibiotics, 
add sufficient penicillinase to diluting fluid A to inactivate the 
residual antibiotic activity on the membrane after filtration. By means 
of a sterile circular blade, paper punch, or any other suitable sterile 
device, cut a circular portion (approximately 17.5 millimeters in 
diameter) from the center of the filtering area. Transfer the cut center 
area to a sterile 38 by 200 millimeter (outside dimensions) test tube 
containing 90plus-minus10 milliliters of sterile medium A. 
Incubate the tube for 7 days at 30 deg.-32 deg. C. Using sterile 
forceps, transfer the remaining outer portion of the membrane into a 
second similar tube containing 90plus-minus10 milliliters of 
medium E. Incubate the second tube for 7 days at 22 deg.-25 deg. C.
    (2) Direct method. From each of 20 immediate containers, transfer 
approximately 300 milligrams of solids if it is not a liquid drug, or 1 
milliliter by volume if it is a liquid drug, or the entire contents if 
it contains less than these amounts, except if it is a liquid drug 
containing penicillin in a concentration greater than 300,000 units per 
milliliter use that volume that contains 300,000 units, into individual 
sterile test tubes (38 millimeters  x  200 millimeters) containing 
90plus-minus10 milliliters of medium A. Incubate all tubes at 
30 deg. C. to 32 deg. C. for 7 days. Gently agitate the tubes every 1 to 
3 days or until complete solubilization occurs. At intervals, examine 
all tubes for visible growth. If growth is observed in any tube, confirm 
by microscopic examination. From each of the same 20 immediate 
containers, transfer a second portion (equivalent to that portion 
initially transferred to the tubes containing medium A) to individual 
sterile test tubes (38 millimeters  x  200 millimeters) containing 
90plus-minus10 milliliters of medium E, except when each 
container does not have sufficient material to provide for the two 
similar-size portions, obtain the second portion from 20 additional 
immediate containers. Incubate all tubes at 22 deg. C. to 25 deg. C. for 
7 days. Gently agitate the tubes every 1 to 3 days or until complete 
solubilization occurs. At intervals, examine all tubes for visible 
growth. If growth is observed in any tube, confirm by microscopic 
examination.

[[Page 357]]

    (3) Bacterial membrane filter method for ophthalmic ointments--(i) 
Ointments that do not contain penicillin. From each of 10 immediate 
containers aseptically transfer 0.1 gram of the product into a sterile 
250-milliliter flask containing 100 milliliters of diluting fluid E 
which has previously been heated to a temperature of 47 deg. C. Repeat 
the process, using 10 additional containers. Swirl both of the flasks to 
dissolve the ointment. Immediately aseptically filter each solution 
through a separate bacteriological membrane filter previously moistened 
with approximately 0.2 milliliter of medium K. Filter all air entering 
the system through air filters capable of removing microorganisms. 
Remove any residual antibiotic from the membranes by rinsing each filter 
five times with 100 milliliters of medium K. The membranes should be 
covered with fluid throughout each step of the filtration procedure 
until the end of the last filtering step. By means of a sterile circular 
blade, paper punch, or other suitable sterile device, cut a circular 
portion (approximately 17.5 millimeters in diameter) from the center of 
the filtering area of each membrane. Transfer the center portion of the 
filtering area of each filter to a sterile test tube 38 millimeters  x  
200 millimeters (outside dimensions) containing 90 
millilitersplus-minus10 milliliters of sterile medium I. 
Incubate the tube for 7 days at 30 deg. C. to 32 deg. C. Using sterile 
forceps transfer the outer portion of each filter to a similar test tube 
containing 90 milliliters plus-minus10 milliliters of sterile 
medium J. Incubate this tube for 7 days at 22 deg. C. to 25 deg. C.
    (ii) Ointments containing penicillin. Proceed as directed in 
paragraph (e)(3)(i) of this section, except in lieu of sterile medium I 
use sterile medium L for the center portion of the filtering area of 
each filter and in lieu of sterile medium J use sterile medium M for the 
remaining outer portion of each filter.
    (f) Evaluation of results--(1) Bacterial membrane-filter method. The 
batch, or the part of the batch represented by a particular filling 
operation meets the requirements of the test if no sample tube shows 
growth. If growth is observed in any sample tube, run a second test in 
the appropriate medium, except perform it in duplicate, using 40 
immediate containers. If in the original test, growth is observed in 
only one of the two media, test both portions of the cut filter membrane 
by placing each into a separate tube of the same medium. The batch meets 
the requirements if no tube on the second test shows growth. If growth 
is observed in any of the control tubes as well as in the sample tubes 
in either the original or the second test such test is invalid and must 
be performed again. In any event, further tests may be justified if 
there is sufficient reason to believe that the results obtained in the 
first and second tests may not be valid. In such instances, the batch is 
satisfactory if on the final test no tube shows growth.
    (2) Direct method. The batch, or the part of the batch represented 
by a particular filling operation, meets the requirements of the test if 
no tube shows growth after incubation. If growth is observed in any 
sample tube, run a second test in the appropriate medium using 40 
immediate containers. The batch is satisfactory if, on the second test, 
no tube shows growth. If growth is observed in any of the control tubes 
(except inoculated tubes, if the sample is penicillin) as well as in the 
sample tubes in either the original or the second test, such test is 
invalid and must be performed again. In any event, further tests may be 
justified if there is sufficient reason to believe that the results 
obtained on the first and second tests may not be valid. In such 
instances the batch is satisfactory if in the final test no tube shows 
growth.
[39 FR 18944, May 30, 1974, as amended at 41 FR 46852, Oct. 26, 1976; 42 
FR 27228, May 27, 1977; 43 FR 43457, Sept. 26, 1978; 49 FR 25846, June 
25, 1984; 49 FR 40006, Oct. 12, 1984; 50 FR 48397, Nov. 25, 1985; 52 FR 
4611, Feb. 13, 1987; 53 FR 13401, Apr. 25, 1988]



                   Subpart C--Biological Test Methods



Sec. 436.31  Equipment and diluents for use in biological testing.

    (a) Equipment--(1) Temperature-measuring devices. Use an accurate 
clinical

[[Page 358]]

thermometer or any other temperature-measuring device of equal 
sensitivity that has been tested to determine the time necessary to 
reach the maximum reading.
    (2) Pyrogen-free glassware. Render all glassware free from pyrogens 
by heating at 250 deg. C. for not less than 30 minutes or by any other 
suitable method.
    (3) Pyrogen-free syringes and needles. Render all syringes and 
needles free from pyrogens by heating at 250 deg. C. for not less than 
30 minutes or by any other suitable method.
    (4) Pyrogen-free sodium chloride. Heat sodium chloride for not less 
than 2 hours at 200 deg. C.
    (5) Pyrogen-free sodium carbonate. Heat anhydrous sodium carbonate 
for not less than 4 hours at 170 deg. C.
    (b) Diluents. (1) Diluent 1 (pyrogen-free water): Prepare pyrogen-
free water by collecting freshly distilled water and sterilizing it in 
an autoclave at 121 deg. C. for not less than 20 minutes. Pyrogen-free 
water meets the requirements for the absence of pyrogens as described in 
Sec. 436.32(a)(3) when 10 milliliters per kilogram are administered as 
described in Sec. 436.32(a)(2). In testing water for the absence of 
pyrogens, the aliquot to be tested is made isotonic by the addition of 
pyrogen-free sodium chloride.
    (2) Diluent 2 (pyrogen-free saline solution): Prepare an isotonic 
solution of sodium chloride by dissolving 9.0 grams of pyrogen-free 
sodium chloride (prepared as described in Sec. 436.31(a)(4) ) in 
pyrogen-free, distilled water (diluent 1) to make 1,000 milliliters. 
Sterilize in an autoclave at 121 deg. C. for not less than 20 minutes. 
Pyrogen-free saline solution meets the requirements for the absence of 
pyrogens as described in Sec. 436.32(a)(3) when 10 milliliters per 
kilogram are administered as described in Sec. 436.32(a)(2).
    (3) Diluent 3 (sterile distilled water): Prepare freshly distilled 
water. Sterilize in an autoclave at 121 deg. C. for 20 minutes.
    (4) Diluent 4 (sterile saline solution): Dissolve 9.0 grams of 
sodium chloride in distilled water to make 1,000 milliliters. Sterilize 
in an autoclave at 121 deg. C. for 20 minutes.
    (5) Diluent 5 (10 percent gum acacia): Dissolve 10 grams of gum 
acacia in approximately 50 milliliters of distilled water. Allow to 
stand overnight at room temperature and dilute to 100 milliliters with 
distilled water. Filter through cotton. Store under refrigeration.
    (6) Diluent 6 (0.5 percent gum acacia in distilled water). 11132
    (7) Diluent 7 (1.0N hydrochloric acid).
    (8) Diluent 8 (0.1N hydrochloric acid).
    (9) Diluent 9 (0.05N sodium hydroxide).
    (10) Diluent 10 (1 percent U.S.P. methylcellulose (4,000 
centipoises) solution): Dissolve 1 gram of U.S.P. methylcellulose (4,000 
centipoises) in 100 milliliters of distilled water. Allow to stand 
overnight at room temperature or until solution is complete. Store under 
refrigeration.
    (11) Diluent 11 (0.12N sodium hydroxide).
    (12) Diluent 12 (0.5 percent methylcellulose (4,000 centipoises) in 
distilled water). Proceed as directed in paragraph (b)(10) of this 
section, except use 0.5 gram of methylcellulose (4,000 centipoises).
    (13) Diluent 13 (pyrogen-free sodium carbonate solution). Dissolve 
25.6 grams of anhydrous pyrogen-free sodium carbonate (prepared as 
described in paragraph (a)(5) of this section) in 1,000 milliliters 
pyrogen-free, distilled water (diluent 1). Pyrogen-free, sodium 
carbonate solution meets the requirements for the absence of pyrogens as 
described in Sec. 436.32(a)(3) when 1.0 milliliter per kilogram is 
administered as described in Sec. 436.32(a)(2).
    (14) Diluent 14 (0.07M sterile sodium carbonate solution). Dissolve 
7.3 grams of sodium carbonate in distilled water to make 1,000 
milliliters. Sterilize in an autoclave at 121 deg. C. for 20 minutes.
    (15) Diluent 15 (pyrogen-free sodium carbonate solution): Dissolve 
9.9 grams of anhydrous pyrogen-free sodium carbonate (prepared as 
directed in paragraph (a)(5) of this section) in 1,000 milliliters of 
pyrogen-free, distilled water (diluent 1). Pyrogen-free sodium carbonate 
solution meets the requirements for the absence of pyrogens as described 
in Sec. 436.32(a)(3) when 1.0 milliliter per kilogram is administered as 
described in Sec. 436.32(a)(2).

[[Page 359]]

    (16) Diluent 16 (0.13M sterile pyrogen-free sodium carbonate 
solution). Dissolve 14.0 grams of anhydrous pyrogen-free sodium 
carbonate (prepared as described in paragraph (a)(5) of this section) in 
1,000 milliliters pyrogen-free, distilled water. Sterilize in an 
autoclave at 121  deg.C for 20 minutes.
[39 FR 18944, May 30, 1974, as amended at 40 FR 51625, Nov. 6, 1975; 50 
FR 48397, Nov. 25, 1985; 53 FR 13401, Apr. 25, 1988]



Sec. 436.32  Pyrogen test.

    (a) Method 1--(1) Test animal. Use healthy, mature rabbits weighing 
not less than 1,800 grams each that have maintained their weight on an 
antibiotic-free diet for at least 1 week under the environmental 
conditions specified in this section. House the animals individually in 
an area of uniform temperature (plus-minus3 deg. C.) and free 
from disturbances likely to excite them. Do not use animals for pyrogen 
tests more frequently than once every 48 hours or prior to 2 weeks 
following their having been given a test sample that was adjudged 
pyrogenic. Before using an animal that has not been used for a test 
during the previous 2 weeks, condition it 1 to 3 days prior to pyrogen 
testing by conducting a sham test as directed in paragraph (a)(2) of 
this section, omitting the injection.
    (2) Procedure. Using equipment and diluents described in 
Sec. 436.31, as necessary, perform the test in an area where the animals 
are housed or under similar environmental conditions. On the day of the 
test: Withhold all food from the animals being used until after 
completion of the test, except that access to water may be allowed; and 
determine the ``control temperature'' of each animal by inserting the 
temperature-measuring device into the rectum of the test animal to a 
depth of not less than 7.5 centimeters and allowing sufficient time to 
reach a maximum temperature, as previously determined, before taking the 
reading. In any one test use only those animals whose control 
temperatures do not deviate by more than 1 deg. C. from each other and 
do not use any animal with a temperature exceeding 39.8 deg. C. The 
control temperature recorded for each rabbit constitutes the temperature 
from which any subsequent rise following the injection of the material 
is calculated. If the product is packaged for dispensing and is in a 
combination package with a container of diluent, dilute the product as 
directed in the labeling. Warm the product to be tested to approximately 
37 deg. C. Dilute the sample with sterile, pyrogen-free saline (prepared 
as described in Sec. 436.31(b)(2)) to the appropriate concentration 
specified in the individual section for each antibiotic to be tested. 
Inject a test dose of 1 milliliter of the diluted sample per kilogram of 
rabbit weight into an ear vein of each of three rabbits within 30 
minutes subsequent to the control temperature reading. Record the 
temperature at 1, 2, and 3 hours subsequent to the injection.
    (3) Evaluation. If no rabbit shows an individual rise in temperature 
of 0.6 deg. C. or more above its respective control temperature, and if 
the sum of the three temperature rises does not exceed 1.4 deg. C., the 
sample meets the requirements for the absence of pyrogens. If one or two 
rabbits show a temperature rise of 0.6 deg. C. or more, or if the sum of 
the temperature rises exceeds 1.4 deg. C., repeat the test using five 
other rabbits. If not more than three of the eight rabbits show 
individual rises in temperature of 0.6 deg. C. or more, and if the sum 
of the eight temperature rises does not exceed 3.7 deg. C., the sample 
meets the requirements for the absence of pyrogens.
    (b) Method 2. Proceed as directed in paragraph (a) of this section, 
except dilute the sample with pyrogen-free water (diluent 1).
    (c) Method 3. Proceed as directed in paragraph (a) of this section, 
except dilute the sample with pyrogen-free water (diluent 1) and inject 
a test dose of 2.0 milliliters of the diluted sample per kilogram of 
rabbit weight.
    (d) Method 4. Proceed as directed in paragraph (a) of this section, 
except inject a test dose of 0.5 milliliter of the diluted sample per 
kilogram of rabbit weight.
    (e) Method 5. Proceed as directed in paragraph (a) of this section, 
except dilute the sample with pyrogen-free water (diluent 1) and inject 
a test dose of 0.5 milliliter of the diluted sample per kilogram of 
rabbit weight.

[[Page 360]]

    (f) Method 6. Proceed as directed in paragraph (a) of this section, 
except dilute sample with 0.05N sodium hydroxide (diluent 9).
    (g) Method 7. Proceed as directed in paragraph (a) of this section, 
except dilute sample with sodium carbonate solution (diluent 13).
    (h) Method 8. Proceed as directed in paragraph (a) of this section, 
except inject a test dose of 2.0 milliliters of the diluted sample per 
kilogram of rabbit weight.
    (i) Method 9. Proceed as directed in paragraph (a) of this section, 
except dilute sample with pyrogen-free sodium carbonate solution 
(diluent 15).
    (j) Method 10. Proceed as directed in paragraph (a) of this section, 
except dilute the sample with sodium carbonate solution (diluent 16).
[39 FR 18944, May 30, 1974, as amended at 40 FR 51625, Nov. 6, 1975; 45 
FR 22921, Apr. 4, 1980; 50 FR 48397, Nov. 25, 1985; 53 FR 13401, Apr. 
25, 1988]



Sec. 436.35  Depressor substances test.

    Proceed as directed in the USP XX depressor substances test. Prepare 
the sample test solution as follows: For each antibiotic listed in the 
table below, select the appropriate diluent and test dose (concentration 
and volume). If the product is packaged for dispensing and is in a 
combination package with a container of diluent, dilute the product as 
directed in the labeling.

                                                                        
------------------------------------------------------------------------
                                                              Volume of 
                                             Concentration      test    
          Antibiotic            Diluent \1\     of test      solution to
                                              solution \2\   be injected
                                                                 \3\    
------------------------------------------------------------------------
Bleomycin sulfate.............          4      \4\ 0.5             1.0  
Capreomycin sulfate...........          4          3.0             1.0  
Chlortetracycline                                                       
 hydrochloride................          3          5.0              .6  
Clindamycin phosphate.........          4          5.0             1.0  
Daunorubicin hydrochloride....          4          1.5             1.0  
Dihydrostreptomycin sulfate...          4          3.0             1.0  
Doxorubicin hydrochloride.....          4          1.5             1.0  
Doxycycline hyclate...........          4          5.0             1.0  
Lincomycin hydrochloride                                                
 monohydrate..................          4          3.0             1.0  
Minocycline hydrochloride.....          4          5.0              .6  
Plicamycin....................          3          0.050           1.0  
Mitomycin.....................          4          0.050           1.0  
Oxytetracycline \5\...........          3          5.0              .6  
Oxytetracycline hydrochloride.          4          5.0              .6  
Rolitetracycline..............          4          5.0              .6  
Rolitetracycline nitrate......          4          5.0              .6  
Sodium colistimethate.........          4          3.0             1.6  
Spectinomycin hydrochloride...          4         15.0             1.0  
Streptomycin sulfate..........          4          3.0             1.0  
Tetracycline hydrochloride....          4          5.0              .6  
Tetracycline phosphate \5\....          3          5.0              .6  
Vidarabine monohydrate \5\....          4          1.0             1.0  
------------------------------------------------------------------------
\1\ Diluent number as listed in sec. 436.31(b).                         
\2\ Milligrams of activity per milliliter.                              
\3\ Milliliters per kilogram of body weight.                            
\4\ The concentration of the test solution is expressed in units per    
  milliliter in lieu of milligrams of activity per milliliter.          
\5\ To prepare the test solution, proceed as directed in the individual 
  section of the antibiotic drug regulation in this chapter for the     
  antibiotic to be tested.                                              

[46 FR 60568, Dec. 11, 1981, as amended at 46 FR 61071, Dec. 15, 1981; 
49 FR 5096, Feb. 10, 1984]



                Subpart D--Microbiological Assay Methods



Sec. 436.100  Laboratory equipment.

    Equipment should be selected which is adequate for its intended use 
and should be thoroughly cleansed after each use to remove any 
antibiotic residues. The equipment should be kept covered when not in 
use. Clean glassware intended for holding and transferring the test 
organisms should be sterilized in a hot air oven at 200-220 deg. C. for 
2 hours. Volumetric flasks, pipettes, or accurately calibrated diluting 
devices should be used when diluting standard and sample solutions.
    (a) Microbiological agar diffusion assay--(1) Cylinders. Use 
stainless steel cylinders with an outside diameter of 8 millimeters 
(plus-minus0.1 millimeter), an inside diameter of 6 
millimeters (plus-minus0.1 millimeter), and a length of 10 
millimeters (plus-minus0.1 millimeter).
    (2) Plates. Plastic or glass Petri dishes may be used, having 
dimensions of 20 by 100 millimeters. Covers should be of suitable 
material.
    (b) Microbiological turbidimetric assay--(1) Tubes. Tubes which give 
satisfactory results and have uniform length and diameter should be 
used. If reusable tubes are employed, care must be taken to remove not 
only all antibiotic residues from the previous test but also all traces 
of cleaning solution.
    (2) Colorimeter. Use a suitable photoelectric colorimeter at a 
wavelength of 530 millimicrons. Set the instrument at zero absorbance 
with clear,

[[Page 361]]

uninoculated broth prepared as described in the applicable method for 
the antibiotic being assayed.
[39 FR 18944, May 30, 1974, as amended at 41 FR 34743, Aug. 17, 1976]



Sec. 436.101  Solutions.

    (a) Antibiotic assay solutions are prepared as follows (solution 
numbers 1, 2, 3, 4, and 6 correspond to those used in ``Assay Methods of 
Antibiotics,'' D. C. Grove and W. A. Randall, Medical Encyclopedia, 
Inc., New York, N.Y. (1955), p. 222), which is incorporated by 
reference. Copies are available from the Medical Encyclopedia Inc., 30 
East 60th St., New York, NY 11220, or available for inspection at the 
Office of the Federal Register, 800 North Capitol Street, NW., suite 
700, Washington, DC.
    (1) Solution 1 (1 percent potassium phosphate buffer, pH 6.0).

Dibasic potassium phosphate: 2.0 gm.
Monobasic potassium phosphate: 8.0 gm.
Distilled water, q.s: 1,000.0 ml.

    Adjust with 18N phosphoric acid or 10N potassium hydroxide to yield 
a pH 5.95 to 6.05 after sterilization.
    (2) Solution 2 (citrate buffer solution pH 6.3).

Citric acid: 13.2 gm.
Sodium hydroxide: 7.06 gm.
Sodium citrate: 97.0 gm.
Distilled water, q.s: 1,000.0 ml.

    Adjust with 10 percent citric acid solution or 10N sodium hydroxide 
to yield pH 6.2 to 6.4 after sterilization.
    (3) Solution 3 (0.1M potassium phosphate buffer, pH 8.0).

Dibasic potassium phosphate: 16.73 gm.
Monobasic potassium phosphate: 0.523 gm.
Distilled water, q.s: 1,000.0 ml.

    Adjust with 18N phosphoric acid or 10N potassium hydroxide to yield 
a pH 7.9 to 8.1 after sterilization.
    (4) Solution 4(0.1M potassium phosphate buffer, pH 4.5).

Monobasic potassium phosphate: 13.6 gm.
Distilled water, q.s: 1,000.0 ml.

    Adjust with 18N phosphoric acid or 10N potassium hydroxide to yield 
a pH 4.45 to 4.55 after sterilization.
    (5) [Reserved]
    (6) Solution 6 (10 percent potassium phosphate buffer, pH 6.0).

Dibasic potassium phosphate: 20.0 gm.
Monobasic potassium phosphate: 80.0 gm.
Distilled water, q.s: 1,000.0 ml.

    Adjust with 18N phosphoric acid or 10N potassium hydroxide to yield 
a pH 5.95 to 6.05 after sterilization.
    (7)-(9) [Reserved]
    (10) Solution 10 (0.2M potassium phosphate buffer, pH 10.5).

Dibasic potassium phosphate: 35.0 gm.
10 N potassium hydroxide: 2.0 ml.
Distilled water, q.s: 1,000.0 ml.

    Adjust with 18N phosphoric acid or 10N potassium hydroxide to yield 
a pH 10.4 to 10.6 after sterilization.
    (11) Solution 11 (10 percent potassium phosphate buffer, pH 2.5).

Monobasic potassium phosphate: 100.0 gm.
Concentrated hydrochloric acid: 0.2 ml. (approximately).
Distilled water, q.s: 1,000.0 ml.

    Adjust with 18N phosphoric acid or 10N potassium hydroxide to yield 
a pH 2.0 to 2.8 after sterilization.
    (12) Solution 12 (10 percent potassium phosphate buffer, pH 7.0).

Monobasic potassium phosphate: 100.0 gm.
Distilled water, q.s: 1,000.0 ml.

    Adjust with 18N phosphoric acid or 10N potassium hydroxide to yield 
a pH 6.95 to 7.05 after sterilization.
    (13) Solution 13 (0.01N methanolic hydrochloric acid).

1.0N hydrochloric acid: 10.0 ml.
Methyl alcohol, q.s: 1,000.0 ml.

    (14) Solution 14 (2 percent sodium bicarbonate solution).

Sodium bicarbonate: 20.0 gm.
Distilled water, q.s: 1,000.0 ml.

    Prepare daily.
    (15) Solution 15 (80 percent isopropyl alcohol solution).

Isopropyl alcohol: 800.0 ml.
Distilled water, q.s: 1,000.0 ml.

    (16) Solution 16 (0.1 M potassium phosphate buffer, pH 7.0).

Dibasic potassium phosphate: 13.6 gm.
Monobasic potassium phosphate: 4.0 gm.
Distilled water, q.s.: 1,000.0 ml.

    Adjust with 18 N phosphoric acid or 10 N potassium hydroxide to 
yield a pH 6.8 to 7.2 after sterilization.
    (17) Solution 17 (5 percent methyl alcohol in 1 percent potassium 
phosphate buffer, pH 6.0).


[[Page 362]]


Methyl alcohol: 50.0 ml.
1 percent potassium phosphate buffer, pH 6.0, q.s.: 1,000.0 ml.

    (18) Solution 18 (0.054M sodium phosphate buffer, pH 6.9).

    Sodium dihydrogen phosphate monohydrate: 3.97 gm.
    Disodium hydrogen phosphate anhydrous: 3.55 gm.
    Distilled water, q.s.: 1,000.0 mL.
[39 FR 18944, May 30, 1974, as amended at 40 FR 52004, Nov. 7, 1975; 45 
FR 75194, Nov. 14, 1980; 47 FR 9396, Mar. 5, 1982]



Sec. 436.102  Culture media.

    (a) Ingredients. Use ingredients that conform to the standards, if 
any, prescribed by the U.S.P. or N.F. In lieu of preparing the media 
from the individual ingredients specified, they may be made from 
dehydrated mixtures that, when reconstituted with distilled water, have 
the same composition as such media. Minor modifications of the 
individual ingredients specified in this section are permissible if the 
resulting media possess growth-promoting properties at least equal to 
the media described.
    (b) Description of media. Medium numbers 1, 2, 3, 4, 5, 8, 9, 10, 
11, and 13 correspond to those used in ``Assay Methods of Antibiotics,'' 
D. C. Grove and W. A. Randall, Medical Encyclopedia, Inc., New York, 
N.Y. (1955) p. 220, which is incorporated by reference. Copies are 
available from Medical Encyclopedia Inc., 30 East 60th St., New York, 
NY, or available for inspection at the Office of the Federal Register, 
800 North Capitol Street, NW., suite 700, Washington, DC. Medium numbers 
18 through 21 correspond to those used in ``Outline of Details for 
Official Microbiological Assays of Antibiotics,'' A. Kirshbaum and B. 
Arret, ``Journal of Pharmaceutical Sciences,'' vol. 56, No. 4, April 
1967, p. 512, which is incorporated by reference. Copies are available 
from the American Pharmaceutical Association, 2215 Constitution Ave. 
NW., Washington, DC 20037, or available for inspection at the Office of 
the Federal Register (see address in this paragraph).
    (1) Medium 1.

Peptone: 6.0 gm.
Pancreatic digest of casein: 4.0 gm.
Yeast extract: 3.0 gm.
Beef extract: 1.5 gm.
Dextrose: 1.0 gm.
Agar: 15.0 gm.
Distilled water, q.s: 1,000.0 ml.
pH 6.5 to 6.6 after sterilization.

    (2) Medium 2.

Peptone: 6.0 gm.
Yeast extract: 3.0 gm.
Beef extract: 1.5 gm.
Agar: 15.0 gm.
Distilled water, q.s: 1,000.0 ml.
pH 6.5 to 6.6 after sterilization.

    (3) Medium 3.

Peptone: 5.0 gm.
Yeast extract: 1.5 gm.
Beef extract: 1.5 gm.
Sodium chloride: 3.5 gm.
Dextrose: 1.0 gm.
Dipotassium phosphate: 3.68 gm.
Potassium dihydrogen phosphate: 1.32 gm.
Distilled water, q.s: 1,000.0 ml.
pH 6.95 to 7.05 after sterilization.

    (4) Medium 4.

Peptone: 6.0 gm.
Yeast extract: 3.0 gm.
Beef extract: 1.5 gm.
Dextrose: 1.0 gm.
Agar: 15.0 gm.
Distilled water, q.s: 1,000.0 ml.
pH 6.5 to 6.6 after sterilization.

    (5) Medium 5. Medium 5 is the same as medium 2, except adjust the 
final pH to 7.8 to 8.0 after sterilization.
    (6)-(7) [Reserved]
    (8) Medium 8. Medium 8 is the same as medium 2, except adjust the 
final pH to 5.8 to 6.0 after sterilization.
    (9) Medium 9.

Pancreatic digest of casein: 17.0 gm.
Papaic digest of soybean: 3.0 gm.
Sodium chloride: 5.0 gm.
Dipotassium phosphate: 2.5 gm.
Dextrose: 2.5 gm.
Agar: 20.0 gm.
Distilled water, q.s: 1,000.0 ml.
pH 7.2 to 7.3 after sterilization.

    (10) Medium 10. Medium 10 is the same as medium 9, except:

Agar: 12.0 gm.
Polysorbate 80 (add polysorbate 80 after boiling the medium to dissolve 
the agar): 10.0 ml.
pH 7.2 to 7.3 after sterilization.

    (11) Medium 11. Medium 11 is the same as medium 1, except adjust the 
final pH to 7.8 to 8.0 after sterilization.
    (12) [Reserved]
    (13) Medium 13.

Peptone: 10.0 gm.

[[Page 363]]

Dextrose: 20.0 gm.
Distilled water, q.s: 1,000.0 ml.
pH 5.6 to 5.7 after sterilization.

    (14)-(18) [Reserved]
    (19) Medium 19.

Peptone: 9.4 gm.
Yeast extract: 4.7 gm.
Beef extract: 2.4 gm.
Sodium chloride: 10.0 gm.
Dextrose: 10.0 gm.
Agar: 23.5 gm.
Distilled water, q.s: 1,000.0 ml.
pH 6.0 to 6.2 after sterilization.

    (20)-(31) [Reserved]
    (32) Medium 32. Prepare as medium 1, except add 300 milligrams of 
hydrated manganese sulfate (MnSO4H2O) to 
each liter of medium.
    (33) Medium 33. Use medium 1, sterilized and cooled to 50 deg. C. 
Aseptically add sufficient sterile sodium novobiocin solution to give a 
final concentration of 10 micrograms of novobiocin activity per 
milliliter of medium. Sterile sodium novobiocin solution is prepared by 
filtering a solution containing 2.5 milligrams of novobiocin per 
milliliter of distilled water through a membrane filter of 0.22-micron 
porosity.
    (34) Medium 34.

Glycerol: 10.0 gm.
Peptone: 10.0 gm.
Beef extract: 10.0 gm.
Sodium chloride: 3.0 gm.
Distilled water, q.s.: 1,000.0 ml.
pH 7.0 after sterilization.

    (35) Medium 35. Same as medium 34, except add 17.0 grams of agar to 
each liter of medium.
    (36) Medium 36.

                                                                        
                                                                        
                                                                        
Pancreatic digest of casein.................................    15.0 gm.
Papaic digest of soybean....................................     5.0 gm.
Sodium chloride.............................................     5.0 gm.
Agar........................................................    15.0 gm.
Distilled water, q.s........................................     1,000.0
                                                                     ml.
pH 7.3 after sterilization..................................  ..........
                                                                        

    (37) Medium 37.

Pancreatic digest of casein: 17.0 gm.
Soybean peptone: 3.0 gm.
Dextrose: 2.5 gm.
Sodium chloride: 5.0 gm.
Dipotassium phosphate: 2.5 gm.
Distilled water, q.s.: 1,000.0 ml.
pH 7.3 after sterilization.

    (38) Medium 38.

Peptone: 15.0 gm.
Papaic digest of soybean meal: 5.0 gm.
Sodium chloride: 4.0 gm.
Sodium sulfite: 0.2 gm.
L-cystine: 0.7 gm.
Dextrose: 5.5 gm.
Agar: 15.0 gm.
Distilled water, q.s.: 1,000.0 ml.
pH 7.0 after sterilization.
[39 FR 18944, May 30, 1974, as amended at 40 FR 52004, Nov. 7, 1975; 42 
FR 14092, Mar. 15, 1977; 47 FR 9396, Mar. 5, 1982; 47 FR 22514, May 25, 
1982]



Sec. 436.103  Test organisms.

    (a) Preparation of test organism suspensions. For each test organism 
listed in the following table, select the media (as listed by medium 
number in Sec. 436.102(b)), incubation period of the Roux bottle, 
suggested dilution factor, and suggested storage period for the 
particular test organism and proceed by the appropriate method described 
in paragraph (b) of this section. Test organism letters A through K, M, 
and N correspond to those used in ``Outline of Details for Official 
Microbiological Assays of Antibiotics,'' A. Kirshbaum and B. Arret, 
``Journal of Pharmaceutical Sciences,'' Vol. 56, No. 4, p. 512 (April 
1967), which is incorporated by reference. Copies are available from the 
American Pharmaceutical Association, 2215 Constitution Ave. NW., 
Washington, DC 20037, or available for inspection at the Office of the 
Federal Register, 800 North Capitol Street, NW., suite 700, Washington, 
DC.

                                                                                                                
----------------------------------------------------------------------------------------------------------------
                                               Medium used for the--  Incubation               Suggested storage
                                    Method   ------------------------  period of   Suggested       period of    
         Test organisms              used                    Roux        Roux      dilution    suspensions under
                                                Slants      bottles     bottle      factor       refrigeration  
----------------------------------------------------------------------------------------------------------------
Test organism A--Staphylococcus            1           1           1    24 hours        1:20  1 week.           
 aureus (ATCC 6538P) \2\.                                                                                       
Test organism B--Micrococcus               1           1           1    24 hours        1:30  2 weeks.          
 luteus (ATCC 7468) \2\.                                                                                        
Test organism C--Micrococcus               1           1           1    24 hours        1:40  2 weeks.          
 luteus (ATCC 9341) \2\.                                                                                        
Test organism D--Staphyloccocus            1           1           1    24 hours    \1\ 1:14  1 week.           
 epidermidis (ATCC 12228) \2\.                                                                                  

[[Page 364]]

                                                                                                                
Test organism E--Saccharomyces             6          19  ..........  ..........        1:30  4 weeks.          
 cerevisiae (ATCC 9763) \2\.              or                                                                    
                                           7          19          19    48 hours        1:30  4 weeks.          
Test organism F--Bordetella                1           1           1    24 hours        1:20  2 weeks.          
 bronchiseptica (ATCC 4617) \2\.                                                                                
Test organism G--Bacillus cereus           3           1           1      1 week  ..........  6 months.         
 var. mycoides (ATCC 11778) \2\.                                                                                
Test organism H--Bacillus                  1           1           1    24 hours  ..........  6 months.         
 subtilis (ATCC 6633) \2\.                or                                                                    
                                           2           1          32      5 days  ..........  6 months.         
Test organism I--Klebsiella                1           1           1    24 hours        1:25  1 week.           
 pneumoniae (ATCC 10031) \2\.                                                                                   
Test organism J--Escherichia               1           1           1    24 hours        1:20  2 weeks.          
 coli (ATCC 10536).                                                                                             
Test organism K--Streptococcus             5  ..........  ..........  ..........  ..........  24 hours.         
 faecium (ATCC 10541) \2\.                                                                                      
Test organism L--Micrococcus               1           1           1    24 hours        1:35  4 weeks.          
 luteus (ATCC 10240) \2\.                                                                                       
Test organism O--Staphylococcus            1          33          33    24 hours        1:10  4 weeks.          
 aureus, resistant to novobiocin                                                                                
 (ATCC 12692) \2\.                                                                                              
Test organism T--Saccharomyces             7          19          19    48 hours        1:30  4 weeks.          
 cerevisiae (ATCC 2601) \2\.                                                                                    
Test organism V--                          1           1           1    48 hours        1:35  4 weeks.          
 Micrococcusluteus, resistant to                                                                                
 dihydrostreptomycin (ATCC                                                                                      
 10240A) \2\.                                                                                                   
Test organism W--Pseudomonas               1           1           1    24 hours        1:25  2 weeks.          
 aeruginosa (ATCC 25619) \2\.                                                                                   
Test organism X--Mycobacterium             8          36  ..........  ..........  ..........  2 weeks.          
 smegmatis (ATCC 607)..                                                                                         
Test organism Y--Pseudomonas               9          36          36    24 hours        1:50  1 week.           
 aeruginosa (ATCC 29336) \2\.                                                                                   
----------------------------------------------------------------------------------------------------------------
\1\ If the antibiotic to be tested is paromomycin, the dilution factor is 1:25.                                 
\2\ Available from American Type Culture Collection, 12301 Parklawn Dr., Rockville, MD. 20852.                  

    (b) Methods for preparation of test organism suspensions--(1) Method 
1--(i) Preparation of suspension. Maintain organisms on agar slants 
containing 10 milliliters of the appropriate medium. Incubate the slants 
at 32 deg. C.-35 deg. C. for 24 hours. Using 3 milliliters of sterile 
U.S.P. saline T.S., wash the growth from the agar slant onto a large 
agar surface, such as a Roux bottle, containing 250 milliliters of the 
appropriate medium. Spread the suspension of organisms over the entire 
surface of the Roux bottle with the aid of sterile glass beads. Incubate 
the Roux bottle at 32 deg. C.-35 deg. C. Wash the resulting growth from 
the agar surface with 50 milliliters of sterile U.S.P. saline T.S.
    (ii) Standardization of suspension. Determine the dilution factor 
that will give a 25-percent light transmission at a wavelength of 580 
millimicrons using a suitable photoelectric colorimeter and a 13-
millimeter diameter test tube as an absorption cell. It may be necessary 
to adjust the suspension. Determine the amount of suspension to be added 
to each 100 milliliters of agar or nutrient broth by the use of test 
plates or test broth. Store the test organism suspension under 
refrigeration.
    (2) Method 2. Proceed as directed in paragraph (b)(1) of this 
section, except in lieu of paragraph (b)(1)(ii) thereof, heat-shock and 
standardize the suspension as follows: Centrifuge and decant the 
supernatant liquid. Resuspend the sediment with 50 to 70 milliliters of 
sterile U.S.P. saline T.S. and heat the suspension for 30 minutes at 
70 deg. C. Use test plates to assure the viability of the spores and to 
determine the amount of spore suspension to be added to each 100 
milliliters of agar. Maintain the spore suspension under refrigeration.
    (3) Method 3. Proceed as directed in paragraph (b)(1) of this 
section, except in lieu of paragraph (b)(1)(ii) thereof, heat-shock and 
standardize the suspension as follows: Heat the suspension for

[[Page 365]]

30 minutes at 70 deg. C. Wash the spore suspension three times with 25 
to 50 milliliters of sterile distilled water. Resuspend the organisms in 
50 to 70 milliliters of sterile distilled water and heat-shock again for 
30 minutes at 70 deg. C. Use test plates to assure the viability of the 
spores and to determine the amount of spore suspension to be added to 
each 100 milliliters of agar. Maintain the spore suspension under 
refrigeration.
    (4)  [Reserved]
    (5) Method 5. Maintain the test organisms in 100-milliliter 
quantities of nutrient broth--Medium 3 as described in 
Sec. 436.102(b)(3). For the test prepare a fresh subculture by 
transferring a loopful of the stock culture to 100 milliliters of the 
same nutrient broth and incubate for 16 to 18 hours at 37 deg. C. Store 
this broth culture under refrigeration.
    (6) Method 6. Maintain the test organisms on agar slants containing 
10 milliliters of the medium specified in paragraph (a) of this section. 
Incubate the slants at 32 deg. C.-35 deg. C. for 24 hours. Inoculate 100 
milliliters of nutrient broth--Medium 13 as described in 
Sec. 436.102(b)(13). Incubate for 16 to 18 hours at 37 deg. C. Proceed 
as directed in paragraph (b)(1)(ii) of this section.
    (7) Method 7. Proceed as directed in paragraph (b)(1) of this 
section, except incubate the slants at 30 deg. C. for 24 hours and 
incubate the Roux bottle at 30 deg. C. for 48 hours.
    (8) Method 8. Maintain organisms on agar slants containing 10 
milliliters of the appropriate medium and transfer to a fresh slant 
about once a week. Incubate the slants at 37 deg. C for 48 hours. Using 
3 milliliters of sterile U.S.P. saline T.S., wash the growth from the 
agar slant into a 500-milliliter Erlenmeyer flask containing 100 
milliliters of medium 34, as described in Sec. 436.102(b) (34), and 50 
grams of glass beads. Agitate the culture by rotation at a speed of 130 
cycles per minute and a radius of 3.5 centimeters at 27 deg. C for 5 
days. Determine the amount of suspension to be added to each 100 
milliliters of agar by the use of test plates. Store the test organism 
suspension under refrigeration.
    (9) Method 9. Proceed as directed in paragraph (b)(1) of this 
section, except incubate the slant and Roux bottle at 37 deg. C and wash 
the resulting growth from the agar surface with 50 milliliters of Medium 
37 as described in Sec. 436.102(b)(37).
[39 FR 18944, May 30, 1974, as amended at 40 FR 52004, Nov. 7, 1975; 42 
FR 14092, Mar. 15, 1977; 42 FR 18058, Apr. 5, 1977; 44 FR 10378, Feb. 
20, 1979; 47 FR 22514, May 25, 1982; 47 FR 27552, June 25, 1982]



Sec. 436.104  Penicillin activity.

    Use penicillin-free equipment and glassware.
    (a) Preparation of inoculated plates. Proceed as directed in 
Sec. 436.105(a), using 10 milliliters of medium 1 for the base layer. 
For the seed layer, use 4 milliliters of medium 4, inoculated with the 
amount of test organism C which gave the clearest, sharpest zones of 
inhibition measuring 17 to 21 millimeters in diameter when standardized 
as described in Sec. 436.103(b)(1)(ii). Use the plates the same day they 
are prepared.
    (b) Preparation of working standard stock solutions and standard 
response lines solutions. Proceed as directed for penicillin G in 
Sec. 436.105(b), except dilute the working standard stock solution to a 
final concentration of 100 units of penicillin G per milliliter and use 
the following final concentrations for the standard response line: 
0.005, 0.0125, 0.025, 0.050, 0.100, and 0.200 unit of penicillin G per 
milliliter. The 0.050 unit of penicillin G-per-milliliter solution is 
the reference concentration of the assay.
    (c) Sample preparation. Dissolve 1.0 gram of the sample in 
sufficient distilled water to make 18 milliliters. Filter if not clear. 
Transfer 9.0 milliliters to a separatory funnel, and add 20 milliliters 
of amyl acetate. Add 1 milliliter of 10 percent potassium phosphate 
buffer, pH 2.5 (solution 11 as described in Sec. 436.101), shake, allow 
to separate, and draw off the aqueous layer into a second separatory 
funnel. Check the pH of the aqueous solution with pH paper, and readjust 
with concentrated hydrochloric acid if the pH is three or above. Extract 
again with 20 milliliters or amyl acetate, discard the aqueous phase, 
and combine the amyl acetate extracts. Wash the extracts with 10 
milliliters of 1 percent potassium phosphate buffer, pH 2.5, and discard 
the

[[Page 366]]

buffer wash. Extract the penicillin from the amyl acetate with a 10-
milliliter aliquot of 1 percent potassium phosphate buffer, pH 6.0 
(solution 1 as described in Sec. 436.101). This is the assay solution.
    (d) Procedure for assay. For the standard response line, use a total 
of 15 plates (three plates for each response line solution, except the 
reference concentration solution, which is included on each plate). On 
each set of three plates, fill three alternate cylinders with the 
reference concentration solution and the other three cylinders with the 
concentration of the response line under test. Thus, there will be 45 
reference concentration zones of inhibition and nine zones of inhibition 
for each of the other concentrations of the response line. Treat a 
portion of the sample solution (2 to 5 milliliters) with 0.1 milliliter 
of penicillinase solution and incubate at 37 deg. C. for 1 hour. For 
each sample tested, use three plates. On each plate fill two cylinders 
with the 0.050 unit of penicillin G per milliliter standard, two 
cylinders with the untreated sample, and two cylinders with the 
penicillinase-treated sample. Incubate all plates, including those of 
the standard response line, overnight at 30 deg. C. A zone of inhibition 
with the untreated sample and no zone with the penicillinase-treated 
sample are a positive test for penicillin. If a positive test is 
obtained, measure the diameters of the zones of inhibition using an 
appropriate measuring device such as a millimeter rule, calipers, or an 
optical projector.
    (e) Estimation of penicillin G activity. To prepare the standard 
response line, average the diameters of the standard reference 
concentration and average the diameters of the standard response line 
concentration tested for each set of three plates. Average also all 45 
diameters of the reference concentration. The average of the 45 
diameters of the reference concentration is the correction point of the 
response line. Correct the average diameter obtained for each 
concentration to the figure it would be if the average reference 
concentration diameter for that set of three plates were the same as the 
correction point. Thus, if in correcting the 0.025 penicillin G 
concentration, the average of the 45 readings of the 0.050 unit of 
penicillin G-per-milliliter concentration is 18.5 millimeters and the 
average of the 0.050 unit of pencillin G-per milliliter concentration of 
this set of three plates is 18.3 millimeters, the correction is +0.2 
millimeters. If the average reading of the 0.025 unit of pencillin G-
per-milliliter concentration of these same three plates is 15.5 
millimeters, the corrected value is 15.7 millimeters. Plot these 
corrected values, including the average of the 0.050 unit of penicillin 
G-per-milliliter concentration, on semilogarithmic graph paper using the 
pencillin concentration in units per milliliter on the logarithmic scale 
and the diameter of the zone of inhibition on the arithmetic scale. Draw 
the line of best fit through these points. To estimate the sample 
potency, average the zone diameters of the standard and the zone 
diameters of the sample on the three plates used. If the average zone 
diameter of the sample is lower than that of the standard, subtract the 
difference between them from the reference concentration diameter of the 
standard response line. From the response line, read the concentrations 
corresponding to these corrected values of zone diameters. Multiply the 
concentration by the dilution factor to obtain the units of penicillin G 
per sample size tested.
[39 FR 18944, May 30, 1974, as amended at 41 FR 34743, Apr. 17, 1976]



Sec. 436.105  Microbiological agar diffusion assay.

    Using the sample solution prepared as described in the section for 
the particular antibiotic to be tested, proceed as described in 
paragraphs (a), (b), (c), and (d) of this section.
    (a) Preparation of inoculated plates. For each antibiotic listed in 
the table in this paragraph, select the media (as listed by medium 
number in Sec. 436.102(b)), the amount of media to be used in the base 
and seed layers, the test organism (as listed in Sec. 436.103(a)), and 
the suggested inoculum and prepare the inoculated plates as follows: 
Prepare the base layer by adding the appropriate amount of melted agar 
to each Petri dish (nominal dimensions 20 by 100 millimeters). 
Distribute the agar

[[Page 367]]

evenly in each dish on a flat, level surface, placing a cover on each 
plate in turn; if a nonporous cover is used, leave it slightly ajar to 
prevent accumulation of condensed moisture from the hot agar base layer. 
After the agar hardens, seat the nonporous cover on each plate. To 
prepare the seed layer, add the suggested inoculum of the test organism 
suspension to a sufficient amount of agar, which has been melted and 
cooled to 48 deg. C-50 deg. C. Swirl the flask to obtain a homogeneous 
suspension, and add the appropriate amount of the inoculated media to 
each of the plates containing the uninoculated base agar. Spread evenly 
over the agar surface, cover, and allow to harden on a flat, level 
surface. After the agar has hardened, place 6 cylinders described in 
Sec. 436.100(a)(1) on the inoculated agar surface so that they are at 
approximately 60 deg. intervals on a 2.8--centimeter radius.

                                                                                                                
----------------------------------------------------------------------------------------------------------------
                                Media to be used (as  Milliliters of media               Suggested              
                                  listed by medium      to be used in the                volume of              
                                   number in Sec.     base and seed layers             standardized   Incubation
                                     436.102(b))     ----------------------    Test     inoculum to  Temperature
          Antibiotic           ----------------------                        organism   be added to    for the  
                                                         Base       Seed                 each 100       plates  
                                   Base       Seed      layer      layer                milliliters             
                                  layer      layer                                     of seed agar             
----------------------------------------------------------------------------------------------------------------
                                                                                        Milliliters   Degrees C.
Amoxicillin...................         11         11         21          4          C           0.5        32-35
Amphotericin B................       None         19       None          8          E           1.0        29-31
Ampicillin....................         11         11         21          4          C           0.5        32-35
Bacitracin....................          2          1         21          4          B           0.3        32-35
Bacitracin....................          2          1         21          4          L           0.3        32-35
Bleomycin.....................         35         35         10          6          X           1.0        32-35
Carbenicillin.................          9         10         21          4          W       \2\ 0.5      36-37.5
Cefactor......................          2          1         21          5          A          0.05      36-37.5
Cefadroxil....................          2          1         21          4          A          0.05      36-37.5
Cefamandole...................          2          1         21          5          A          0.06      36-37.5
Cefazolin.....................          2          1         21          4          A          0.05        32-35
Cefotaxime....................          2          1         21          5          A           0.1      36-37.5
Cefoxitin.....................          2          1         21          5          A           0.1      36-37.5
Cephalexin....................          2          1         21          4          A          0.05        32-35
Cephaloglycin.................          2          1         21          4          A           0.2        32-35
Cephaloridine.................          2          1         21          4          A           0.1        32-35
Cephalothin...................          2          1         21          4          A           0.1        32-35
Cephapirin....................          2          1         21          4          A          0.08        32-35
Cephradine....................          2          1         21          4          A          0.05        32-35
Clindamycin...................         11         11         21          4          C           1.5      36-37.5
Cloxacillin...................          2          1         21          4          A           0.1        32-35
Colistimethate, sodium........          9         10         21          4          F           0.1      36-37.5
Colistin......................          9         10         21          4          F           0.1      36-37.5
Cyclacillin...................         11         11         21          4          C           0.5      36-37.5
Dactinomycin..................          5          5         10          4          H         (\1\)      36-37.5
Dicloxacillin.................          2          1         21          4          A           0.1        32-35
Dihydrostreptomycin...........          5          5         21          4          H         (\1\)      36-37.5
Erythromycin..................         11         11         21          4          C           1.5        32-35
Gentamicin....................         11         11         21          4          D          0.03      36-37.5
Kanamycin B...................          5          5         21          4          H         (\1\)      36-37.5
Methicillin...................          2          1         21          4          A           0.3        32-35
Mitomycin.....................          8          8         10          4          H           0.5      36-37.5
Nafcillin.....................          2          1         21          4          A           0.3        32-35
Natamycin.....................       None         19       None          8          E           0.8        29-31
Neomycin......................         11         11         21          4          A           0.4        32-35
Neomycin......................         11         11         21          4          D           1.0      36-37.5
Netilmicin....................         11         11         20          5          D          0.25      36-37.5
Novobiocin....................          2          1         21          4          D           4.0        34-36
Nystatin......................       None         19       None          8          T           1.0        29-31
Oleandomycin..................         11         11         21          4          D           1.0      36-37.5
Oxacillin.....................          2          1         21          4          A           0.3        32-35
Paromomycin...................         11         11         21          4          D           2.0      36-37.5
Penicillin G..................          2          1         21          4          A           1.0        32-35
Penicillin V..................          2          1         21          4          A           1.0        32-35
Plicamycin....................          8          8         10          4          A           0.1        32-35
Polymyxin B...................          9         10         21          4          F           0.1      36-37.5
Rifampin......................          2          2         21          4          H           0.1        29-31
Sisomicin.....................         11         11         21          4          D          0.03      36-37.5
Streptomycin..................          5          5         21          4          H         (\1\)      36-37.5
Ticarcillin...................         38         38         21          4          Y           1.5      36-37.5

[[Page 368]]

                                                                                                                
Vancomycin....................          8          8         10          4          H         (\1\)      36-37.5
----------------------------------------------------------------------------------------------------------------
\1\ Determine the amount of the inoculum by the use of test plates.                                             
\2\ Use dilution of the suspension that gives 25 percent light transmission in lieu of the stock suspension.    

    (b) Preparation of working standard stock solutions and standard 
response line solutions. For each antibiotic listed in the table in this 
paragraph, select the working standard drying conditions, solvent(s), 
concentrations, and storage time for the standard solutions and proceed 
as follows: If necessary, dry the working standard as described in 
Sec. 436.200; dissolve and dilute an accurately weighed portion to the 
proper concentration to prepare the working standard stock solution. 
Store the working standard stock solution under refrigeration and do not 
use longer than the recommended storage time. Further dilute an aliquot 
of the working standard stock solution to the proper concentrations to 
prepare the standard response line solutions. The reference 
concentration of the assay is the mid concentration of the response 
line.

[[Page 369]]



--------------------------------------------------------------------------------------------------------------------------------------------------------
                                                              Working standard stock solutions                                  Standard response line  
                               ---------------------------------------------------------------------------------------------        concentrations      
                                                                                                                            ----------------------------
                                                                                                                                             Final      
                                     Drying                         Diluent (solution        Final                                      concentrations, 
          Antibiotic               conditions                       number as listed     concentration       Storage time                   units or    
                                 (method number   Initial solvent        in Sec.            units or            under         Diluent    micrograms of  
                                  as listed in                         436.101(a))       milligrams per     refrigeration                  antibiotic   
                                 Sec.  436.200)                                            milliliter                                     activity per  
                                                                                                                                           milliliter   
--------------------------------------------------------------------------------------------------------------------------------------------------------
Amoxicillin...................  Not dried......  ................  Distilled water...  1.0 mg...........  7 days...........         3  0.064, 0.080,    
                                                                                                                                        0.100, 0.125 and
                                                                                                                                        0.156, g. (Prepare   
                                                                                                                                        the standard    
                                                                                                                                        response line   
                                                                                                                                        simultaneously  
                                                                                                                                        with the sample 
                                                                                                                                        solution.)      
Amphotericin B................  1..............  ................  Dimethylsulfoxide.  1 mg.\1\.........  Use same day.....        10  0.64 0.80, 1.00, 
                                                                                                                                        1.25, 1.56      
                                                                                                                                        g.     
                                                                                                                                        (Prepare the    
                                                                                                                                        standard        
                                                                                                                                        response line   
                                                                                                                                        simultaneously  
                                                                                                                                        with the sample 
                                                                                                                                        solution.)      
Ampicillin....................  Not dried......  ................  Distilled water...  0.1..............  1 week...........         3  0.064, 0.080,    
                                                                                                                                        0.100, 0.125    
                                                                                                                                        0.156 g. (Prepare   
                                                                                                                                        the standard    
                                                                                                                                        response line   
                                                                                                                                        simultaneously  
                                                                                                                                        with the sample 
                                                                                                                                        solution.)      
Bacitracin zinc...............  1..............  ................  0.01N HCl.........  100 units........  Use same day.....         1  0.64, 0.80, 1.0, 
                                                                                                                                        1.25, 1.56      
                                                                                                                                        units.          
Bleomycin.....................  7..............  ................  16................  2 units..........  2 weeks..........        16  0.01, 0.02, 0.04,
                                                                                                                                        0.08, 0.16 unit.
Carbenicillin.................  Not dried......  ................  1.................  1 mg.............  2 weeks..........         1  12.8, 16.0, 20.0,
                                                                                                                                        25.0, 31.2      
                                                                                                                                        g.     
Cefaclor......................  Not dried......  ................  1.................  1 mg.............  1 day............         1  3.2, 4.0, 5.0,   
                                                                                                                                        6.25, 7.81      
                                                                                                                                        g.     
Cefadroxil....................  Not dried......  ................  1.................  1mg..............  Use same day.....         1  12.8,16.0, 20.0, 
                                                                                                                                        25.0, and       
                                                                                                                                        31.2g. 
Cefamandole...................  Not dried......  ................  3.................  1mg \5\..........  1 day............         1  1.28, 1.60, 2.00,
                                                                                                                                        2.50, 3.12      
                                                                                                                                        g.     
Cefazolin.....................  Not dried......  10,000 g per ml. in                                                                        1.25, 1.56      
                                                  solution 6.                                                                           g.     
Cefotaxime....................  Not dried......  1...............  ..................  1 mg.............  Use same day.....         1  6.4, 8.0, 10.0,  
                                                                                                                                        12.5, 15.6      
                                                                                                                                        g.     
Cefoxitin.....................  Not dried......  ................  1.................  1 mg.............  Use same day.....         1  12.8, 16.0, 20.0,
                                                                                                                                        25, 31.2 g.            
Cephalexin....................  Not dried......  ................  1.................  1 mg.............  7 days...........         1  12.8, 16.0, 20.0,
                                                                                                                                        25.0, 31.2      
                                                                                                                                        g.     
Cephaloglycin.................  Not dried......  ................  Distilled water...  100 g...  1 week...........         4  6.4, 8.0, 10.0,  
                                                                                                                                        12.5, 15.6      
                                                                                                                                        g.     
Cephaloridine.................  1..............  ................  1.................  1 mg.............  5 days...........         1  0.64, 0.80, 1.00,
                                                                                                                                        1.25, 1.56      
                                                                                                                                        g.     
Cephalothin...................  1..............  ................  1.................  1 mg.............  5 days...........         1  0.64, 0.80, 1.00,
                                                                                                                                        1.25, 1.56      
                                                                                                                                        g.     
Cephapirin....................  Not dried......  ................  1.................  1 mg.............  3 days...........         1  0.64, 0.80, 1.00,
                                                                                                                                        1.25, 1.56      
                                                                                                                                        g.     
Cephradine....................  Not dried......  ................  1.................  1 mg.............  5 days...........         1  6.4, 8.0, 10.0,  
                                                                                                                                        12.5, 15.6      
                                                                                                                                        g.     
Clindamycin...................  Not dried......  ................  Distilled water...  1 mg.............  1 month..........         3  0.64, 0.80, 1.00,
                                                                                                                                        1.25 1.56 g.            
Cloxacillin...................  Not dried......  ................  1.................  1 mg.............  7 days...........         1  3.20, 4.00, 5.00,
                                                                                                                                        6.25, 7.81      
                                                                                                                                        g.     
Colistimethate, sodium........  1..............  10,000 g. per ml. in                                                                       1.25, 1.56      
                                                  distilled water.                                                                      g.     
Colistin......................  1..............  10,000 g. per ml. in                                                                       1.25, 1.56      
                                                  distilled water.                                                                      g.     
Cyclacillin...................  Not dried......  ................  Distilled water...  1 mg.............  1 day............         3  0.64, 0.80, 1.0, 
                                                                                                                                        1.25, 1.56      
                                                                                                                                        g.     
                                                                                                                                        (Prepare the    
                                                                                                                                        standard        
                                                                                                                                        response line   
                                                                                                                                        simultaneously  
                                                                                                                                        with the sample 
                                                                                                                                        solution.)      
Dactinomycin..................  1..............  10,000 g. per ml. in                                                                       1.41, 2.00      
                                                  methyl alcohol.                                                                       g.     
Dicloxacillin.................  Not dried......  ................  1.................  1 mg.............  7 days...........         1  3.20, 4.00, 5.00,
                                                                                                                                        6.25, 7.81      
                                                                                                                                        g.     
Dihydrostreptomycin...........  5..............  ................  3.................  1 mg.............  30 days..........         3  0.64, 0.80, 1.00,
                                                                                                                                        1.25, 1.56      
                                                                                                                                        g.     

[[Page 370]]

                                                                                                                                                        
Erythromycin..................  1..............  10,000 g. per ml. in                                                                       1.25, 1.56      
                                                  methyl alcohol.                                                                       g.     
Gentamicin....................  3..............  ................  3.................  1 mg.............  1 month..........         3  0.064, 0.080,    
                                                                                                                                        0.100, 0.125,   
                                                                                                                                        0.156 g.            
Kanamycin B (use the kanamycin  Not dried......  ................  3.................  1 mg.............  1 month..........         3  0.64, 0.80, 1.00,
 sulfate working standard).                                                                                                             1.25, 1.56      
                                                                                                                                        g.     
Methicillin...................  Not dried......  ................  1.................  1 mg.............  4 days...........         1  6.4, 8.0, 10.0,  
                                                                                                                                        12.5, 15.6      
                                                                                                                                        g.     
Mitomycin.....................  Not dried......  ................  1.................  1 mg.............  14 days..........         1  0.50, 0.71, 1.0, 
                                                                                                                                        1.41, 2.0 g.            
Nafcillin.....................  Not dried......  ................  1.................  1 mg.............  2 days...........         1  1.28, 1.60, 2.00,
                                                                                                                                        2.50, 3.12      
                                                                                                                                        g.     
Natamycin.....................  Not dried......  ................  Dimethylsulfoxide.  1 mg \7\.........  Use same day.....        10  3.20, 4.00, 5.00,
                                                                                                                                        6.25, 7.81      
                                                                                                                                        g.     
                                                                                                                                        (Prepare the    
                                                                                                                                        standard        
                                                                                                                                        response line   
                                                                                                                                        solutions       
                                                                                                                                        simultaneously  
                                                                                                                                        with the sample 
                                                                                                                                        solution to be  
                                                                                                                                        tested using red
                                                                                                                                        low actinic     
                                                                                                                                        glassware. Use  
                                                                                                                                        solutions within
                                                                                                                                        2 hours after   
                                                                                                                                        preparation.)   
Neomycin......................  1..............  ................  3.................  1 mg.............  2 weeks..........         3  0.64, 0.80, 1.00,
                                                                                                                                        1.25, 1.56      
                                                                                                                                        g. (if 
                                                                                                                                        test organism D 
                                                                                                                                        is used); 6.4,  
                                                                                                                                        8.0, 10.0, 12.5,
                                                                                                                                        15.6 g.
                                                                                                                                        (if test        
                                                                                                                                        organism A is   
                                                                                                                                        used).          
Netilmicin....................  Not dried \9\..  ................  3.................  1 mg.............  7 days...........         3  0.064, 0.080,    
                                                                                                                                        0.100, 0.125,   
                                                                                                                                        0.156  
                                                                                                                                        g.              
Novobiocin....................  5..............  10,000 g. per ml. in                                                                       0.500, 0.625,   
                                                  absolute ethyl                                                                        0.781 g.            
Nystatin \10\.................  4..............  ................  Dimethylformamide.  1,000 units \2\..  Use same day.....         6  12.8, 16.0, 20.0,
                                                                                                                                        25.0, 31.2      
                                                                                                                                        units. (Prepare 
                                                                                                                                        the standard    
                                                                                                                                        response line   
                                                                                                                                        solutions       
                                                                                                                                        simultaneously  
                                                                                                                                        with the sample 
                                                                                                                                        solution to be  
                                                                                                                                        tested using red
                                                                                                                                        low actinic     
                                                                                                                                        glassware.)     
Oleandomycin..................  Not dried......  10,000 g. per ml. in                                                                       6.25, 7.81      
                                                  ethyl alcohol.                                                                        g.     
Oxacillin.....................  Not dried......  ................  1.................  1 mg.............  3 days...........         1  3.20, 4.00, 5.00,
                                                                                                                                        6.25, 7.81      
                                                                                                                                        g.     
Paromomycin...................  1..............  ................  3.................  1 mg.............  3 weeks..........         3  0.64, 0.80, 1.00,
                                                                                                                                        1.25, 1.56      
                                                                                                                                        g.     
Penicillin G..................  Not dried......  ................  1.................  1,000 units......  4 days...........         1  0.64, 0.80, 1.00,
                                                                                                                                        1.25, 1.56      
                                                                                                                                        units.          
Penicillin V Potassium........  Not dried......  ................  1.................  100 units........  4 days...........         1  0.64, 0.80, 1.00,
                                                                                                                                        1.25, 1.56      
                                                                                                                                        units.          
Plicamycin....................  7..............  ................  Distilled water...  0.1 mg...........  1 day............         1  0.50, 0.7, 1.00, 
                                                                                                                                        1.41, 2.00      
                                                                                                                                        g.     
Polymyxin B...................  1..............  Distilled water   6.................  10,000 units.....  2 weeks..........         6  6.4, 8.0, 10.0,  
                                                  \3\.                                                                                  12.5, 15.6      
                                                                                                                                        units.          
Rifampin......................  Not dried......  ................  Methyl alcohol....  1 mg.............  1 day............         1  3.20, 4.00, 5.00,
                                                                                                                                        6.25, 7.81,     
                                                                                                                                        g.     
Streptomycin..................  1..............  ................  3.................  1 mg.............  30 days..........         3  0.64, 0.80, 1.00,
                                                                                                                                        1.25, 1.56      
                                                                                                                                        g.     

[[Page 371]]

                                                                                                                                                        
Sisomicin \6\.................  Not dried \8\..  ................  3.................  1 mg.............  14 days..........         3  0.064, 0.080,    
                                                                                                                                        0.100, 0.125,   
                                                                                                                                        0.156 g.            
Ticarcillin...................  Not dried......  ................  1.................  1 mg.............  1 day............         1  3.20, 4.00, 5.00,
                                                                                                                                        6.25, 7.81      
                                                                                                                                        g.     
Vancomycin....................  1..............  ................  Distilled water...  1 mg.............  1 week...........         4  6.4, 8.0, 10.0,  
                                                                                                                                        12.5, 15.6      
                                                                                                                                        g.     
--------------------------------------------------------------------------------------------------------------------------------------------------------
\1\ Further dilute aliquots of the working standard stock solution with dimethylsulfoxide to give concentrations of 12.8, 16, 20.0, 25, and 31.2        
  micrograms per milliliter.                                                                                                                            
\2\ Further dilute aliquots of the working standard stock solution with dimethylformamide to give concentrations of 256, 320, 400, 500, and 624 units   
  per milliliter.                                                                                                                                       
\3\ Add 2 milliliters of distilled water for each 5 milligrams of weighed working standard material.                                                    
\4\ Further dilute aliquots of the working standard stock solution with dimethylformamide to give concentrations of 64, 80, 100, 125, and 156 micrograms
  per milliliter.                                                                                                                                       
\5\ The final concentration of the working standard stock solution is allowed to hydrolyze in a 37 deg. C. constant temperature water bath for 60       
  minutes.                                                                                                                                              
\6\ Working standard should be stored below minus 20 deg. C under an atmosphere of nitrogen. Sisomicin is hygroscopic and care should be exercised      
  during weighing.                                                                                                                                      
\7\ Further dilute aliquots of the working standard stock solution with dimethylsulfoxide to give concentrations 64.0, 80.0, 100, 125, and 156          
  micrograms per milliliter.                                                                                                                            
\8\ Weigh a separate portion of the working standard and determine the loss on drying by the method described in Sec.  436.200(c) of this chapter. Use  
  this value to determine the anhydrous content of the working standard.                                                                                
\9\ Working standard should be stored below minus 10 deg. C under an atmosphere of nitrogen. Netilmicin sulfate is hygroscopic and care should be       
  exercised during weighing.                                                                                                                            
\10\ For assay of nystatin pastilles, use 80 percent aqueous dimethylformamide as the initial solvent and as diluent for all dilutions where            
  dimethylformamide is required.                                                                                                                        

[[Page 372]]

                                                                                                                                                        

    (c) Procedure for assay. For the standard response line, use a total 
of 12 plates--three plates for each response line solution, except the 
reference concentration solution which is included on each plate. On 
each set of three plates, fill three alternate cylinders with the 
reference concentration solution and the other three cylinders with the 
concentration of the response line under test. Thus, there will be 36 
reference concentration zones of inhibition and nine zones of inhibition 
for each of the four other concentrations of the response line. For each 
sample tested use three plates. Fill three alternate cylinders on each 
plate with the standard reference concentration solution and the other 
three cylinders with the sample reference concentration solution. After 
all the plates have incubated for 16 to 18 hours at the appropriate 
incubation temperature for each antibiotic listed in the table in 
paragraph (b) of this section, measure the diameters of the zones of 
inhibition using an appropriate measuring device such as a millimeter 
rule, calipers, or an optical projector.
    (d) Estimation of potency. To prepare the standard response line, 
average the diameters of the standard reference concentration and 
average the diameters of the standard response line concentration tested 
for each set of three plates. Average also all 36 diameters of the 
reference concentration for all four sets of plates. The average of the 
36 diameters of the reference concentration is the correction point of 
the response line. Correct the average diameter obtained for each 
concentration to the figure it would be if the average reference 
concentration diameter for that set of three plates were the same as the 
correction point. Thus, if in correcting the highest concentration of 
the response line, the average of the 36 diameters of the reference 
concentration is 16.5 millimeters and the average of the reference 
concentration of the set of three plates (the set containing the highest 
concentration of the response line) is 16.3 millimeters, the correction 
is +0.2 millimeter. If the average reading of the highest concentration 
of the response line of these same three plates is 16.9 millimeters, the 
corrected diameter is then 17.1 millimeters. Plot these corrected 
diameters, including the average of the 36 diameters of the reference 
concentration on 2-cycle semilog paper, using the concentration of the 
antibotic in micrograms or units per milliliter as the ordinate (the 
logarithmic scale), and the diameter of the zone of inhibition as the 
abscissa. The response line is drawn either through these points by 
inspection or through points plotted for highest and lowest zone 
diameters obtained by means of the following equation:
[GRAPHIC] [TIFF OMITTED] TR01JA93.000

where:
L=Calculated zone diameter for the lowest concentration of the standard 
response line;
H=Calculated zone diameter for the highest concentration of the standard 
response line;
c=Average zone diameter of 36 readings of the reference point standard 
solution;
a, b, d, e=Corrected average values for the other standard solutions, 
lowest to highest concentration, respectively.


To estimate the potency of the sample, average the zone diameters of the 
standard and the zone diameters of the sample on the three plates used. 
If the average zone diameter of the sample is larger than that of the 
standard, add the difference between them to the reference concentration 
diameter of the standard response line. If the average zone diameter of 
the sample is lower than that of the standard, subtract the difference 
between them from the reference concentration diameter of the standard 
response line. From the response line, read the concentrations 
corresponding to these corrected values of zone diameters. Multiply the 
concentration by the appropriate dilution factor to obtain the 
antibiotic content of the sample.
[39 FR 18944, May 30, 1974]

    Editorial Note: For Federal Register citations affecting 
Sec. 436.105, see the List of CFR Sections Affected appearing in the 
Finding Aids section of this volume.

[[Page 373]]



Sec. 436.106  Microbiological turbidimetric assay.

    Using the sample solution prepared as described in the section for 
the particular antibiotic to be tested, proceed as described in 
paragraphs (a), (b), and (c) of this section.
    (a) Preparation of working standard stock solutions and standard 
response line solutions. For each antibiotic listed in the table in this 
paragraph, select the working standard, drying conditions, solvent(s), 
concentrations, and storage time for the standard solutions and proceed 
as follows: If necessary, dry the working standard as described in 
Sec. 436.200; dissolve and dilute an accurately weighed portion to the 
proper concentration for the working standard stock solution. Store the 
working standard stock solution under refrigeration and do not use 
longer than the recommended storage time. Prepare the proper 
concentrations for the standard response line solutions by further 
diluting an aliquot of the working standard stock solution. The 
reference concentration of the assay is the mid concentration of the 
standard response line.

[[Page 374]]



                                                                                                                                                                                                
------------------------------------------------------------------------------------------------------------------------------------------------------------------------------------------------
                                                                    Working standard stock solutions                                                  Standard response line concentrations     
                         -----------------------------------------------------------------------------------------------------------------------------------------------------------------------
                            Drying conditions                                                                                                                            Final concentrations-- 
       Antibiotic           (method number as                             Diluent (solution number   Final concentration   Storage time under     Diluent (solution      units or micrograms of 
                             listed in Sec.          Initial solvent          as listed in Sec.      units or milligrams      refrigeration      number as listed in    antibiotic activity per 
                                436.200)                                         436.101(a))           per milliliter                             Sec.  436.101(a))            milliliter       
------------------------------------------------------------------------------------------------------------------------------------------------------------------------------------------------
Amikacin................  Not dried...........  ........................  Distilled water.........  1 mg................  2 weeks.............  Distilled water......  8.0, 8.9, 10.0, 11.2,    
                                                                                                                                                                        12.5 g.        
Candicidin \1\..........  6...................  ........................  Dimethyl sulfoxide......  1 mg................  Use same day........  Distilled water......  0.030, 0.043, 0.060,     
                                                                                                                                                                        0.085, 0.120 g.
                                                                                                                                                                        (Prepare standard       
                                                                                                                                                                        response line           
                                                                                                                                                                        simultaneously with the 
                                                                                                                                                                        sample solution.)       
Capreomycin.............  5...................  Distilled water.........  ........................  1 mg................  7 days..............  Distilled water......  80.0, 89.0, 100.0, 112,0,
                                                                                                                                                                        125.0 g.       
Chloramphenicol.........  Not dried...........  Ethyl alcohol (10,000     Distilled water.........  1 mg................  1 month.............  Distilled water......  2.00, 2.24, 2.50, 2.80,  
                                                 g. per ml.).                                                                                                  3.12 g.        
Chlortetracycline.......  Not dried...........  ........................  0.01N HCl...............  1 mg................  4 days..............  Distilled water......  0.048, 0.054, 0.060,     
                                                                                                                                                                        0.067, 0.075 g.
Cycloserine.............  1...................  ........................  Distilled water.........  1 mg................  1 month.............  Distilled water......  40.0, 44.5, 50.0, 56.0,  
                                                                                                                                                                        62.5 g.        
Demeclocycline..........  1...................  ........................  0.1N HCl................  1 mg................  4 days..............  Distilled water......  0.080, 0.089, 0.100,     
                                                                                                                                                                        0.112, 0.125 g.
Dihydrostreptomycin.....  5...................  ........................  Distilled water.........  1 mg................  30 days.............  Distilled water......  24.0, 26.8, 30.0, 33.5,  
                                                                                                                                                                        37.5 g.        
Doxycycline.............  Not dried...........  ........................  0.1N HCl................  1 mg................  5 days..............  Distilled water......  0.080, 0.089, 0.100,     
                                                                                                                                                                        0.112, 0.125 g.
Gramicidin..............  1...................  ........................  alcohol U.S.P. XX.......  1 mg................  30 days.............  alcohol U.S.P. XX....  0.032, 0.0356, 0.040,    
                                                                                                                                                                        0.0448, 0.050 g.                    
Kanamycin...............  Not dried...........  ........................  Distilled water.........  1 mg................  1 month.............  Distilled water......  8.0, 8.9, 10.0, 11.2,    
                                                                                                                                                                        12.5 g.        
Lincomycin..............  Not dried...........  ........................  Distilled water.........  1 mg................  1 month.............  Distilled water......  0.400, 0.447, 0.500,     
                                                                                                                                                                        0.559, 0.625 g.
Meclocycline............  Not dried...........  ........................  13......................  1 mg................  Use same day........  Distilled water......  0.048, 0.054, 0.06,      
                                                                                                                                                                        0.067, 0.075 g.
Methacycline............  1...................  ........................  Distilled water.........  1 mg................  7 days..............  Distilled water......  0.048, 0.054, 0.060,     
                                                                                                                                                                        0.067, 0.075 g.
Oxytetracycline.........  Not dried...........  ........................  0.1N HCl................  1 mg................  4 days..............  Distilled water......  0.192, 0.215, 0.240,     
                                                                                                                                                                        0.268, 0.300 g.
Rolitetracycline........  1...................  ........................  Distilled water.........  1 mg................  1 day...............  Distilled water......  0.192, 0.215, 0.240,     
                                                                                                                                                                        0.268, 0.300 g.
Spectinomycin...........  Not dried...........  ........................  Distilled water.........  1 mg................  1 month.............  Distilled water......  24.0, 26.8, 30.0, 33.5,  
                                                                                                                                                                        37.5 g.        
Streptomycin............  1...................  ........................  Distilled water.........  1 mg................  30 days.............  Distilled water......  24.0, 26.8, 30.0, 33.5,  
                                                                                                                                                                        37.5 g.        
Tetracycline............  Not dried...........  ........................  0.1N HCl................  1 mg................  1 day...............  Distilled water......  0.192, 0.215, 0.240,     
                                                                                                                                                                        0.268, 0.300 g.
Tobramycin..............  Not dried...........  ........................  Distilled water.........  1 mg................  2 weeks.............  Distilled water......  2.00, 2.236, 2.5, 2.795, 
                                                                                                                                                                        3.125 g.       
Troleandomycin..........  1...................  ........................  15......................  1 mg................  Use same day........  Distilled water......  20.0, 22.25, 25.0, 28.0, 
                                                                                                                                                                        31.25 g.       
Tyrothricin \2\.........  ....................  ........................  ........................  ....................  ....................  .....................  .........................
------------------------------------------------------------------------------------------------------------------------------------------------------------------------------------------------
\1\ Use sterile equipment for all stages of this assay.                                                                                                                                         
\2\ The gramicidin working standard and the gramicidin standard response line concentrations are used for the assay of tyrothricin.                                                             


[[Page 375]]

    (b) Procedure for assay. For each antibiotic listed in the table in 
this paragraph, select the test organism (as listed in Sec. 436.103(a)), 
nutrient broth (as listed by medium number in Sec. 436.102(b)), and 
suggested inoculum and proceed as follows: Place 1.0 milliliter (or 0.1 
milliliter in the case of gramicidin and tyrothricin) of each 
concentration of the standard response line (prepare as described in 
paragraph (a) of this section) and of the sample solution in each set of 
three replicate tubes (as described in Sec. 436.100(b)(1)). Fifteen 
tubes are used for the five-point standard response line and three for 
each sample. To each tube add 9 milliliters of the inoculated broth and 
place immediately in a water bath at the appropriate temperature for 2 
to 4 hours. The exact length of the incubation period should be 
determined by observation of growth in the reference concentration tube 
of the standard. Remove the tubes from the water bath and add 0.5 
milliliter of a 12-percent formaldehyde solution to each tube. Determine 
the absorbance value of each tube in a suitable photoelectric 
colorimeter, at a wavelength of 530 millimicrons. Set the instrument at 
zero absorbance with an uninoculated blank composed of the same amounts 
of nutrient broth and formaldehyde used in the assay.

    Note: The amount of working standard and sample solutions may be 
reduced as long as all other solutions used are reduced proportionately.

----------------------------------------------------------------------------------------------------------------
                                                                                         Suggested              
                                                                                         volume of              
                                                                                       standardized             
                                                                                        inoculum to             
                                                                  Test       Medium     be added to   Incubation
                          Antibiotic                            organism    (nutrient    each 100    temperature
                                                                             broth)     milliliters             
                                                                                         of medium              
                                                                                         (nutrient              
                                                                                          broth)                
----------------------------------------------------------------------------------------------------------------
                                                                                       Milliliters    Degrees C.
Amikacin.....................................................           A           3          0.1       36-37.5
Candicidin \1\...............................................           E          13          0.2         27-29
Capreomycin..................................................           I           3         0.05       36-37.5
Chloramphenicol..............................................           J           3          0.7       36-37.5
Chlortetracycline............................................           A           3          0.1       36-37.5
Cycloserine..................................................           A           3          0.4       36-37.5
Demeclocycline...............................................           A           3          0.1       36-37.5
Dihydrostreptomycin..........................................           I           3          0.1       36-37.5
Doxycycline..................................................           A           3          0.1       36-37.5
Gramicidin...................................................           K           3          1.0       36-37.5
Kanamycin....................................................           A           3          0.2       36-37.5
Lincomycin...................................................           A           3          0.1       36-37.5
Meclocycline.................................................           A           3          0.2       36-37.5
Methacycline.................................................           A           3          0.1       36-37.5
Oxytetracycline..............................................           A           3          0.1       36-37.5
Rolitetracycline.............................................           A           3          0.1       36-37.5
Spectinomycin................................................           J           3          0.1       36-37.5
Streptomycin.................................................           I           3          0.1       36-37.5
Tetracycline.................................................           A           3          0.1       36-37.5
Tobramycin...................................................           A           3         0.15       36-37.5
Troleandomycin...............................................           I           3          0.1       36-37.5
Tyrothricin..................................................           K           3          1.0       36-37.5
----------------------------------------------------------------------------------------------------------------
\1\ Use sterile equipment for all stages of this assay.                                                         

[GRAPHIC] [TIFF OMITTED] TC01AP94.000

where:
L=Calculated absorbance value for the lowest concentration of the 
standard response line.
H=Calculated absorbance value for the highest concentration of the 
standard response line.

[[Page 376]]

a, b, c, d, e=Average absorbance values for each concentration of the 
standard response line, lowest to the highest, respectively.

    (c) Estimation of potency. To prepare the standard response line, 
plot the average absorbance values for each concentration of the 
standard response line on one-cycle semilogarithmic graph paper with the 
absorbance values on the arithmetric scale and concentrations on the 
logarithmic scale. The response line is drawn either through these 
points by inspection or through points plotted for highest and lowest 
absorbance values obtained by means of the following equations.

To estimate the potency of the sample, average the absorbance values for 
the sample and determine the antibiotic concentration from the standard 
response line. Multiply the concentration by the appropriate dilution 
factor to obtain the antibiotic content of the sample.
[39 FR 18944, May 30, 1974, as amended at 40 FR 57797, Dec. 12, 1975; 41 
FR 49483, Nov. 9, 1976; 44 FR 22057, Apr. 13, 1979; 46 FR 3835, 3839, 
Jan. 16, 1981; 46 FR 16682, Mar. 13, 1981; 46 FR 33512, June 30, 1981; 
46 FR 61072, Dec. 15, 1981; 47 FR 22515, May 25, 1982; 47 FR 23708, June 
1, 1982; 48 FR 3960, Jan. 28, 1983; 53 FR 32607, Aug. 26, 1988]



            Subpart E--General Chemical Tests for Antibiotics



Sec. 436.200  Loss on drying.

    Use the method specified in the individual section for each 
antibiotic.
    (a) Method 1. In an atmosphere of about 10 percent relative 
humidity, grind the sample, if necessary, to obtain a fine powder. When 
tablets, troches, or capsules are to be tested, use four tablets, 
troches, or capsules in preparing the sample. Transfer about 100 
milligrams of the sample to a tared weighing bottle equipped with a 
ground-glass stopper. Weigh the bottle and place it in a vacuum oven, 
tilting the stopper on its side so that there is no closure during the 
drying period. Dry at a temperature of 60 deg. C. and a pressure of 5 
millimeters of mercury or less for 3 hours. At the end of the drying 
period, fill the vacuum oven with air dried by passing it through a 
drying agent such as sulfuric acid or silica gel. Replace the stopper 
and place the weighing bottle in a desiccator over a desiccating agent, 
such as phosphorous pentoxide or silica gel, allow to cool to room 
temperature, and reweigh. Calculate the percent of loss.
    (b) Method 2. Proceed as directed in paragraph (a) of this section, 
except use a tared weighing bottle or weighing tube equipped with a 
capillary-tube stopper, the capillary having an inside diameter of 0.20-
0.25 millimeter, and place it in a vacuum oven without removing the 
stopper.
    (c) Method 3. Proceed as directed in paragraph (a) of this section, 
except dry the sample at a temperature of 110 deg. C. and a pressure of 
5 millimeters of mercury or less for 3 hours.
    (d) Method 4. Proceed as directed in paragraph (a) of this section, 
except dry the sample at a temperature of 40 deg. C. and a pressure of 5 
millimeters of mercury or less for 2 hours.
    (e) Method 5. Proceed as directed in paragraph (a) of this section, 
except dry the sample at a temperature of 100 deg. C. and a pressure of 
5 millimeters of mercury or less for 4 hours.
    (f) Method 6. Proceed as directed in paragraph (a) of this section, 
except dry the sample at a temperature of 40 deg. C. and a pressure of 5 
millimeters of mercury or less for 3 hours.
    (g) Method 7. Proceed as directed in paragraph (a) of this section, 
except dry the sample at a temperature of 25 deg. C. and a pressure of 5 
millimeters of mercury or less for 4 hours.
    (h) Method 8. Proceed as directed in paragraph (a) of this section, 
except transfer approximately 300 milligrams of the sample to a tared 
weighing bottle equipped with a ground-glass stopper; dry the sample at 
a temperature of 25  deg. C and a pressure of 5 millimeters of mercury 
or less for 4 hours, and then dry the sample at a temperature of 100 
deg.C and a pressure of 5 millimeters of mercury or less for 3 
additional hours.
    (i) Method 9. Use a suitable thermogravimetric apparatus prepared 
for vacuum operation. Rapidly and thoroughly grind a portion of the 
sample and promptly transfer 5 to 10 milligrams to the sample pan. Place 
the system under vacuum and allow it to come to equilibrium before 
heating. Obtain an accurate sample weight and

[[Page 377]]

continuously record the weight loss as the sample is heated at a rate of 
20 deg. per minute from room temperature to about 200  deg. C. The 
weight loss plateau, or inflection, at about 150  deg. C is taken as the 
total volatile weight loss. Calculate the percent weight loss on drying.
[39 FR 18944, May 30, 1974, as amended at 50 FR 48397, Nov. 25, 1985; 51 
FR 11572, Apr. 4, 1986]



Sec. 436.201  Moisture determination.

    (a) Equipment--(1) Apparatus. Use a closed system consisting of all 
glass automatic burettes, platinum electrodes, and a magnetic stirrer 
connected to a suitable electrometric apparatus. This apparatus embodies 
a simple electrical circuit which serves to pass 5 to 10 microamperes of 
direct current between a pair of platinum electrodes immersed in the 
solution to be titrated. At the endpoint of the titration a slight 
excess of the reagent increases the flow of current to between 50 and 
150 microamperes for 30 seconds or longer, depending upon the solution 
being titrated.
    (2) Titrating vessel. Use a suitable titrating vessel which has been 
previously dried at 105 deg. C. and cooled in a desiccator.
    (b) Reagents--(1) Karl Fischer reagent. Dissolve 125 grams of iodine 
in 170 milliliters of pyridine, add 670 milliliters of methanol and 
cool. To 100 milliliters of pyridine kept in an ice bath, add sulfur 
dioxide until the volume reaches 200 milliliters. Slowly add this 
solution to the cooled iodine-methanol-pyridine mixture and shake well. 
(A commercially prepared Karl Fischer reagent, pyridine containing or 
pyridine-free, may be used.) Preserve the reagent in glass-stoppered 
bottles protected from light and from moisture in the air.
    (2) Methanol solution. Add sufficient water (usually 2 milligrams 
per milliliter) to methanol so that each milliliter of the resulting 
methanol solution is equivalent to about 0.5 milliliter of Karl Fischer 
reagent.
    (3) Solvents--(i) Solvent A. Methanol:chloroform:carbon 
tetrachloride (1:2:2 by volume).
    (ii) Solvent B. Chloroform:carbon tetrachloride (1:1 by volume).
    (iii) Solvent C. Anhydrous methanol.
    (c) Standardization of reagents--(1) Water equivalence of Karl 
Fischer reagent. Standardize the Karl Fischer reagent no longer than 1 
hour before use by one of the following methods.
    (i) Accurately weigh 25-35 milligrams of water into a dry titration 
vessel and add 20 milliliters of solvent A. Start the stirrer and 
titrate to the endpoint by adding measured quantities of Karl Fischer 
reagent. Calculate the water equivalence of the Karl Fischer reagent as 
follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.001

where:
  e=Water equivalence of the Karl Fischer reagent in terms of milligrams 
          of water per milliliter;
  W=Milligrams of water;
  VT=Milliliters of Karl Fischer reagent used;
  VA=Milliliters of Karl Fischer reagent equivalent to the 20 
          milliliters of solvent A, determined as directed in paragraph 
          (c)(3) of this section.

    (ii) Accurately weigh about 25-35 milligrams of water into a dry 
titration vessel, add an excess of Karl Fischer reagent, start the 
stirrer, and titrate to the endpoint with methanol solution. Calculate 
the water equivalence of the Karl Fischer reagent as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.001

where:
  e=Water equivalence of the Karl Fischer reagent in terms of milligrams 
          of water per milliliter;
  W=Milligrams of water;
  VT=Milliliters of Karl Fischer reagent used;
  Vm=Milliliters of methanol solution used;
  f=Milliliters of Karl Fischer reagent equivalent to each milliliter of 
          methanol solution determined as directed in paragraph (c)(2) 
          of this section.

    (2) Karl Fischer reagent equivalence of methanol solution. Titrate a 
known volume of Karl Fischer reagent with methanol solution until the 
endpoint is reached. Calculate the milliliters of Karl Fischer reagent 
equivalent to each milliliter of methanol solution as follows:

[[Page 378]]

[GRAPHIC] [TIFF OMITTED] TC01AP94.002


where:
  f=Milliliters of Karl Fischer reagent equivalent to each milliliter of 
          methanol solution;
  VT=Milliliters of Karl Fischer reagent used;
  Vm=Milliliters of methanol solution used.

    (3) Karl Fischer reagent equivalence of solvents. (i) Solvent A: Use 
20 milliliters of solvent A as the sample. Start the stirrer and titrate 
to the endpoint by adding measured quantities of Karl Fischer reagent.
    (ii) Solvent B: Use 10 milliliters of solvent B as the sample. Add 
an excess of Karl Fischer reagent to the sample and start the stirrer. 
Titrate to the endpoint with methanol solution.
    (iii) Solvent C. Use 20 milliliters of solvent C as the sample. 
Start the stirrer and titrate to the endpoint by adding measured 
quantities of Karl Fischer reagent.
    (iv) Calculate the Karl Fischer reagent equivalence of the solvents 
as follows:

VA=VC=VT,

VB=(VT-Vm) X f

where:
  VA,VB, and VC=Milliliters of Karl 
          Fischer reagent equivalent to the aliquots used of solvents A, 
          B, and C, respectively;
  VT=Milliliters of Karl Fischer reagent used;
  Vm=Milliliters of methanol solution used;
  f=Milliliters of Karl Fischer reagent equivalent to each milliliter of 
          methanol solution determined as directed in paragraph (c)(2) 
          of this section.

    (d) Sample preparation--(1) Powders. In the case of tablets, grind 4 
tablets to a fine powder. In the case of capsules containing enteric-
coated pellets, grind the pellets to a fine power. If the maximum 
moisture limit is greater than 1 percent, accurately weigh about 300 
milligrams of the sample into a dry titrating vessel. If the maximum 
moisture limit is less than 1 percent, accurately weigh 1 to 2 grams of 
the sample. Proceed as directed in paragraph (e)(1) or (2) of this 
section.
    (2) Ointments and oils. (i) Transfer about 1 to 2 grams, accurately 
weighed, into a dry titrating vessel. Proceed as directed in paragraph 
(e)(1) of this section; or
    (ii) Transfer about 1 to 2 grams, accurately weighed, into a dry 
titrating vessel. Add 10 milliliters of solvent B and proceed as 
directed in paragraph (e)(2) of this section.
    (3) Aerosols with propellant. Place the immediate container to be 
tested in a suitable freezing unit having a temperature of not higher 
than 0 deg. C. for at least 2 hours. Remove the container from the 
freezing unit, puncture it, mix the entire contents by swirling. Proceed 
as directed in paragraph (e)(3) of this section, using an accurately 
measured 10-milliliter aliquot from the container as the sample and 
allowing the solution to warm to at least 10 deg. C. before determining 
the endpoint.
    (4) Hygroscopic powders. Weigh the immediate container. Using a 
suitable dry hypodermic needle and syringe, inject 3 milliliters of 
anhydrous methanol into the container and shake to dissolve the 
contents. Using the same syringe, remove the withdrawable contents and 
transfer into the titration vessel. Rinse the syringe and needle by 
drawing in an additional 3 milliliters of anhydrous methanol. Add the 
rinsings to the titration vessel. Titrate the solution immediately, 
proceeding as directed in paragraph (e)(3) of this section. Determine 
the Karl Fischer equivalent (in milliliters), if any, of the anhydrous 
methanol by titrating a blank of the same total volume used in preparing 
the sample and rinsing the syringe and needle. Dry the immediate 
container and its closure for three hours at 100 deg. C., cool to room 
temperature in a desiccator, and weigh. Determine the weight of sample 
tested by subtracting the weight obtained from the original weight of 
the immediate container.
    (5) Solutions. Proceed as directed in paragraph (e)(3) of this 
section, using about 1 to 2 grams of the sample, accurately weighted.
    (e) Titration procedures and calculations--(1) Procedure 1. Add 20 
milliliters of solvent A to the sample. Start the stirrer and titrate to 
the endpoint by adding measured quantities of Karl Fischer reagent. 
Determine the percent moisture in the sample as follows:

[[Page 379]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.001


where:
  e=Water equivalence of the Karl Fischer reagent determined as directed 
          in paragraph (c)(1) of this section;
  VT=Milliliters of Karl Fischer reagent used;
  VA=Milliliters of Karl Fischer reagent equivalent to the 20 
          milliliters of solvent A, determined as directed in paragarph 
          (c)(3) of this section;
  Ws=Weight of the sample in milligrams.

    (2) Procedure 2. Add an excess of Karl Fischer reagent to the 
sample, start the stirrer, and titrate to the endpoint with methanol 
solution. Calculate the percent moisture in the sample as follows:
    (i) For powders:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.002
    
    (ii) For oils and ointments:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.003
    
where:
VT=Milliliters of Karl Fischer reagent used;
Vm=Milliliters of methanol solution used;
f=Milliliters of Karl Fischer reagent equivalent to each milliliter of 
methanol solution determined as directed in paragraph (c)(2) of this 
section.
VB=Milliliters of Karl Fischer reagent equivalent to the 10 
milliliters of solvent B determined as directed in paragraph (c)(3) of 
this section;
e=Water equivalence of the Karl Fischer reagent determined as directed 
in paragraph (c)(1) of this section;
Ws=Weight of the sample in milligrams.

    (3) Procedure 3. Add about 20 milliliters of solvent A to a dry 
titrating vessel and proceed as directed in titration procedure 1 or 2. 
Disregard the volume of reagents used to determine the endpoint. 
Promptly introduce an accurately weighed or measured quantity of sample 
into the titrating vessel and titrate to the endpoint using either 
titration procedure 1 or 2 without additional solvents. Calculate the 
percent moisture in the sample as follows:
    (i) If titration procedure 1 is used:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.004
    
    [GRAPHIC] [TIFF OMITTED] TR01JA93.005
    
Percent moisture in hygroscopic powders=
[GRAPHIC] [TIFF OMITTED] TC01AP94.003

    (ii) If titration procedure 2 is used:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.006
    
    [GRAPHIC] [TIFF OMITTED] TR01JA93.007
    
    [GRAPHIC] [TIFF OMITTED] TR01JA93.008
    
where:
VT=Milliliters of Karl Fischer reagent used;
Vm=Milliliters of methanol solution used;
f=Milliliters of Karl Fischer reagent equivalent to each milliliter of 
methanol solution determined as directed in paragraph (c)(2) of this 
section;
Vb=Karl Fischer equivalent (in milliliters) of the methanol 
used as a sample solvent;
e=Water equivalence of the Karl Fischer reagent determined as directed 
in paragraph (c)(1) of this section.
[39 FR 18944, May 30, 1974, as amended at 48 FR 51292, Nov. 8, 1983; 50 
FR 41679, Oct. 15, 1985; 51 FR 22071, June 18, 1986; 51 FR 27532, Aug. 
1, 1986]



Sec. 436.202  pH.

    (a) Apparatus. A suitable potentiometer fitted with two electrodes, 
one being constructed of glass and sensitive to hydrogen ion activity 
and the other being a calomel or a silver/silver chloride reference 
electrode. A combination electrode with glass electrode and reference 
electrode contained in the same system may be used.
    (b) Standardization. Select two standard buffer solutions such that 
the expected pH value of the sample is within their pH range and is also 
within 2 pH

[[Page 380]]

unit of one of the standard buffer solutions. Standardize the pH meter 
with the two buffer solutions. Make any necessary adjustment of the 
meter if the observed pH value of either standard solution differs by 
more than 0.05 pH units of its known value.
    (c) Sample preparation. If necessary, dilute the sample with carbon 
dioxide-free distilled water to the concentration specified in the 
individual section for each antibiotic.
    (d) Test procedure. Determine the pH of the sample at 
25 deg.plus-minus2 deg. C. Rinse the electrode(s) between 
determinations first with distilled water and then with a portion of the 
next sample to be tested. Store electrode(s) with tips immersed in 
water.
[39 FR 18944, May 30, 1974, as amended at 42 FR 29857, June 10, 1977; 42 
FR 31449, June 21, 1977]



Sec. 436.203  Crystallinity.

    Use the method specified in the individual section for each 
antibiotic.
    (a) Method 1. To prepare the sample for examination, mount a few 
particles in mineral oil on a clean glass slide. Examine the sample by 
means of a polarizing microscope. The particles reveal the phenomena of 
birefringence and extinction positions on revolving the microscope 
stage.
    (b) Method 2. Proceed as directed in paragraph (a) of this section, 
except to prepare the sample for examination, mount a few particles in 
mineral oil, add 1 drop of ethyl alcohol, and allow to react for about 
30 seconds.



Sec. 436.204  Iodometric assay.

    (a) Reagents. (1) 0.01N Sodium thiosulfate (2.482 grams 
Na2S2O35H2O and 125 
milligrams Na2CO3 per liter).
    (2) 1.0N Sodium hydroxide.
    (3) 1.2N Hydrochloric acid.
    (4) 0.01N Iodine solution (prepared from 0.1N iodine U.S.P.).
    (5) Starch iodide paste, T.S. (U.S.P.).
    (b) Preparation of sample and working standard solutions--(1) 
Workingstandard solutions. From the following table, select the initial 
solvent, diluent, and final concentration as listed for each antibiotic 
working standard. Dissolve and dilute an accurately weighed portion to 
the specified final concentration and proceed as directed in paragraphs 
(c) and (d) of this section.

                                                                                                                
----------------------------------------------------------------------------------------------------------------
                                                                                          Final concentration in
                                                                Diluent (solution number  units or milligrams of
             Antibiotic                   Initial solvent          as listed in Sec.           activity per     
                                                                      436.101(a))         milliliter of standard
                                                                                                 solution       
----------------------------------------------------------------------------------------------------------------
Amoxicillin.........................  None...................  Distilled water..........  1.0 mg.               
Ampicillin..........................  ......do...............  ......do.................  1.25 mg.              
Cephalexin..........................  ......do...............  ......do.................  2 mg.                 
Cephaloridine.......................  ......do...............  ......do.................  2 mg.                 
Cephalothin.........................  ......do...............  ......do.................  2 mg.                 
Cephapirin..........................  ......do...............  ......do.................  2 mg.                 
Cloxacillin.........................  ......do...............  ......do.................  1.25 mg.              
Cyclacillin.........................  ......do...............  ......do.................  1.0 mg.               
Dicloxacillin.......................  ......do...............  1........................  1.25 mg.              
Methicillin.........................  ......do...............  1........................  1.25 mg.              
Nafcillin...........................  ......do...............  1........................  1.25 mg.              
Oxacillin...........................  ......do...............  1........................  1.25 mg.              
Penicillin G........................  ......do...............  1........................  2,000 units.          
Penicillin V potassium..............  ......do...............  1........................  2,000 units.          
----------------------------------------------------------------------------------------------------------------


                                                                                                                
----------------------------------------------------------------------------------------------------------------
                                                                                          Final concentration in
                                                                Diluent (solution number  units or milligrams of
             Antibiotic                   Initial solvent          as listed in Sec.           activity per     
                                                                      436.101(a))          milliliter of sample 
----------------------------------------------------------------------------------------------------------------
Amoxicillin trihydrate..............  None...................  Distilled water..........  1.0 mg.               
Ampicillin..........................  ......do...............  ......do.................  1.25 mg.              
Ampicillin sodium...................  ......do...............  1........................  1.25 mg.              
Ampicillin trihydrate...............  ......do...............  Distilled water..........  125 mg.               

[[Page 381]]

                                                                                                                
Bacampicillin hydrochloride.........  None...................  ......do.................  1.25 mg.\2\           
Cephalexin..........................  ......do...............  ......do.................  2 mg.                 
Cephaloridine.......................  ......do...............  ......do.................  2 mg.                 
Cephalothin sodium..................  ......do...............  ......do.................  2 mg.                 
Cephapirin sodium...................  ......do...............  ......do.................  2 mg.                 
Cloxacillin sodium monohydrate......  ......do...............  ......do.................  1.25 mg.              
Cloxacillin sodium monohydrate......  ......do...............  ......do.................  1.25 mg.              
Dicloxacillin sodium monohydrate....  ......do...............  1........................  1.25 mg.              
Cyclacillin.........................  ......do...............  Distilled water..........  1.0 mg.               
Methicillin sodium monohydrate......  ......do...............  1........................  1.25 mg.              
Mezlocillin.........................  ......do...............  Distilled water..........  2.0 mg.               
Nafcillin sodium monohydrate........  ......do...............  1........................  1.25 mg.              
Oxacillin sodium monohydrate........  ......do...............  1........................  1.25 mg.              
Penicillin G benzathine blank         ......do...............  Distilled water..........  2,000 units.          
 solution.                                                                                                      
Penicillin G benzathine inactivated   ......do...............  1N NaOH..................  2,000 units.\1\       
 solution.                                                                                                      
Penicillin G potassium..............  ......do...............  1........................  2,000 units.          
Penicillin G procaine...............  2 ml methyl alcohol....  1........................  2,000 units.          
Penicillin G sodium.................  None...................  1........................  2,000 units.          
Penicillin V........................  2 ml methyl alcohol....  1........................  2,000 units.          
Penicillin V potassium..............  ......do...............  1........................  2,000 units.          
----------------------------------------------------------------------------------------------------------------
\1\ Allow to stand in 1N NaOH for 15 minutes before assaying.                                                   
\2\ The final concentration of bacampicillin hydrochloride is calculated in milligrams of ampicillin activity   
  per milliliter of sample. The ampicillin working standard is used for the assay of bacampicillin              
  hydrochloride.                                                                                                

    (3) Finished product solutions. Prepare the sample for assay as 
directed in the individual section for each antibiotic product to be 
tested by diluting to the concentration prescribed in the table in 
paragraph (b)(2) of this section and proceed as described in paragraphs 
(c) and (d) of this section.
    (c) Inactivated sample and standard solutions. (1) Transfer 2.0 
milliliters each of the sample and the appropriate working standard 
solutions to glass-stoppered Erlenmeyer flasks.
    (2) Add 2.0 milliliters of 1N sodium hydroxide, except if the sample 
has been diluted in 1N sodium hydroxide, and allow to stand at room 
temperature for 15 minutes.
    (3) Add 2.0 milliliters of 1.2N hydrochloric acid.
    (4) Add 10.0 milliliters of 0.01N iodine solution, stopper, allow to 
stand at room temperature for 15 minutes, and proceed as directed in 
paragraph (e) of this section.
    (d) Blank determination. Transfer 2.0 milliliters each of the sample 
and the appropriate working standard solutions to glass-stoppered 
Erlenmeyer flasks. Add 10.0 milliliters of 0.01N iodine solution and 
immediately proceed as directed in paragraph (e) of this section.
    (e) Titration procedure. Titrate the excess iodine using 0.01N 
sodium thiosulfate. Toward the end of the titration, add 1 drop of the 
starch iodide paste. Continue the titration by the addition of 0.01- to 
0.02-milliliter portions of 0.01N sodium thiosulfate, shaking vigorously 
after each addition. The endpoint is reached when the blue color of the 
starch-iodine complex is discharged. Calculate the antibiotic content as 
described in paragraph (f) of this section.
    (f) Calculations--(1) F factor determination. Using the appropriate 
working standard for the particular antibiotic to be tested, assay the 
standard as directed in this section. Calculate the F factor (the units 
of micrograms of activity equivalent of each milliliter of 0.01N sodium 
thiosulfate consumed) by means of the following formula:
[GRAPHIC] [TIFF OMITTED] TR01JA93.009

where:
Ws=Actual weight in milligrams of standard in the 2.0 
milliliters titrated;
P=Potency of the working standard in units or micrograms per milligram;
Vs=Milliliters of sodium thiosulfate used in the working 
standard blank determination minus the milliliters of sodium thiosulfate 
used in the titration of the inactivated working standard solution (the 
difference is the equivalent of the number of milliliters of 0.01N 
iodine absorbed by the inactivated standard).


[[Page 382]]


    (2) Bulk antibiotic. Calculate the potency of the sample in units or 
micrograms per milligram by means of the following formula:
[GRAPHIC] [TIFF OMITTED] TR01JA93.010

where:
Vu=Milliliters of sodium thiosulfate used in the sample blank 
determination minus the milliliters of sodium thiosulfate used in the 
titration of the inactivated sample solution (the difference is the 
equivalent of the number of milliliters of 0.01N iodine absorbed by the 
inactivated sample);
Wu=Actual weight in miligrams of sample in the 2.0 
milliliters titrated.

    (3) Finished products. Calculate the potency of the sample in units 
or milligrams by means of the appropriate one of the following formulas:
[GRAPHIC] [TIFF OMITTED] TR01JA93.011

[GRAPHIC] [TIFF OMITTED] TR01JA93.012

where:
d=Dilution factor for the sample;
n=Number of doses or items in the sample assayed.
[39 FR 18944, May 30, 1974, as amended at 39 FR 34032, Sept. 23, 1974; 
42 FR 59856, Nov. 22, 1977; 44 FR 10378, Feb. 20, 1979; 46 FR 2980, Jan. 
13, 1981; 46 FR 25602, May 8, 1981; 46 FR 46312, Sept. 18, 1981; 46 FR 
58298, Dec. 1, 1981; 46 FR 61072, Dec. 15, 1981; 49 FR 6091, Feb. 17, 
1984]



Sec. 436.205  Hydroxylamine colorimetric assay.

    (a) Reagents--(1) Hydroxylamine hydrochloride solution. Dissolve 350 
grams of hydroxylamine hydrochloride in sufficient distilled water to 
make 1 liter.
    (2) Buffer. Dissolve 173 grams of sodium hydroxide and 20.6 grams of 
sodium acetate in sufficient distilled water to make 1 liter.
    (3) Neutral hydroxylamine. Mix 1 volume each of hydroxylamine 
hydrochloride solution described in paragraph (a)(1) of this section and 
the buffer described in paragraph (a)(2) of this section. Check the pH 
and if necessary adjust to pH 7.0plus-minus0.1 by adding an 
additional amount of one of the components. To 1 volume of this 
neutralized solution add 8 volumes of distilled water and 2 volumes of 
95 percent ethanol. This solution should be used for 1 day only.
    (4) Ferric ammonium sulfate. Dissolve 272 grams of ferric ammonium 
sulfate in a mixture of 26 milliliters of concentrated sulfuric acid and 
sufficient distilled water to make 1 liter. This reagent may be used for 
1 week when stored in a brown bottle at room temperature.
    (b) Preparation of working standard solutions. From the following 
table, select the diluent and final concentration as listed for each 
antibiotic working standard. Dissolve and dilute an accurately weighed 
portion to the specified final concentration and proceed as directed in 
paragraph (d) of this section.

                                                                        
------------------------------------------------------------------------
                                                               Final    
                                                           concentration
                                      Diluent (solution    in milligrams
            Antibiotic               number as listed in        per     
                                      Sec.  436.101(a))    milliliter of
                                                              standard  
                                                              solution  
------------------------------------------------------------------------
Amoxicillin......................  Distilled water.......          1.0  
Ampicillin.......................  ......do..............         1.25  
Cefazolin \1\....................  ......................          1.0  
Cephaloridine....................  Distilled water.......          1.0  
Cephalothin......................  ......do..............          2.0  
Cephapirin.......................  ......do..............          1.0  
Cloxacillin......................  1.....................         1.25  
Cyclacillin......................  Distilled water.......         1.25  
Dicloxacillin....................  1.....................         1.25  
Methicillin......................  1.....................         1.25  
Nafcillin........................  1.....................         1.25  
Oxacillin........................  1.....................         1.25  
Penicillin G.....................  1.....................         1.25  
Penicillin G procaine............  17....................          2.0  
Penicillin V Potassium...........  1.....................         1.25  
------------------------------------------------------------------------
\1\ To prepare the working standard solution, proceed as directed in the
  individual section of the antibiotic drug regulation in this chapter  
  for the antibiotic to be tested.                                      

    (c) Preparation of sample solutions. From the following table, 
select the diluent and final concentration as listed for each 
antibiotic. Dissolve an accurately weighed portion of the sample, dilute 
to the appropriate final concentration, and proceed as directed in 
paragraph (d) of this section; if the product is packaged for 
dispensing, dilute an aliquot of the stock solution (prepared as 
described in the individual monograph) to the appropriate concentration 
and then proceed as directed in paragraph (d) of this section.

[[Page 383]]



                                                                        
------------------------------------------------------------------------
                                                               Final    
                                                           concentration
                                      Diluent (solution    in milligrams
            Antibiotic               number as listed in        per     
                                      Sec.  436.101(a))    milliliter of
                                                               sample   
------------------------------------------------------------------------
Amoxicillin trihydrate...........  Distilled water.......          1.0  
Ampicillin.......................  ......do..............         1.25  
Ampicillin sodium................  1.....................         1.25  
Ampicillin trihydrate............  Distilled water.......         1.25  
Bacampicillin hydrochloride......  ......do..............     \1\  1.2  
Cefazolin sodium.................  1.....................          1.0  
Cephaloridine....................  Distilled water.......          1.0  
Cephalothin sodium...............  ......do..............          2.0  
Cephapirin sodium................  ......do..............          1.0  
Cloxacillin sodium monohydrate...  1.....................         1.25  
Cyclacillin......................  Distilled water.......         1.25  
Dicloxacillin sodium monohydrate.  ......do..............         1.25  
Methicillin sodium monohydrate...  1.....................         1.25  
Nafcillin sodium monohydrate.....  1.....................         1.25  
Oxacillin sodium monohydrate.....  1.....................         1.25  
Penicillin G potassium...........  1.....................         1.25  
Penicillin G procaine............  17....................          2.0  
Penicillin G sodium..............  1.....................         1.25  
Penicillin V.....................  17....................         1.25  
Penicillin V potassium...........  1.....................         1.25  
------------------------------------------------------------------------
\1\ The final concentration of bacampicillin hydrochloride is calculated
  in milligrams of ampicillin per milliliter of sample. The ampicillin  
  working standard is used for the assay of bacampicillin hydrochloride.

    (d) Procedure. Using a volume of from 1 to 2 milliliters of standard 
or sample solution, add an equal volume of water and mix. Add the 
following reagents in the specified volumetric proportions with respect 
to the sample or standard solutions: Add 1.25 volumes of neutral 
hydroxylamine reagent and allow to react for 5 minutes. Add 1.25 volumes 
of ferric ammonium sulfate reagent, mix, and after 3 minutes determine 
the absorbance of the resulting solution at the wavelength of 480 
millimicrons, using a suitable spectrophotometer and a reagent blank 
prepared by treating a volume of water in the same manner as the 
standard or sample solution. The time elapsed after the addition of the 
ferric ammonium sulfate reagent and the reading of the absorbance must 
be precisely the same (within 10 seconds) for each solution. Calculate 
the potency of the sample in units or micrograms per milligram as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.013

    A1=Absorbance of sample solution.
    A2=Absorbance of standard solution.
[39 FR 18944, May 30, 1974, as amended at 39 FR 34032, Sept. 23, 1974; 
39 FR 44012, Dec. 20, 1974; 42 FR 59856, Nov. 22, 1977; 44 FR 10378, 
Feb. 20, 1979; 45 FR 16474, Mar. 14, 1980; 46 FR 2981, Jan. 13, 1981; 46 
FR 25602, May 8, 1981; 46 FR 61072, Dec. 15, 1981; 49 FR 34350, Aug. 30, 
1984]



Sec. 436.206  Test for metal particles in ophthalmic ointments.

    (a) Procedure. Extrude the contents of each of 10 tubes as 
completely as practicable into separate, clear, glass Petri dishes (60 
millimeters in diameter), cover the dishes, and heat to 80 deg. C. to 
85 deg. C. for at least 2 hours or until the ointment has melted 
completely and evenly in the dishes. A higher temperature of 100 deg. 
C.plus-minus2 deg. C. may be used if necessary to allow 
adequate settling of metal particles. Allow the ointment to cool to room 
temperature without agitation. Invert each Petri dish on the stage of a 
suitable microscope adjusted to furnish 30 times magnification and 
equipped with an eye-piece micrometer disc which has been calibrated at 
the magnification being used. In addition to the usual source of light, 
direct an illuminator from above the ointment at a 45 deg. angle. 
Examine the entire bottom of the Petri dish for metal particles. By 
varying the intensity of the illuminator from above, such metal 
particles are recognized by their characteristic reflection of light. 
Count the total number of metal particles exceeding 50 microns in any 
single dimension.
    (b) Evaluation. The batch is acceptable if (1) a total of not more 
than 50 such particles is found in 10 tubes; and (2) not more than one 
tube is found to contain more than eight such particles. If the batch 
fails the above test, repeat

[[Page 384]]

the test on 20 additional tubes of ointment. The total number of metal 
particles exceeding 50 microns in any single dimension from the 30 tubes 
tested shall not exceed 150, with not more than three tubes containing 
more than eight such particles.
[39 FR 18944, May 30, 1974; 40 FR 11869, Mar. 14, 1975]



Sec. 436.207  Residue on ignition.

    Use the method specified in the individual section for each 
antibiotic.
    (a) Method 1. Place approximately 1 gram of the sample, accurately 
weighed, in a tared porcelain crucible and carefully ignite at a low 
temperature until thoroughly charred. The crucible may be loosely 
covered with a porcelain lid during the charring. Add 2 milliliters of 
nitric acid and 5 drops of sulfuric acid to the contents of the crucible 
and cautiously heat until white fumes are evolved, then ignite, 
preferably in a muffle furnace, at 500 deg. C. to 600 deg. C. until the 
carbon is all burned off. Cool the crucible in a desiccator and weigh. 
From the weight of residue obtained, calculate the sulfated ash content.
    (b) Method 2. Proceed as directed in paragraph (a) of this section, 
except use 2 milliliters of sulfuric acid and do not use the nitric 
acid.



Sec. 436.208  Heavy metals determination.

    (a) Reagents--(1) Ammonia solution. Prepare an aqueous solution 
containing not less than 9 grams and not more than 10 grams of ammonia 
(NH3) per 100 milliliters.
    (2) 6 percent acetic acid. Dilute 60 milliliters of glacial acetic 
acid with sufficient water to give a solution of 1,000 milliliters.
    (3) Hydrogen sulfide solution. Prepare a saturated solution of 
hydrogen sulfide by passing hydrogen sulfide into cold water for a 
sufficient time. It is suitable if it produces an immediate copious 
precipitate when added to an equal volume of 1N ferric chloride. Prepare 
a fresh hydrogen sulfide solution each time a heavy metals test is to be 
performed.
    (4) Lead nitrate stock solution. Dissolve 159.8 milligrams of lead 
nitrate with 100 milliliters of 0.15N nitric acid, and dilute with water 
to a volume of 1,000 milliliters. Prepare and store this solution in 
glass containers free from soluble lead salts.
    (5) Standard lead solution. Dilute a 10-milliliter aliquot of the 
lead nitrate stock solution to 100 milliliters with water. This solution 
must be freshly prepared each time a heavy metals test is performed. One 
milliliter of this standard lead solution represents a lead level of 10 
parts per million in a 1.0-gram sample or 20 parts per million in a 0.5-
gram sample.
    (b) Preparation of the sample. Use the sulfated ash obtained as 
described in Sec. 436.207(a). If the heavy metal limit is greater than 
30 parts per million, the sulfated ash may be obtained from a 0.5-gram 
sample. Add 2 milliliters of hydrochloric acid to the sulfated ash and 
slowly evaporate to dryness on a steam bath. Moisten the residue with 1 
drop of hydrochloric acid, add 10 milliliters of hot water, and digest 
by heating on the steam bath for 2 minutes. After cooling to room 
temperature, add ammonia solution dropwise until a pH of 7.2 is reached, 
then add 2 milliliters of 6 percent acetic acid. Filter the solution, if 
necessary, and wash the crucible and the filter with about 10 
milliliters of water. Combine the washings with the filtrate and dilute 
to exactly 25 milliliters with water.
    (c) Procedure. Prepare a series of five standard lead solutions, in 
increments of 10 parts per million, in which the solution of lowest 
concentration contains 20 parts of lead per million less than the 
maximum limit of heavy metals permitted for the sample. Transfer the 
necessary quantities of standard lead solution described in paragraph 
(a)(5) of this section directly into metal-free 50-milliliter Nessler 
tubes of uniform diameter, add 2 milliliters of 6 percent acetic acid to 
each, and adjust each to a final volume of 25 milliliters with water. 
Transfer the 25-milliliter solution of the sample described in paragraph 
(b) of this section to another Nessler tube. Add 10 milliliters of 
hydrogen sulfide solution to each standard and sample solution, mix 
well, and allow to stand for 10 minutes. View downward over a white 
surface; the color of the solution of the sample should be no darker 
than the standard

[[Page 385]]

that contains the lead equivalent of the heavy metals limit of the test.



Sec. 436.209  Melting range or temperature.

    (a) Apparatus. Melting range apparatus consists of a glass container 
for a bath of colorless fluid, a suitable stirring device, an accurate 
thermometer, and a controlled source of heat. Any apparatus or method of 
equal accuracy may be used. The accuracy should be checked periodically 
by use of melting point standards, preferably those that melt near the 
expected melting range of the product to be tested. The bath fluid is 
selected with a view to the temperature required, but light paraffin is 
used generally and certain liquid silicones are well adapted to the 
higher temperature ranges. The fluid is deep enough to permit immersion 
of the thermometer to its specified immersion depth so that the bulb is 
still 2 centimeters above the bottom of the bath.
    (b) Sample preparation. If necessary, reduce the sample to a fine 
powder and store it in a desiccator over sulfuric acid for 24 hours. If 
a method for loss on drying is included in the section for the 
antibiotic to be tested, a sample dried by that method may be used.
    (c) Test procedure. Use a capillary glass tube about 10 centimeters 
long and 0.8 to 1.2 millimeters internal diameter with the wall 0.2 to 
0.3 millimeter in thickness. Charge the tube with a sufficient amount of 
the dry power to form a column 2.5 to 3.5 millimeters high from the 
sealed end when packed down as closely as possible by moderate tapping 
on a solid surface. Heat the bath until a temperature 
10 deg.plus-minus1 deg. C. below the expected melting range 
is reached, then introduce the charged tube, and heat at a rate of rise 
of 3 deg.plus-minus0.5 deg. C. per minute until melting is 
completed. The temperature at which the column of the sample is observed 
to collapse definitely against the side of the tube at any point is 
defined as the beginning of melting, and the temperature at which the 
sample becomes liquid throughout is defined as the end of melting or the 
melting point.
[39 FR 18944, May 30, 1974, as amended at 41 FR 24883, June 21, 1976]



Sec. 436.210  Specific rotation.

    (a) Test procedure. The appropriate solvent, test concentration, and 
polarimeter tube length are specified in the section for each antibiotic 
to be tested. Accurately weigh the sample to be tested in a glass-
stoppered volumetric flask, dissolve in the appropriate solvent, and 
dilute to the specified test concentration at 25 deg. C. Maintain the 
solution at 25 deg. C. and transfer to the appropriate polarimeter tube. 
Determine the angular rotation of both solvent and sample solution in a 
suitable polarimeter, using a sodium light source or a white light 
source with a 589.3-millimicron filter. The zero correction is the 
average of the blank readings and is subtracted from the average 
observed rotation of the sample solution if the two figures are of the 
same sign, or is added if they are opposite in sign, to give the 
corrected angular rotation of the sample solution. The determination 
must be completed within one-half hour from the time the solution is 
prepared.
    (b) Calculations. Determine the specific rotation. [], by 
the following formula:
[GRAPHIC] [TIFF OMITTED] TC01AP94.004

where:
a=The corrected angular rotation of the sample solution in degrees at 
temperature t using a light source of a wavelength of x millimicrons;
l=The length of the polarimeter tube in decimeters;
c=The concentration of the solution expressed as number of grams of 
substance in 100 milliliters of solution.



Sec. 436.211  Identity test by infrared spectrophotometry.

    (a) Apparatus--(1) Spectrophotometer. A suitable spectrophotometer 
capable of recording the infrared absorption spectrum in the 2 to 15 
micron range.
    (2) Hydraulic press. A 30-ton hydraulic press with 12-inch square 
platens.
    (b) Sample preparation methods. Use the sample preparation method 
specified in the individual section for each antibiotic.
    (1) Potassium bromide discs. Quantities of materials specified are 
for a 13-millimeter die. Appropriate adjustments

[[Page 386]]

should be made in the quantities of materials when dies of other sizes 
are used. To prepare a 1.0 percent mixture weigh approximately 2 
milligrams of the sample and mix thoroughly with 200 milligrams of dried 
potassium bromide (infrared spectrophotometric quality). For a 0.5 
percent potassium bromide mixture, use 1 milligram of sample. For a 0.25 
percent potassium bromide mixture, use 0.5 milligram of sample. A mortar 
and pestle, a ball mill, or other suitable mixing device may be used. 
Transfer the uniformly milled mixture to the die, evacuate gradually 
while raising the pressure to 3,000 pounds per square inch until 
evacuation is complete, then raise the pressure to 16,000 pounds per 
square inch, and hold that pressure for 2 to 3 minutes. Release the 
pressure, dismantle the die, and recover the potassium bromide disc. 
Mount the disc in a suitable holder and proceed as directed in paragraph 
(c) of this section.
    (2) Mineral oil mull. Weigh approximately 20 milligrams of the 
sample into an agate mortar and add 2 drops of mineral oil. Triturate 
thoroughly with a pestle until a uniform consistency is obtained. Use 
two rock salt plates as an absorption cell. Place a small drop of the 
mull in the center of one of the plates. Gently put the other plate on 
the mull and slowly squeeze the plates together to spread the mull 
uniformly. Clamp the two plates firmly together in a metal holder. 
Examine the assembled cell by holding it up to the light. It should 
appear smooth and free of any air bubbles. Proceed as directed in 
paragraph (c) of this section.
    (3) 1 percent solution. Prepare a 1 percent solution of the sample 
in chloroform and use 1.0 millimeter matched absorption cells. Proceed 
as directed in paragraph (c) of this section.
    (c) Procedure. Place the sample, prepared as directed in paragraph 
(b) of this section, in the spectrophotometer. Determine the infrared 
absorbance spectrum between the wavelengths of 2 to 15 microns. To be 
suitable the spectrum should have a transmittance of between 20 and 70 
percent at most of the wavelengths showing significant absorption. 
Compare the spectrum to that of an authentic sample of the same 
antibiotic prepared in an identical manner. To pass the infrared 
identity test, the absorption spectrum of the sample should compare 
qualitatively with that of the authentic sample.



Sec. 436.212  Disintegration test.

    (a) Apparatus--(1) Basket-rack assembly. The basket-rack assembly 
consists of 6 open-ended glass tubes, each 7.75plus-minus0.25 
centimeters long and having an inside diameter of approximately 21.5 
millimeters and a wall approximately 2 millimeters thick; the tubes are 
held in a vertical position by two plastic plates, each about 9 
centimeters in diameter and 6 millimeters in thickness, with six holes, 
each about 24 millimeters in diameter, equidistant from the center of 
the plate and equally spaced from one another. Attached by screws to the 
undersurface of the lower plate is 10-mesh No. 23 (0.025 inch) W. and M. 
gauge woven stainless steel wire cloth. The glass tubes and the upper 
plastic plate are secured in position at the top by means of a stainless 
steel plate, about 9 centimeters in diameter and 1 millimeter in 
thickness, having six perforations each about 20 millimeters in 
diameter, which coincide with those of the upper plastic plate and the 
upper open ends of the glass tubes. A central shaft about 8 centimeters 
in length, the upper end of which terminates in an eye through which a 
string or wire may be inserted, is attached to the stainless steel 
plate. The parts of the apparatus are assembled and rigidly held by 
means of three bolts passing through the two plastic plates and the 
steel plate. The design of the basket-rack assembly may be varied 
somewhat provided the specifications for the glass tubes and the screen 
mesh size are maintained.
    (2) Disks. Each tube is provided with a slotted and perforated 
cylindrical disk 9.5plus-minus0.15 millimeters thick and 
20.7plus-minus0.15 millimeters in diameter. The disk is made 
of a suitable, transparent plastic material having a specific gravity of 
between 1.18 and 1.20. Five 2-millimeter holes extend between the ends 
of the cylinder, one of the holes being through the cylinder axis and 
the others parallel with it and equally spaced on a 6-millimeter radius 
from it.

[[Page 387]]

Equally spaced on the sides of the cylinder are four notches that form 
V-shaped planes perpendicular to the ends of the cylinder. The 
dimensions of each notch are such that the openings on the bottom of the 
cylinder are 1.60 millimeters square and those on the top are 9.5 
millimeters wide and 2.55 millimeters deep. All surfaces of the disk are 
smooth.
    (3) Raising and lowering device. Use a device for raising and 
lowering the basket in the immersion fluid at a constant rate between 28 
and 32 cycles per minute through a distance of not less than 5 
centimeters and not more than 6 centimeters.
    (b) Immersion fluids. During the performance of the tests all 
immersion fluids are maintained at a temperature of 
37 deg.plus-minus2 deg. C. by using a thermostatically 
controlled water bath.
    (1) Distilled water.
    (2) Simulated gastric fluid: Dissolve 2.0 grams of sodium chloride 
and 7.0 milliliters of hydrochloric acid in about 500 milliliters of 
water. Dissolve 3.2 grams of pepsin in this solution and add sufficient 
water to make 1,000 milliliters. This solution has a pH of about 1.2.
    (3) Simulated intestinal fluid: Dissolve 6.8 grams of monobasic 
potassium phosphate in 250 milliliters of water, mix and add 190 
milliliters of 0.2N sodium hydroxide and 400 milliliters of water. Add 
10.0 grams of pancreatin, mix, and adjust the resulting solution with 
0.2N sodium hydroxide to a pH of 7.5plus-minus0.1. Dilute to 
1,000 milliliters.
    (c) Immersion vessel. Use a suitable vessel, such as a 1-liter 
beaker.
    (d) Operation. Add enough immersion fluid to the immersion vessel so 
that when the basket-rack assembly is placed on the raising and lowering 
device at the highest point of the upward stroke, the wire mesh remains 
at least 2.5 centimeters below the surface of the fluid and descends to 
not less than 2.5 centimeters from the bottom of the immersion vessel.
    (e) Procedure--(1) Uncoated or filmcoated tablets. Place one tablet 
into each of the six tubes of the basket, add a disk to each tube, and 
operate the apparatus, using simulated gastric fluid as the immersion 
fluid. At the end of the time limit specified in the individual section 
for the particular antibiotic tablet being tested, lift the basket from 
the fluid and observe the tablets.
    (2) Plain-coated tablets. Place one tablet in each of the six tubes 
of the basket, add a disk to each tube, and operate the apparatus, using 
simulated gastric fluids as the immersion fluid. After 30 minutes, lift 
the basket from the fluid and observe the tablets. If the tablets have 
not disintegrated completely, substitute simulated intestinal fluid as 
the immersion fluid and continue the test for a total period of time 
(including previous immersion in simulated gastric fluid) equal to the 
time limit specified in the individual section for the particular 
antibiotic tablet being tested. Lift the basket and observe the tablets.
    (3) Enteric-coated tablets. Place one tablet in each of the six 
tubes of the basket and operate the apparatus, using simulated gastric 
fluid as the immersion fluid. One hour later, lift the basket from the 
fluid and observe the tablets. If the tablets show no distinct evidence 
of dissolution or disintegration, add a disk to each tube and operate 
the apparatus, using simulated intestinal fluid as the immersion fluid, 
for a total period of time (including the previous immersion in 
simulated gastric fluid) equal to the time limit specified in the 
individual section for the particular antibiotic tablet being tested. 
Lift the basket and observe the tablets.
    (4) Pastilles. Place one pastille into each of the six tubes of the 
basket, add a disk to each tube, and operate the apparatus, using 
distilled water as the immersion fluid. At the end of the time limit 
specified in the individual section for the particular antibiotic 
pastille being tested, lift the basket from the fluid and observe the 
pastilles.
    (5) Capsules. Place one capsule into each of the six tubes of the 
basket, add a disk to each tube, and operate the apparatus, using 
distilled water as the immersion fluid. At the end of the time limit 
specified in the individual section for the capsules being tested, lift 
the basket from the fluid and observe the capsules.
    (f) Evaluation. Complete disintegration is defined as the state in 
which any residue of the tablet, pastille, or

[[Page 388]]

capsule (except fragments of the insoluble coating) remaining on the 
screen is a soft mass having no palpably firm core. The tablets, 
pastilles, or capsules pass the disintegration test if all of the units 
tested disintegrate completely under the conditions and time specified 
in the individual section for the antibiotic tablet, pastille, or 
capsule being tested. If one or two tablets, pastilles, or capsules fail 
to disintegrate completely, repeat the test on 12 additional tablets, 
pastilles, or capsules. The tablets, pastilles, or capsules pass the 
disintegration test if not less than 16 of the total 18 tested 
disintegrate completely. Enteric coated tablets fail the disintegration 
test if they show any distinct evidence of dissolution or disintegration 
after 1 hour immersion in simulated gastric fluid.
[39 FR 18944, May 30, 1974, as amended at 52 FR 4617, Feb. 13, 1987; 55 
FR 19873, May 14, 1990]



Sec. 436.213  Nonaqueous titrations.

    (a) Equipment--(1) Apparatus. Use a closed system consisting of a 
suitable titrimeter equipped with a potentiometer, an automatic burette, 
a chart recorder, and a glass calomel combinatin electrode (with 
saturated methanolic potassium chloride as the electrolyte).
    (2) Titration vessel. Use a 100-milliliter tall form beaker without 
a spout.
    (b) Reagents--(1) Methyl alcohol, reagent grade, anhydrous.
    (2) Dimethylsulfoxide, A.C.S., reagent grade.
    (3) Glacial acetic acid, A.C.S., reagent grade.
    (4) Lithium methoxide reagent: 0.02N lithium methoxide in methyl 
alcohol, standardized against primary grade benzoic acid.
    (5) Perchloric acid reagent: 0.02N perchloric acid in glacial acetic 
acid, standardized against primary grade potassium acid phthalate.
    (c) Preparation of sample solutions. Select the weight of the sample 
and the solvent listed for each antibiotic. Transfer the accurately 
weighed sample to a titration vessel. Add the appropriate solvent, 
cover, and stir magnetically until the sample is dissolved. Proceed as 
directed in paragraph (e) of this section, using the procedure or 
procedures specified in the individual section for each antibiotic.

                                                                        
------------------------------------------------------------------------
                                       Weight in                        
             Antibiotic               milligrams          Solvent       
                                       of sample                        
------------------------------------------------------------------------
Amoxicillin-acid titration..........         100  20 milliliters        
                                                   dimethylsulfoxide and
                                                   30 milliliters methyl
                                                   alcohol.*            
Amoxicillin-base titration..........         100  50 milliliters glacial
                                                   acetic acid.         
Ampicillin-acid titration...........         100  20 milliliters        
                                                   dimethylsulfoxide and
                                                   30 milliliters methyl
                                                   alcohol.*            
Ampicillin-base titration...........         100  50 milliliters glacial
                                                   acetic acid.         
Ampicillin sodium-base titration....          50      Do.               
Cephaloglycin-base titration........          50      Do.               
Cephapirin sodium-base titration....          50  50 milliliters glacial
                                                   acetic acid.         
Cyclacillin-acid titration..........         100  20 milliliters        
                                                   dimethylsulfoxide and
                                                   30 milliliters methyl
                                                   alcohol.*            
Cyclacillin-base titration..........         100  50 milliliters glacial
                                                   acetic acid.         
Tobramycin-base titration...........          30  50 ml glacial acetic  
                                                   acid.                
------------------------------------------------------------------------
*The methyl alcohol is added after the sample has dissolved in          
  dimethylsulfoxide.                                                    

    (d) Blank determination. Place the same volume of solvent used to 
prepare the sample solution into a titration vessel and proceed as 
directed in paragraph (e) of this section, using the procedure or 
procedures specified in the individual section for each antibiotic.
    (e) Titration procedures--(1) Acid titration. Equilibrate the 
electrode by soaking it overnight in the solvent used for preparing the 
sample solution. Start the magnetic stirrer and titrate the sample 
solution with the lithium methoxide reagent. Record the change in 
potential of the solution with the addition of the titrant. Determine 
the number of milliliters of reagent consumed at neutralization (the 
inflection point of the titration curve). Calculate the antibiotic 
content as directed in the individual section.
    (2) Base titration. Proceed as directed in paragraph (e)(1) of this 
section, except use the perchloric acid reagent as the titrant and 
calculate the antibiotic

[[Page 389]]

content as directed in the individual section.
[39 FR 18944, May 30, 1974, as amended at 40 FR 22251, Apr. 22, 1975; 40 
FR 23725, June 2, 1975; 40 FR 57797, Dec. 12, 1975; 46 FR 2981, Jan. 13, 
1981]



Sec. 436.214  Heat stability.

    Store an accurately weighed portion of the sample of approximately 
30 milligrams in an unstoppered 50-milliliter Erlenmeyer flask for 4 
days in an electric oven at 100 deg. Cplus-minus1 deg. C. At 
the end of this period, remove the flask from the oven and allow to cool 
in a desiccator. Accurately weigh an unheated portion of the original 
sample of approximately 30 milligrams. Assay both the heated and 
unheated samples for potency as directed in Sec. 436.204 or Sec. 436.205 
of this chapter. Determine the percent loss from the difference in 
potency between the unheated original sample and the heat-treated 
sample.
[42 FR 59856, Nov. 22, 1977]



Sec. 436.215  Dissolution test.

    (a) Equipment. Use either Apparatus 1 or 2 as described in the 
United States Pharmacopeia XXI dissolution test.
    (b) Procedure. For each dosage form listed in the table in this 
paragraph select the appropriate dissolution medium, rotation rate, 
sampling time, and apparatus, and proceed as set forth in either 
Apparatus 1 or 2 methodology of the United States Pharmacopeia XXI 
dissolution test. Determine the amount of drug substance dissolved by 
performing the assay described in paragraph (c) of this section. The 
amount of dissolution medium removed for sampling purposes may be 
disregarded if the amount removed is not more than 15 milliliters. If 
more than 15 milliliters is removed, then correct for the volume 
removed.

                                                                                                                
----------------------------------------------------------------------------------------------------------------
                                                                   Rotation                                     
              Dosage form                   Dissolution medium     rate \1\      Sampling time(s)      Apparatus
----------------------------------------------------------------------------------------------------------------
Amoxicillin trihydrate and clavulanate  900 mL distilled water...        75  30 min..................          2
 potassium chewable tablets..                                                                                   
Amoxicillin trihydrate and clavulanate  ......do.................        75  ......do................          2
 potassium tablets.                                                                                             
Azithromycin capsules.................  900 mL 0.10 M sodium            100  45 min..................          2
                                         phosphate buffer, pH                                                   
                                         6.0, 0.1 mg/mL trypsin.                                                
Bacampicillin hydrochloride tablets...  ......do.................        75  ......do................          2
Cefadroxil hemihydrate capsules.......  900 mL distilled water...       100  45 min..................          1
Cefadroxil hemihydrate tablets........  900 mL distilled water...        50  30 min..................          2
Cefixime tablets......................  900 mL 0.05 M potassium         100  45 min..................          1
                                         phosphate buffer, pH 7.2.                                              
Cefpodoxime proxetil tablets..........  900 mL pH 3.0 glycine            75  30 min..................          2
                                         buffer.                                                                
Cefprozil tablets.....................  900 mL purified water....       100  45 min..................          1
Cefuroxime axetil for oral suspension.  900 mL Sorenson's                50  30 min..................          2
                                         Modified Phosphate                                                     
                                         Buffer, pH 7.0.                                                        
Cefuroxime axetil tablets.............  900 mL 0.07N hydrochloric        55  15 min. and 45 min......          2
                                         acid.                                                                  
Cephalexin hydrochloride monohydrate    900 mL distilled water...       150  45 min..................          1
 tablets..                                                                                                      
Cephradine dihydrate capsules.........  900 mL 0.12N hydrochloric        75  60 min..................          2
                                         acid.                                                                  
Clarithromycin tablets................  900 mL 0.10 M sodium             50  30 min..................          2
                                         acetate buffer, pH 5.0.                                                
Doxycycline hyclate tablets...........  900 mL distilled water...        75  60 min and 90 min.......          2
Doxycycline monohydrate hydrochloric    900 mL 0.1N hydrochloric         75  60 min..................          2
 acid capsules..                         acid..                                                                 
Erythromycin particles in tablets.....  900 mL 0.05M potassium           75  45 min..................          2
                                         phosphate buffer, pH 6.8.                                              
Loracarbef capsules...................  900 mL distilled water...        50  30 min..................          2
Oxytetracycline hydrochloride           900 mL distilled water...        75  30 min and 60 min.......          2
 capsules..                                                                                                     
Rifabutin capsules....................  900 mL 0.01 N                   100  45 min..................          1
                                         hydrochloric acid.                                                     
Tetracycline hydrochloride capsules     ......do.................        75  ......do................          2
 (except 500-mg)..                                                                                              
Tetracycline hydrochloride capsules     ......do.................        75  30 min, 60 min, and 90            2
 (500-mg)..                                                                   min.                              
Tetracycline hydrochloride tablets....  ......do.................        75  30 min and 60 min.......          2
Vancomycin hydrochloride capsules.....  900 mL distilled water...       100  45 min..................         1 
----------------------------------------------------------------------------------------------------------------
\1\ Rotation rate of basket or paddle stirring element (revolutions per minute).                                

[[Page 390]]

                                                                                                                

    (c) Antibiotic drug content--(1) Tetracycline hydrochloride--(i) 
Preparation of working standard solution. Accurately weigh 20 to 30 
milligrams of tetracycline hydrochloride working standard into a 
suitable-sized volumetric flask. Dissolve and dilute to volume with 
water. Further dilute an accurately measured portion with distilled 
water to obtain a known concentration of 0.01 to 0.02 milligram of 
tetracycline hydrochloride per milliliter.
    (ii) Preparation of sample solutions. Dilute an accurately measured 
portion of the sample with sufficient distilled water to obtain a 
concentration of 0.01 to 0.02 milligram of tetracycline hydrochloride 
per milliliter (estimated).
    (iii) Procedure. Using a suitable spectrophotometer and water as the 
blank, determine the absorbance of each standard and sample solution at 
the absorbance peak at approximately 276 nanometers. Determine the exact 
position of the absorption peak for the particular instrument used.
    (iv) Calculation. Determine the total amount of tetracycline 
hydrochloride dissolved as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.014

where:

    T=Total milligrams of drug dissolved;
    Au=Absorbance of sample;
    c=Concentration of standard in milligrams;
    d=Dilution factor of sample filtrate;
    As=Absorbance of standard.

    *If more than 15 mL of dissolution medium is removed, correct for 
the volume removed.

    (2) Oxytetracycline hydrochloride; preparation of working standard-
solution. (i) Accurately weigh 30 milligrams of oxytetracycline-base 
working standard into a suitable-sized volumetric flask. Add 5 
milliliters of 0.1N hydrochloric acid and swirl the flask to dissolve 
oxytetracycline base. Dilute an accurately measured portion with 
distilled water to obtain a known concentration of 0.01 to 0.02 
milligram of oxytetracycline per milliliter.
    (ii) Proceed as directed in paragraphs (c)(1) (ii), (iii), and (iv) 
of this section except measure the absorbance at the absorption peak at 
approximately 273 nanometers.
    (3) Doxycycline hyclate. Proceed as directed in paragraph (c)(1) of 
this section, except use the doxycycline working standard.
    (4) Bacampicillin hydrochloride. Use the ampicillin working standard 
as the standard of comparison and assay for ampicillin content by either 
of the following methods.
    (i) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except dilute the working standard to a final concentration of 
0.3 milligram of ampicillin per milliliter and use the sample solution 
as it is removed from the dissolution vessel without further dilution.
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, except:
    (a) Buffer. In lieu of the buffer described in 
Sec. 442.40(b)(1)(ii)(b)(2) of this chapter, use the buffer prepared as 
follows: Dissolve 200 grams of primary standard tris (hydroxymethyl) 
aminomethane in sufficient distilled water to make 1 liter. Filter 
before use.
    (b) Preparation of the working standard solution. Dissolve and 
dilute an accurately weighed portion of the ampicillin working standard 
with sufficient distilled water to obtain a final concentration of 0.3 
milligram of ampicillin per milliliter;
    (c) Sample solution. Use the sample solution as it is removed from 
the dissolution vessel without further dilution; and
    (d) Calculations. Determine the total amount of ampicillin dissolved 
as follows:

[GRAPHIC] [TIFF OMITTED] TR01JA93.409

where:

    T=Total milligrams of ampicillin equivalent dissolved;
    Au=Absorbance of sample;
    c=Concentration of working standard solution in milligrams per 
milliliter;
    d=Dilution factor of sample filtrate;
    As=Absorbance of standard.

    *If more than 15 mL of dissolution medium is removed, correct for 
the volume removed.

    (5) Cephradine dihydrate--(i) Preparation of working standard 
solution. Accurately weigh approximately 40 milligrams of cephradine 
working standard

[[Page 391]]

into a suitable-sized volumetric flask. Dissolve and dilute to volume 
with 0.12N hydrochloric acid. Further dilute with a buffer solution 
(prepared by dissolving 27.2 grams of sodium acetate trihydrate in a 
mixture of 12 milliliters of glacial acetic acid and sufficient 
distilled water to make 2 liters) to obtain a known concentration of 
0.01 to 0.03 milligram of cephradine per milliliter.
    (ii) Preparation of sample solution. Filter the sample and dilute an 
accurately measured portion of the filtrate with sufficient buffer 
solution, described in paragraph (c)(5)(i) of this section, to obtain a 
concentration of 0.01 to 0.03 milligram of cephradine per milliliter 
(estimated).
    (iii) Proceed as directed in paragraphs (c)(1) (iii) and (iv) of 
this section, except measure the absorbance at the absorption peak at 
approximately 262 nanometers.
    (6) Amoxicillin trihydrate. Assay for the amoxicillin content as 
described in Sec. 440.103d of this chapter, except use the sample as it 
is removed from the dissolution vessel.
    (7) Vancomycin hydrochloride. Assay for the vancomycin content as 
described in Sec. 436.105 of this chapter, except use the sample as it 
is removed from the dissolution test.
    (8) Erythromycin--(i) Preparation of working standard solution. 
Accurately weigh approximately 140 milligrams of erythromycin working 
standard into a 250-milliliter volumetric flask and dissolve in 10 
milliliters of methyl alcohol. Add water nearly to volume, mix, and 
allow the solution to cool. Dilute to volume with water and mix. On the 
day of use, dilute an accurately measured aliquot with water to obtain a 
known concentration of 0.28 milligram of erythromycin per milliliter 
(before adjusting for standard potency).
    (ii) Preparation of sample solution. Dilute an accurately measured 
portion of the filtered sample with sufficient 0.05M potassium phosphate 
buffer, pH 6.8, to obtain a concentration of about 0.28 milligram of 
erythromycin per milliliter (estimated).
    (iii) Procedure. Transfer 5.0-milliliter aliquots of the working 
standard solution and sample solution to 25-milliliter volumetric flasks 
and treat as follows: Add 2.0 milliliters of water, allow to stand for 5 
minutes with intermittent swirling. Add 15.0 milliliters of 0.25N sodium 
hydroxide, dilute to volume with sufficient 0.05M potassium phosphate 
buffer, pH 6.8, and mix. Heat to 60  deg.C for 5 minutes and allow to 
cool. Using a suitable spectrophotometer and a blank (prepared as per 
the procedure above except that 2.0 milliliters of 0.5N sulfuric acid is 
substituted for the 2.0 milliliters of water) for each solution, 
determine the absorbance of each working standard and sample solution at 
the absorbance peak at approximately 236 nanometers. Determine the exact 
position of the absorption peak for the particular instrument used.
    (iv) Calculation. Proceed as directed in paragraph (c)(1)(iv) of 
this section.
    (9) Cefuroxime axetil tablets and powder for oral suspension--(i) 
Preparation of working standard solution--(a) Cefuroxime axetil tablets. 
Accurately weigh approximately 60 milligrams of cefuroxime axetil 
working standard into a suitable-sized volumetric flask. Dissolve in 5 
milliliters of methanol and dilute to volume with 0.07N hydrochloric 
acid to obtain a known concentration equivalent to 0.01 to 0.02 
milligram of cefuroxime activity per milliliter.
    (b) Cefuroxime axetil for oral suspension. Accurately weigh 
approximately 15 milligrams of cefuroxime axetil working standard into a 
100-milliliter volumetric flask. Dissolve in 5 milliliters of methanol 
and dilute to volume with Sorenson's Modified Phosphate Buffer, pH 7.0 
(4.2 grams of sodium dihydrogen orthophosphate dihydrate and 14.3 grams 
of hydrogen disodium orthophosphate dodecahydrate per liter of water).
    (ii) Preparation of sample solution--(a) Cefuroxime axetil tablets. 
Filter through a 0.45-micron filter and dilute an accurately measured 
portion of the filtrate with sufficient 0.07N hydrochloric acid to 
obtain a concentration equivalent to 0.01 to 0.02 milligram of 
cefuroxime activity per milliliter (estimated).
    (b) Cefuroxime axetil for oral suspension. Filter the sample through 
an 8-micron filter. A coarse prefilter may be used to prevent clogging. 
Use the filtrate solution without further dilution.

[[Page 392]]

    (iii) Procedure--(a) Cefuroxime axetil tablets. Using a suitable 
spectrophotometer and 0.07N hydrochloric acid as the blank, determine 
the absorbance of each standard and sample solution at the absorbance 
peak at approximately 280 nanometers. Determine the exact position of 
the absorption peak for the particular instrument used.
    (b) Cefuroxime axetil for oral suspension. Using a suitable 
spectrophotometer and Sorenson's Modified Phosphate Buffer, pH 7.0 (4.2 
grams of sodium dihydrogen orthophosphate dihydrate and 14.3 grams of 
hydrogen disodium orthophosphate dodecahydrate per liter of water) as 
the blank, determine the absorbance of each standard and sample solution 
at the absorbance peak at approximately 280 nanometers. Determine the 
exact position of the absorption peak for the particular instrument 
used.
    (iv) Calculations. Determine the total amount of cefuroxime activity 
dissolved as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.015

where:
T = Total milligrams of cefuroxime activity dissolved;
AU = Absorbance of sample;
c = Cefuroxime activity of working standard solution in milligrams per 
          milliliter;
d = Dilution factor of sample filtrate; and
As = Absorbance of standard.

    (10) Cefixime--(i) Preparation of working standard solution. 
Accurately weigh approximately 25 milligrams of cefixime working 
standard into a 500-milliliter volumetic flask. Wet the powder with 0.5 
milliliters of methanol, and dilute to volume with 0.05 M potassium 
phosphate buffer, pH 7.2 (prepared by dissolving 6.8 grams of monobasic 
potassium phosphate in distilled water to a volume of one liter. The pH 
is adjusted to 7.2 with 1.0N NaOH). Sonicate to assure dissolution and 
mix.
    (ii) Preparation of sample solution. Forty-five minutes after the 
beginning of the rotation, withdraw and filter a portion of the 
solution. For the 400-milligram tablets, pipet 10.0 milliliters of the 
filtered sample solution into a 100-milliliter volumetric flask. For the 
200-milligram tablets, pipet 10.0 milliliters of the filtered sample 
into a 50-milliliter volumetric flask. Dilute to volume with 0.05 M 
postassium phosphate buffer, pH 7.2.
    (iii) Procedure. Proceed as directed in paragraphs (c)(1) (iii) and 
(iv) of this section, except measure the absorbance of the peak at 
approximately 320 nanometers using 0.05 M potassium phosphate buffer, pH 
7.2 as the blank.
    (11) Cephalexin hydrochloride monohydrate. Assay for cephalexin 
activity of the cephalexin hydrochloride monohydrate as directed in 
Sec. 442.28 of this chapter, and use U.S.P. dissolution apparatus 1 (10 
mesh basket). Use the sample as it is removed from the dissolution 
vessel.
    (12) Doxycycline monohydrate. Proceed as directed in paragraph 
(c)(1) of this section, except use the doxycycline standard.
    (13) Clarithromycin. Proceed as directed in Sec. 452.50(b)(1) of 
this chapter except:
    (i) Dissolution medium. Instead of the mobile phase described in 
Sec. 452.50(b)(1)(i) of this chapter, use 0.10 M sodium acetate buffer 
prepared as follows: Weigh 13.6 grams of sodium acetate trihyrate into a 
container sufficient to hold 1 liter of solution. Dissolve the salt in 
750 milliliters of distilled water. Adjust the pH of the solution to 
5.00.05 with glacial acetic acid. Dilute to 1,000 
milliliters with distilled water.
    (ii) Preparation of the standard and sample solutions--(a) Standard 
solution. Dissolve (with shaking or sonication) an accurately weighed 
portion of the clarithromycin working standard, in sufficient methanol 
to obtain a solution having a known concentration of approximately 625 
micrograms per milliliter of clarithromycin. Quantitatively transfer and 
dilute an aliquot of this solution with mobile phase (described in 
Sec. 452.50(b)(1)(i) of this chapter) and mix to obtain a solution of 
known concentration of approximately 125 micrograms per milliliter of 
clarithromycin.
    (b) Sample solution. Use the sample solution as it is removed from 
the dissolution vessel after diluting and mixing with mobile phase 
(described in Sec. 452.50(b)(1)(i) of this chapter) 1:2 for

[[Page 393]]

the 250-milligram tablet and 1:4 for the 500-milligram tablet.
    (c) Calculations. Determine the total amount of clarithromycin 
activity dissolved as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.016

where:
T = Total milligrams of clarithromycin activity dissolved;
AU = Area of the clarithromycin peak (at a retention time 
          equal to that observed for the standard) in the chromatogram 
          of the sample;
AS = Area of the clarithromycin peak in the chromatogram of 
          the clarithromycin standard;
c = Clarithromycin activity in the clarithromycin working standard 
          solution in milligrams per milliliter; and
d = Dilution factor of sample filtrate.
    (14) Azithromycin. Proceed as directed in Sec. 452.60(b)(1) of this 
chapter, except:
    (i) Dissolution medium. Dissolve 85.2 grams of sodium phosphate 
dibasic and dilute to volume with ultrapure deionized or high-
performance liquid chromatographic-grade water in a stoppered 2-liter 
graduated cylinder. Dilute this entire solution in an appropriate, 
suitably sized container with 4 liters of ultrapure deionized or high-
performance liquid chromatographic-grade water. Adjust the pH to 
6.00.05 with concentrated hydrochloric acid (about 40.5 
milliliters). Add 600 milligrams of trypsin and mix well.
    (ii) Preparation of the standard and sample solutions--(a) Standard 
solution. Accurately weigh approximately 15 milligrams of the 
azithromycin working standard into a 50-milliliter volumetric flask. Add 
25 milliliters of the dissolution medium and sonicate briefly. Dilute to 
volume with dissolution medium and mix well. Pipet 2.0 milliliters of 
this solution into a 25-milliliter volumetric flask and dilute to volume 
with the mobile phase described in Sec. 452.60(b)(1)(i) of this chapter. 
Pipet 4.0 milliliters of this solution into a 25-milliliter volumetric 
flask and bring to volume with the mobile phase.
    (b) Sample solution. Filter the sample solutions through a 0.45-
micron filter before use. Pipet 2.0 milliliters of the filtered aliquot 
into a 25-milliliter volumetric flask and dilute to volume with the 
mobile phase described in Sec. 452.60(b)(1)(i) of this chapter. Pipet 
4.0 milliliters of this solution into another 25-milliliter volumetric 
flask and bring to volume with the mobile phase. The solution is stable 
at room temperature for 24 hours.
    (c) Calculations. Determine the percent of azithromycin dissolved as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.017

where:
AU =Area of the azithromycin peak (at a retention time equal 
          to that observed for the standard) in the chromatogram of the 
          sample;
AS =Area of the azithromycin peak in the chromatogram of the 
          azithromycin standard;
PS =Azithromycin activity in the azithromycin working 
          standard solution in micrograms per milliliter;
          [GRAPHIC] [TIFF OMITTED] TR01JA93.018
          
    1 If more than 15 milliliters of dissolution medium are 
removed, correct for the volume removed; and
WU =Theoretical azithromycin content (mg) of capsule.

    (15) Cefprozil. Proceed as directed in Sec. 442.80(b)(1) of this 
chapter except:
    (i) Sample solutions. Filter the sample solutions through a 0.45-
micron filter before use. Use the sample solution as it is removed from 
the dissolution vessel without further dilution for the 250-milligram 
tablet; prepare the sample solution for the 500-milligram tablet by 
diluting a 5-milliliter aliquot of the filtered solution to volume in a 
10-milliliter volumetric flask with distilled water.
    (ii) Calculations. Determine the total percent of cefprozil 
dissolved as follows:

[[Page 394]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.019


[GRAPHIC] [TIFF OMITTED] TR01JA93.020

where:
AU = Area of the cefprozil (Z) or cefprozil (E) response in 
          the chromatogram of the sample (at a retention time equal to 
          that observed for the standard);
AS = Area of the cefprozil (Z) or cefprozil (E) response in 
          the chromatogram of the cefprozil (Z) or cefprozil (E) 
          standard;
c = Concentration of the cefprozil (Z) or cefprozil (E) working standard 
          solution in milligrams per milliliter; and
d = Dilution factor of the sample filtrate.

    (16) Loracarbef--(i) Preparation of the working standard solution. 
Accurately weigh approximately 110 milligrams of the loracarbef working 
standard into a suitable-sized volumetric flask. Dissolve and dilute to 
volume with water. Further dilute an accurately measured portion with 
distilled water to obtain a known concentration of 0.02 milligram of 
loracarbef activity per milliliter.
    (ii) Preparation of sample solutions. Dilute an accurately measured 
portion of the sample with sufficient distilled water to obtain a 
concentration of 0.02 milligram of loracarbef activity per milliliter 
(estimated).
    (iii) Procedure. Using a suitable spectrophotometer and water as the 
blank, determine the absorbance of each standard and sample solution at 
the absorbance maximum at approximately 260 nanometers. Determine the 
exact position of the absorbance maximum for the particular instrument 
used.
    (iv) Calculations. Determine the total amount of loracarbef 
dissolved as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.021

where:
T = Total milligrams of loracarbef activity dissolved;
AU = Absorbance of sample;
AS = Absorbance of the standard;
c = Concentration of the working standard solution in milligrams per 
          milliliter; and
d = Dilution factor of the sample filtrate.

    (17) Cefadroxil hemihydrate. Proceed as directed in paragraph (c)(1) 
of this section, except use the cefadroxil working standard and measure 
the absorbance at the absorption peak of approximately 264 nanometers.
    (18) Rifabutin--(i) Preparation of the working standard solution. 
Accurately weigh approximately 45 milligrams of the rifabutin working 
standard into a suitable-sized volumetric flask. Dissolve and dilute to 
volume with 0.01N hydrochloric acid (prepared by diluting 5.0 
milliliters of hydrochloric acid (37 percent) to 6 liters with distilled 
water) to obtain a concentration of approximately 13 micrograms 
rifabutin activity per milliliter.
    (ii) Preparation of sample solutions. Forty-five minutes after the 
beginning of the rotation, withdraw a 10-milliliter aliquot from the 
vessel. Dilute a 2-milliliter portion of the sample to 25 milliliters 
with 0.01N hydrochloric acid.
    (iii) Procedure. Using a suitable spectrophotometer and 0.01N 
hydrochloric acid as the blank, determine the absorbance of each 
standard and sample solution at the absorbance maximum at approximately 
280 nanometers. Determine the exact position of the absorbance maximum 
for the particular instrument used.
    (iv) Calculations. Determine the total amount of rifabutin dissolved 
as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.022

where:
T = Total milligrams of rifabutin activity dissolved;
AU = Absorbance of sample;
AS = Absorbance of the standard;
c = Rifabutin activity of the working standard solution in micrograms 
          per milliliter; and
d = Dilution factor of the sample filtrate.


[[Page 395]]


    (19) Cefpodoxime proxetil--(i) Dissolution fluid: 0.04 molar glycine 
buffer, pH 3.0--(A) Stock solution. Dissolve 54.5 grams of glycine 
(aminoacetic acid) and 42.6 grams of sodium chloride in about 500 
milliliters of deionized water in a 1-liter volumetric flask. Add 
cautiously, and with swirling, 14.2 milliliters of concentrated 
hydrochloric acid. Cool to room temperature. Dilute to volume with 
deionized water and mix. Check the pH of the solution obtained by 
diluting 50 milliliters of the stock solution to 900 milliliters with 
deionized water. The pH should be 3.00.1. If necessary, 
adjust the pH of the stock solution with 50 percent sodium hydroxide or 
concentrated hydrochloric acid. Recheck that the pH of the working 
solution is 3.00.1.
    (B) Working solution. Dilute 50 milliliters of stock solution to 900 
milliliters with deionized water.
    (ii) Preparation of the working standard solutions. Accurately weigh 
approximately 28 milligrams for the 100-milligram tablets and 56 
milligrams for the 200-milligram tablets of the cefpodoxime proxetil 
working standard and dissolve in 10 milliliters of methanol. Dilute to 
200 milliliters with dissolution fluid. Prepare fresh daily.
    (iii) Sample solutions. Filter the sample solutions through a 0.45-
micron filter before use. Use the sample solution as it is removed from 
the dissolution vessel without further dilution.
    (iv) Procedure. Using a suitable spectrophotometer and water as the 
blank, determine the absorbance of each standard and sample solution at 
the absorbance peak at approximately 259 nanometers. Determine the exact 
position of the absorption peak for the particular instrument used.
    (v) Calculations. Determine the percent of label dissolved as 
follows:
    Percent dissolved = (Asam/Astd) X 
(Cs/L) X V X P X F1

where:
Asam = Absorbance of the sample at 259 nanometers;
Astd = Absorbance of the working standard solution at 259 
          nanometers;
Cs = Concentration of the working standard preparation in 
          milligrams per milliliter;
L = Tablet strength, in milligrams per tablet;
P = Purity of the reference standard in percent;
V = Volume of dissolution fluid used in milliliters (900); and
F1 = 0.7666 (conversion factor to free acid equivalents).

    (d) Evaluation. Use the dissolution acceptance table and 
interpretation in the United States Pharmacopeia XXI.
[44 FR 48188, Aug. 17, 1979]

    Editorial Note: For Federal Register citations affecting 
Sec. 430.215, see the List of CFR Sections Affected appearing in the 
Finding Aids section of this volume.



Sec. 436.216  High-performance liquid chromatographic assay.

    (a) Equipment. A suitable high-performance liquid chromatograph 
equipped with:
    (1) A suitable detection system specified in the monograph for the 
drug being tested;
    (2) A suitable recording device of at least 25-centimeter 
deflection;
    (3) A suitable chromatographic data managing system; and
    (4) An analytical column, 3 to 30 centimeters long, packed with a 
material as defined in the monograph for the drug being tested; and if 
specified in that monograph, the inlet of this column may be connected 
to a guard column, 3 to 5 centimeters in length, packed with the same 
material of 40 to 60 micrometers particle size.
    (b) Procedure. Perform the assay and calculate the drug content 
using the temperature, instrumental conditions, flow rate, and 
calculations specified in the monograph for the drug being tested. Use a 
detector sensitivity setting that gives a peak height for the working 
standard solution that is at least 50 percent of scale with typical 
chart speed of not less than 2.5 millimeters per minute. Use the 
equipment described in paragraph (a) of this section. Use the reagents, 
working standard solution, and sample solution described in the 
monograph for the drug being tested. Equilibrate and condition the 
column by passage of 10 to 15 void volumes of mobile phase followed by 
five replicate injections of the same volume of the working standard 
solution. Allow an operating time sufficiently long to obtain 
satisfactory separation and elution of the expected components after 
each injection. Record the peak responses and calculate the prescribed

[[Page 396]]

system suitability requirements described for the system suitability 
test in paragraph (c) of this section.
    (c) System suitability test. Select the system suitability 
requirements specified in the monograph for the drug being tested. Then, 
using the equipment and procedure described in this section, test the 
chromatographic system for assay as follows:
    (1) Trailing factor or asymmetry factor. Calculate either the 
trailing factor (T), from distances measured along the horizontal line 
at 5 percent of the peak height above the baseline or the asymmetry 
factor (As) measured at a point 10 percent of the peak height 
from the baseline; whichever is required in the appropriate monograph, 
as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.023

where:
W0.05=Width of peak at 5 percent height; and
f=Horizontal distance from point of ascent to a point coincident with 
          maximum peak height.
          [GRAPHIC] [TIFF OMITTED] TR01JA93.024
          
where:
a=Horizontal distance from point of ascent to point of maximum peak 
          height; and
b=Horizontal distance from the point of maximum peak height to point of 
          descent.

    (2) Efficiency of the column. Calculate the number of theoretical 
plates (n) of the column as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.025

where:
n=Efficiency, as number of theoretical plates for column;
tR=Retention time of solute; and
Wh=Peak width at half-height.
Calculate the absolute efficiency of the column, (reduced plate height) 
(hr
[GRAPHIC] [TIFF OMITTED] TR01JA93.026

where:
L=Length of column in centimeters;
n=Number of theoretical plates; and
dp

    (3) Resolution. Calculate the resolution (R) as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.027
    
where:
tRj=Retention time of a solute eluting after i (tRj 
          is larger than tRi)
tRi=Retention time for any solute;
wi=Width of peak at baseline for any solute; and
wj=Width of peak at baseline for any solute eluting after i.

    (4) Coefficient of variation (relative standard deviation). 
Calculate the coefficient of variation (SR in percent) as 
follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.005

where:
X is the mean of N of individual measurements of Xi.


If the complete operating system meets the system suitability 
requirements of the monograph for the drug being tested, proceed as 
described in paragraph (b) of this section, except alternate injections 
of the working standard solution with injections of the sample solution.
    (5) Capacity factor. Calculate the capacity factor (k), if required 
in the monograph as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.028

where:
tr=Retention time of solute; and
tm=Retention time of solvent or unretained substance, 
          calculated as follows:
          [GRAPHIC] [TIFF OMITTED] TR01JA93.029
          
where:
D=Column diameter in centimeters;
L=Column length in centimeters;
0.75=Average total column porosity; and
F=Flow rate in milliliters per minute.
[51 FR 11572, Apr. 4, 1986, as amended at 54 FR 47351, Nov. 14, 1989]

[[Page 397]]



Sec. 436.217  Film-coat rupture test.

    (a) Immersion fluid. Dilute 6.0 milliliters of hydrochloric acid to 
1,000 milliliters with water. During the performance of the test 
maintain the immersion fluid at a temperature of 370.5 
deg.C by using a thermostatically controlled water bath.
    (b) Immersion vessel. Use a suitable vessel, such as a 1-liter 
beaker.
    (c) Operation. Add 750 milliliters of immersion fluid to the 
immersion vessel.
    (d) Procedure. Drop a tablet into the immersion fluid and record the 
time for the tablet coat to rupture. Repeat the test with a further 19 
tablets, testing not more than 10 tablets with a given volume of 
immersion fluid.
    (e) Evaluation. The tablets pass the film-coat rupture test if the 
mean coat rupture time does not exceed 20 seconds and not more than 2 
tablets have a coat rupture time exceeding 40 seconds.
[52 FR 42432, Nov. 5, 1987]



           Subpart F--Chemical Tests for Specific Antibiotics



Sec. 436.300  Polarimetric assay of carbenicillin indanyl sodium.

    (a) Equipment. Polarimeter capable of measuring optical rotatory 
activity at 365 nanometers: Perkin-Elmer Model 141 or equivalent, with a 
suitable 1-decimeter polarimeter tube.
    (b) Reagents--(1) 4-methyl-2-pentanone. Meets ACS specifications.
    (2) Phosphate-citrate buffer. Dissolve 61.0 grams of anhydrous 
disodium phosphate and 11.0 grams of citric acid in 950 milliliters of 
distilled water. Adjust the pH to 6.0 with 6N hydrochloric acid. Dilute 
to 1,000 milliliters with distilled water.
    (c) Preparation of carbenicillin indanyl sodium sample and working 
standard solutions. Accurately weigh approximately 125 milligrams of the 
carbenicillin indanyl sodium sample or working standard into a 25-
milliliter volumetric flask. Dissolve and dilute to volume with 
distilled water. Transfer a 5-milliliter aliquot to a 50-milliliter 
glass-stoppered centrifuge tube. Add 15 milliliters of the phosphate-
citrate buffer and 20 milliliters of 4-methyl-2-pentanone; stopper and 
shake the tube for 10 seconds. Centrifuge at 2,000 revolutions per 
minute for 10 minutes to separate the phases. Remove about 15 
milliliters of the upper (4-methyl-2-pentanone solvent) phase and 
proceed as directed in paragraph (e) of this section.
    (d) Preparation of the blank. Place a 5-milliliter aliquot of 
distilled water into a 50-milliliter glass-stoppered centrifuge tube, 
add 15 milliliters of phosphate-citrate buffer and 20 milliliters of 4-
methyl-2-pentanone; stopper and shake the tube for 10 seconds. 
Centrifuge at 2,000 revolutions per minute for 10 minutes to separate 
the phases. Remove about 15 milliliters of the upper phase and proceed 
as directed in paragraph (e) of this section.
    (e) Procedure. Fill the polarimeter tube with the blank solution 
prepared as described in paragraph (d) of this section. Place the tube 
in the polarimeter. Adjust the polarimeter to zero rotation using a 
light source with a wavelength of 365 nanometers. Use the same procedure 
to determine the optical rotation of both the sample solution and the 
working standard solution prepared as directed in paragraph (c) of this 
section.
    (f) Calculations. Calculate the carbenicillin content (potency) of 
the sample on an anhydrous basis as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.030


[[Page 398]]


where: m=moisture content of the sample.



Sec. 436.301  Thin layer chromatography identity test for carbenicillin indanyl.

    Using the sample solution prepared as described in the section for 
the antibiotic drug to be tested, proceed as described in paragraphs 
(a), (b), (c), and (d) of this section.
    (a) Equipment--(1) Chromatography tank. A rectangular tank, 
approximately 9  x  9  x  3.5 inches lined with Whatman's 3MM 
chromatographic paper (0.3 millimeters) or equivalent.
    (2) Iodine vapor chamber. A rectangular tank approximately 9  x  9 
x  3.5 inches, with a suitable cover, containing iodine crystals.
    (3) Plates. Use 20  x  20 centimeters thin layer chromatography 
plates coated with silica gel G or equivalent to a thickness of 250 
microns.
    (b) Reagents--(1) Extraction solvent. Mix ethyl acetate, acetone, 
pyridine, water, and acetic acid in volumetric proportions of 
100:200:25:75:1.5 respectively.
    (2) Developing solvent. Mix ethyl acetate, acetone, pyridine, water, 
and acetic acid in volumetric proportions of 300:400:25:75:2 
respectively.
    (3) Ferric chloride-potassium ferricyanide reagent. Immediately 
before use, mix 100 milliliters of a 1 percent ferric chloride solution 
in 1 percent hydrochloric acid with 100 milliliters of a 1 percent 
potassium ferricyanide solution and 75 milliliters of methanol.
    (c) Preparation of working standard solution. Weigh an amount of the 
carbenicillin indanyl working standard equivalent to approximately 10 
milligrams of carbenicillin into a 50-milliliter Erlenmeyer flask. 
Dissolve the material in sufficient extraction solvent to make a 
solution containing 1 milligram carbenicillin per milliliter.
    (d) Procedure. Pour developing solvent into the bottom of the 
chromatography tank. Cover and seal the tank. Allow it to equilibrate 
for 1 hour. Prepare a plate as follows: On a line 2 centimeters from the 
base of the silica gel plate, and at intervals of 2 centimeters, spot 10 
microliters of the standard solution and the sample solution. The plate 
should be air dried for 30 minutes. Place the plate into the 
chromatography tank. Allow the solvent front to travel about 15 
centimeters from the starting line and then remove the plate from the 
tank. Heat the plate for 30 minutes at 80 deg. C. in a circulating air 
oven and then allow the plate to cool to room temperature. Place the 
plate in the iodine vapor chamber for about 30 seconds, remove the plate 
and spray it with the ferric chloride-potassium ferricyanide reagent. 
Carbenicillin indanyl appears as a blue spot on a yellow-green 
background at an Rf of about 0.5. The test is satisfactory if 
the sample compares qualitatively with the standard.
[39 FR 18944, May 30, 1974, as amended at 41 FR 18509, May 5, 1976]



Sec. 436.302  Clindamycin vapor phase chromatography.

    (a) Equipment. Gas chromatograph equipped with a flame ionization 
detector: Barber-Colman 5,000 or equivalent.
    (b) Reagents. (1) Pyridine, reagent grade, dried over sodium 
sulfate.
    (2) Chloroform, reagent grade.
    (3) Acetic anhydride, reagent grade, used as acteylating agent.
    (4) Internal standard: Prepare a solution containing 3 milligrams of 
cholestane per milliliter in pyridine.
    (c) Typical conditions. (1) Column: 4 feet  x  4 millimeters ID, 
glass, with 1 percent SE-30 on Diatoport S (60/80 mesh), or equivalent.
    (2) Temperatures: Column 200 deg. C.; detector 215 deg. C.; 
injection port, ambient temperature.
    (3) Carrier gas: Helium approximately 120 milliliters per minute.
    (4) Detector: Hydrogen flame--hydrogen at 120 pounds per square 
inch, air at 40 pounds per square inch.
    (5) Sensitivity: 1,000; attenuation, 2 for clindamycin, 1 for 
internal standard: 2 x 10-8 amperes.
    (d) Preparation of clindamycin sample and working standard 
solutions. Accurately weigh approximately 15 milligrams of sample or 
working standard into a glass-stoppered conical 15-milliliter centrifuge 
tube. Add 1.0 milliliter of chloroform, 1.0 milliliter of internal 
standard solution, and 0.6 milliliter of acetic anhydride. Agitate the 
tubes to insure dissolution of the sample and

[[Page 399]]

complete mixing of the liquids. Proceed as directed in paragraph (e) of 
this section.
    (e) Procedure. Cover the top of each centrifuge tube with a plastic 
cap. Punch a small hole in the top of each cap to allow vapor to escape. 
Place the tubes in a 100 deg. C. drying oven for 2.5 hours. Remove the 
tubes from the oven and allow to cool. Take the plastic cap from each 
tube and replace with the glass stopper. Centrifuge 10-15 minutes at 
2,000-2,500 r.p.m. to separate the white solid from the liquid in the 
tube. Inject 0.5 microliter of the clear liquid into the gas 
chromatograph. Use the conditions and materials listed in paragraphs 
(a), (b), and (c) of this section. The conditions should be adequate to 
maintain a stable baseline and provide at least 60 percent deflection of 
the recorder scale by the clindamycin peak. The resolution of the peaks 
should be complete. The elution order is: Internal standard, 
clindamycin, and epiclindamycin (if present). Calculate the clindamycin 
content as directed in paragraph (f) of this section.
    (f) Calculations. Calculate the clindamycin content of the sample as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.031

where:
Ru=Area of the clindamycin sample peak (at a retention time 
equal to that observed for the clindamycin standard)/Area of internal 
standard peak;
Rs=Area of the clindamycin standard peak/Area of internal 
standard peak;
Ws=Weight of the clindamycin working standard in milligrams;
Wu=Weight of the sample in milligrams;
f=Potency of the clindamycin working standard in micrograms per 
milligram.



Sec. 436.303  Clindamycin content of clindamycin palmitate hydrochloride by vapor phase chromatography.

    (a) Equipment. Gas chromatograph equipped with a flame ionization 
detector: Hewlett-Packard 7606 \4\ or equivalent.
---------------------------------------------------------------------------

    \4\ Available from: Hewlett Packard Co., P.O. Box 301, Loveland, CO 
80537.
---------------------------------------------------------------------------

    (b) Reagents. (1) Acetic anhydride, reagent grade.
    (2) Pyridine, reagent grade.
    (3) Chloroform, reagent grade.
    (4) Internal standard: Prepare a solution containing 5 milligrams of 
cholesteryl benzoate per milliliter in chloroform.
    (c) Typical conditions. (1) Column: 6 feet  x  2 millimeters ID, 
glass, with 1 percent UC-W98 on Chromosorb WHP (80/100 mesh) or 
equivalent.
    (2) Temperatures: Column 275 deg. C.; detector 290 deg. C.; 
injection port 280 deg. C.
    (3) Carrier gas: Helium approximately 60 milliliters per minute.
    (4) Detector: Hydrogen flame ionization--hydrogen at 12 pounds per 
square inch, air at 32 pounds per square inch.
    (5) Sensitivity: 1,000; attenuation, 16; 1  x  10- 9 
amperes.
    (d) Preparation of clindamycin palmitate hydrochloride sample and 
working standard solutions. Accurately weigh approximately 15 milligrams 
of both the sample and the working standard into separate glass-
stoppered, conical 15-milliliter centrifuge tubes. Add 1.0 milliliter of 
internal standard solution, 1.0 milliliter of pyridine, and 0.5 
milliliter of acetic anhydride to each tube. Agitate the tubes to insure 
dissolution and complete mixing of the liquids. Proceed as directed in 
paragraph (e) of this section.
    (e) Procedure. Cover the top of each centrifuge tube with a plastic 
cap. Punch a small hole in the top of each cap to allow vapor to escape. 
Place the tubes in a 100 deg. C. drying oven for 2.5 hours. Remove the 
tubes from the oven and allow to cool. Take the plastic cap from each 
tube and replace with the glass stopper. Centrifuge 10-15 minutes at 
2,000-2,500 r.p.m. to separate the white solid from the liquid in the 
tube. Inject 1 microliter of the clear liquid into the gas 
chromatograph. Use the conditions and materials listed in paragraphs 
(a), (b), and (c) of this section. The conditions should be adequate to 
maintain a stable baseline and provide at least 40 percent deflection of 
the recorder scale by the clindamycin palmitate peak. The resolution of 
the peaks should be complete. The internal standard will be eluted 
before the clindamycin palmitate. Calculate the clindamycin content as 
directed in paragraph (f) of this section.

[[Page 400]]

    (f) Calculations. Calculate the clindamycin content of the sample as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.032

where:
Ru=Area of the sample peak (at a retention time equal to that 
observed for the clindamycin palmitate hydrochloride standard)/Area of 
internal standard peak;
Rs=Area of the clindamycin palmitate hydrochloride standard 
peak/Area of internal standard peak;
Ws=Weight of the clindamycin palmitate hydrochloride working 
standard in milligrams;
Wu=Weight of the sample in milligrams;
f=Micrograms of clindamycin activity per milligram of clindamycin 
palmitate hydrochloride working standard.



Sec. 436.304  Clindamycin phosphate vapor phase chromatography.

    (a) Equipment. Gas chromatograph equipped with an electronic 
integrator and with a flame ionization detector that has a sensitivity 
of at least 1  x  10- 1 0 amperes: Hewlett-
Packard 7600 \4\ or equivalent.
---------------------------------------------------------------------------

    \4\ See footnote 4 to Sec. 436.303(a).
---------------------------------------------------------------------------

    (b) Reagents. (1) Trifluoroacetic anhydride.
    (2) Intestinal alkaline phosphatase.
    (3) pH 9.0 borate buffer: Transfer 3.1 grams of boric acid into a 1-
liter volumetric flask containing 500 milliliters of water, mix, and add 
21 milliliters of 1.0N sodium hydroxide and 10 milliliters of 0.1M 
magnesium chloride. Dilute to volume with water and mix well.
    (4) Internal standard: Prepare a chloroform solution containing 
approximately 0.45 milligram hexacosane per milliliter.
    (5) Anhydrous sodium carbonate.
    (c) Typical conditions. (1) Column: 2 feet  x  3 millimeters ID, 
glass, with 1 percent SE-30 on Diatoport S (80/100 mesh), or equivalent.
    (2) Temperatures: Column, 180 deg. C., detector, 215 deg. C., 
injection port, ambient temperature.
    (3) Carrier gas: Helium approximately 60 milliliters per minute.
    (4) Detector: Hydrogen flame--hydrogen flow at 40 milliliters per 
minute. Air flow at 400 milliliters per minute.
    (5) Sensitivity: 1  x  10- 9 amperes.
    (d) Preparation of clindamycin phosphate sample solution. Accurately 
weigh approximately 12 milligrams of the clindamycin phosphate sample 
into a 50-milliliter glass-stoppered centrifuge tube. Pipet 25 
milliliters of the pH 9.0 borate buffer into the centrifuge tube. Add 10 
milliliters chloroform and shake vigorously for 15 minutes. Centrifuge 
the resulting mixture and pipet a 20-milliliter aliquot of the aqueous 
phase into a 35-milliliter centrifuge tube. Add a weighed amount of 
intestinal alkaline phosphatase equivalent to 50 units of activity 
5 and allow the solution to stand until the enzyme has 
completely dissolved. Place the tube into a water bath at 37 deg. 
C.plus-minus2 deg. C. for 2.5 hours. After the 2.5-hour 
hydrolysis, allow the solution to cool and proceed as directed in 
paragraph (f) of this section.
---------------------------------------------------------------------------

    5 Defined such that 50 units hydrolyzes at least 20 
micromoles of a clindamycin phosphate authentic sample under the assay 
conditions described in this section.
---------------------------------------------------------------------------

    (e) Preparation of the clindamycin hydrochloride standard solution. 
Accurately weigh approximately 9 milligrams of the clindamycin 
hydrochloride working standard into a 35-milliliter glass-stoppered 
centrifuge tube and dissolve in 20 milliliters of pH 9.0 borate buffer. 
Proceed as directed in paragraph (f) of this section.
    (f) Procedure. Add 10 milliliters of the internal standard solution 
to each sample and standard solution. Shake the centrifuge tubes 
vigorously for 30 minutes and centrifuge. Remove the aqueous layer and 
discard. Shake the tubes again; mix in an ultrasonic mixer for 2 
minutes, then centrifuge. No emulsion should be present at this stage. 
Remove the remaining aqueous layer by suction and transfer a 3-
milliliter aliquot of the chloroform layer to a 1-dram tablet vial 
containing approximately 1 gram of anhydrous sodium sulfate. Swirl the 
vial to dry the chloroform and transfer a 1-milliliter aliquot to 
another 1-dram tablet vial. Using a 0.25-milliliter pipet, add 0.25 
milliliter of trifluoracetic anhydride to

[[Page 401]]

each of the vials and place into a water bath at 45 deg. 
C.plus-minus2 deg. C. for 30 minutes. Remove the vials from 
the bath, add about 10 granules of anhydrous sodium carbonate to each 
vial, and allow to stand for approximately 30 minutes. Centrifuge the 
vials for approximately 10 minutes at 5,000 r.p.m. Inject 2 microliters 
of each of the resulting solutions into the gas chromatograph. Use the 
conditions and materials listed in paragraphs (a), (b), and (c) of this 
section. The elution order is: Epiclindamycin (if present), clindamycin 
B (if present), clindamycin, and internal standard. Calculate the 
clindamycin content as directed in paragraph (g) of this section.
    (g) Calculations. Calculate the clindamycin content of the sample as 
follows:

Micrograms of clindamycin per milligram=
[GRAPHIC] [TIFF OMITTED] TC01AP94.006

where:
Ru=Area of the clindamycin sample peak (at a retention time 
equal to that observed for the clindamycin standard)/Area of internal 
standard peak;
Ru=Area of the clindamycin standard peak/Area of internal 
standard peak;
Ws=Weight of the clindamycin working standard in milligrams;
Wu=Weight of the sample in milligrams;
f=Potency of the clindamycin working standard in micrograms per 
milligram.
[39 FR 18944, May 30, 1974, as amended at 41 FR 24704, June 18, 1976]



Sec. 436.305  Thin layer chromatographic identity test for hetacillin.

    (a) Equipment--(1) Chromatography tank. A rectangular tank, 
approximately 9  x  9  x  3.5 inches with a glass solvent trough on the 
bottom.
    (2) Plates. Use 20  x  20 centimeter thin layer chromatography 
plates coated with Silica Gel G or equivalent to a thickness of 250 
microns.
    (b) Developing solvent. Mix 650 milliliters acetone with 100 
milliliters distilled water, 100 milliliters benzene, and 25 milliliters 
acetic acid.
    (c) Spray solution. Dissolve 300 milligrams of ninhydrin in 100 
milliliters of ethanol.
    (d) Preparation of spotting solutions--(1) Sample solution. Use the 
sample solution prepared as described in the section for the particular 
product to be tested.
    (2) Reference solutions. Prepare a solution containing 10 milligrams 
of an authentic hetacillin sample per milliliter in a 4:1 solution of 
acetone and 0.1N hydrochloric acid, and a solution of ampicillin 
standard at 1 mg/ml in the same solvent.
    (e) Procedure. Spot a plate as follows: Apply approximately 10 
microliters of the sample solution, 1  l, of the reference 
hetacillin solution, and 1  l, of the ampicillin reference 
solution on a line 1.5 centimeters from the base of the silica gel plate 
and at intervals of not less than 2.0 centimeters. Pour developing 
solvent into the glass trough in the bottom of the chromatography tank. 
After all spots are thoroughly dry, place the silica gel plate directly 
into the glass trough of the chromatography tank. Cover and seal the 
tank. Allow the solvent front to travel about 11.5 centimeters from the 
bottom of the plate, remove the plate from the tank, and allow to air 
dry. Apply the spray solution (do not saturate) and place immediately 
into an oven maintained at 90 deg. C. Heat 15 minutes.
    (f) Evaluation. Measure the distance the solvent front traveled from 
the starting line and the distance the spots are from the starting line. 
Calculate the Rf value by dividing the latter by the former. 
The sample and standard should have spots of corresponding Rf 
values.
[39 FR 18944, May 30, 1974, as amended at 45 FR 16472, Mar. 14, 1980]



Sec. 436.306  Lincomycin gas liquid chromatography.

    (a) Equipment. Gas chromatograph equipped with a flame ionization 
detector; Barber-Colman 5000 or equivalent.
    (b) Reagents. (1) Pyridine, reagent grade, kept over potassium 
hydroxide.
    (2) Methanol, reagent grade, anhydrous.
    (3) Ethanol, absolute, reagent grade.
    (4) Internal standard: Prepare a solution containing 2 milligrams of 
tetraphenylcyclopentadienone per milliliter in pyridine.

[[Page 402]]

    (5) Silylating reagent: Mix (9+1) of hexamethyldisilazane and 
trimethyl chlorosilane.
    (c) Typical conditions. (1) Column: 4 feet  x  3 millimeters ID, 
glass, with 3 percent SE-30 on Gas-Chrom Q (100/120 mesh), or 
equivalent.
    (2) Temperatures: Column 225 deg. C.; detector 280 deg. C., injector 
270 deg. C.
    (3) Carrier gas: Helium at 15 pounds per square inch.
    (4) Detector: Hydrogen flame ionization--hydrogen at 20 pounds per 
square inch, air at 40 pounds per square inch.
    (5) Sensitivity: 100; attenuation 2; current 2  x  10- 8 
amperes.
    (d) Preparation of lincomycin sample and working standard solutions. 
Prepare the sample and working standard as follows: Weigh accurately an 
aliquot of about 40 milligrams into a 10-milliliter volumetric flask, 
add sufficient pyridine to dissolve, and make to mark. Transfer a 1-
milliliter aliquot to a glass-stoppered conical centrifuge tube and 
proceed as directed in paragraph (e) of this section.
    (e) Procedure. Add 0.2 milliliter of the silylating reagent to each 
centrifuge tube and allow to stand at least 30 minutes. Then add exactly 
1 milliliter of the internal standard, shake well, and centrifuge. 
Inject 5 microliters of the supernatant into the gas chromatograph. Use 
the typical conditions and materials listed in paragraphs (a), (b), and 
(c) of this section. The conditions should be adequate to provide at 
least 60 percent scale deflection with the lincomycin peak and to 
maintain a stable base line. The resolution of the peaks should be 
complete. The elution order is lincomycin B, lincomycin, and the 
internal standard. If necessary, adjust the current setting for the 
lincomycin B peak to give a satisfactory response relative to that of 
the lincomycin peak. Calculate the lincomycin content and lincomycin B 
content as directed in paragraph (f) of this section.
    (f) Calculations.
    [GRAPHIC] [TIFF OMITTED] TR01JA93.033
    
where:
Ru=Area of the lincomycin sample peak/Area of internal 
standard peak;
Rs=Area of the lincomycin standard peak/Area of internal 
standard peak;
Ws=Weight of the lincomycin working standard in milligrams;
Wu=Weight of the sample in milligrams;
f=Potency of lincomycin working standard in micrograms per milligram.

[GRAPHIC] [TIFF OMITTED] TR01JA93.034

where:
A=Area of lincomycin peak of the sample;
B=Area of lincomycin B peak of the sample corrected for the attenuation 
adjustment.
[39 FR 18944, May 30, 1974, as amended at 46 FR 3839, Jan. 16, 1981]



Sec. 436.307  Spectinomycin vapor phase chromatography.

    (a) Equipment. Gas chromatograph equipped with a flame ionization 
detector; Barber-Colman 5,000 or equivalent.
    (b) Reagents. (1) Dimethylformamide, reagent grade, kept dry over 
anhydrous sodium sulfate.
    (2) Internal standard: Prepare a solution containing 2 milligrams of 
triphenylantimony per milliliter in dry dimethylformamide.
    (3) Silylating reagent: Hexamethyl-disilazane.
    (c) Typical conditions. (1) Column: 4 feet by 4 millimeters ID, 
glass, with 5 percent SE-52 on Diatoport S (80/100 mesh), or equivalent.
    (2) Temperatures: Column 215 deg. C.; detector 270 deg. C.; 
injection port 265 deg. C.
    (3) Carrier gas: Helium 93 milliliters per minute at 15 pounds per 
square inch.
    (4) Detector: Hydrogen flame--hydrogen at 20 pounds per square inch, 
air at 40 pounds per square inch.
    (5) Sensitivity: 1,000; attenuation, 10 for both spectinomycin and 
internal standard; 2  x  10- 6 amperes.
    (d) Preparation of spectinomycin sample and working standard--(1) 
Working standard and bulk antibiotic solutions. (i) Accurately weigh 
approximately 30 milligrams of sample or working standard into separate 
glass-stoppered 25-milliliter Erlenmeyer flasks.
    (ii) Add 10 milliliters of the internal standard solution and 1.0 
milliliter of hexamethyldisilazane to each flask. Agitate the flasks to 
insure dissolution of the sample and working standard

[[Page 403]]

and complete mixing of the liquids. Shake the flasks intermittently for 
1 hour. Proceed as directed in paragraph (e) of this section.
    (2) Finished product solutions. Prepare the sample for assay as 
directed in the individual section for each antibiotic product to be 
tested.
    (e) Procedure. Inject 2.5 microliters of each solution into the gas 
chromatograph. Use the conditions and materials listed in paragraphs 
(a), (b), and (c) of this section. The conditions should be adequate to 
maintain a stable base line and provide at least 60 percent deflection 
of the recorder scale by the spectinomycin peak. The resolution of the 
peaks should be complete. The internal standard will be eluted before 
spectinomycin. Calculate the spectinomycin content as directed in 
paragraph (f) of this section.
    (f) Calculations. Calculate the spectinomycin content of the sample 
as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.035

where:
Ru=Area of spectinomycin sample peak (at a retention time 
equal to that observed for the spectinomycin standard)/Area of internal 
standard peak;
Rs=Area of the spectinomycin standard peak/Area of internal 
standard peak;
Ws=Weight of the spectinomycin working standard in 
milligrams;
Wu=Weight of the sample in milligrams;
f=Potency of the spectinomycin working standard in micrograms per 
milligram.



Sec. 436.308  Paper chromatography identity test for tetracyclines.

    (a) Equipment--(1) Sheet (chromatographic). Whatman No. 1 filter 
paper for chromatography, 20  x  20 centimeters.
    (2) Chamber (chromatographic). Cylindrical glass chromatographic 
jar, 25 centimeters high by 12 centimeters in diameter, with a ground-
glass lid.
    (3) Preparation of solutions--(i) pH3.5 buffer. Mix 13.93 volumes of 
0.1M citric acid with 6.07 volumes of 0.2M of disodium phosphate.
    (ii) Solvent (organic phase). Mix chloroform, nitromethane, and 
pyridine in volumetric proportions of 10:20:3, respectively.
    (b) Preparation of spotting solutions. Prepare solutions of the 
working standard and sample as follows: Accurately weigh a portion of 
the working standard and sample and dilute with methanol to obtain a 
concentration of 1 milligram per milliliter of antibiotic to be tested.
    (c) Procedure. Fill the chamber to a depth of 0.6 centimeter with 
freshly prepared solvent. Draw a starting line about 2.5 centimeters 
from and parallel to the bottom of the sheet. Wet the sheet thoroughly 
with the pH 3.5 buffer and blot it firmly between sheets of absorbent 
paper. Starting about 5 centimeters from the edge of the sheet and at 
1.5-centimeter intervals, apply to the starting line 2 microliters each 
of standard solution, sample solution, and a 1:1 mixture of the standard 
and sample solutions. Allow a few minutes for the sheet to dry 
partially, and while still damp place it in the chamber with the bottom 
edge touching the solvent. When the solvent front has risen about 10 
centimeters, remove the sheet from the chamber. Expose the paper to 
ammonia vapor. Examine the dried sheet under a strong source of 
ultraviolet light and record the position of any fluorescent spots. 
Measure the distance the solvent front traveled from the starting line 
and the distance that the fluorescent spots are from the starting line. 
Calculate the Rf value by dividing the latter by the former.
[39 FR 18944, May 20, 1974, as amended at 44 FR 30333, May 5, 1979; 45 
FR 16472, 16474, Mar. 14, 1980]



Sec. 436.309  Anhydrotetracyclines and 4-epianhydrotetracycline.

    Determination of 4-epianhydrotetracycline and anhydrotetracyclines 
in tetracycline, tetracycline hydrochloride, tetracycline phosphate, and 
in dosage forms thereof is as follows:
    (a) Screening procedure for total anhydrotetracyclines content--(1) 
Sample solution preparation--(i) Bulk packaged for repacking or for use 
in the manufacture of another drug. Accurately weigh approximately 50 
milligrams of the sample into a 50-milliliter volumetric flask and add 
10 milliliters of 0.1N hydrochloric acid. Shake until sample is

[[Page 404]]

completely dissolved, and then dilute to volume with water.
    (ii) Sterile dispensing containers. Proceed as directed in paragraph 
(a)(1)(i) of this section.
    (iii) Capsules. Transfer a representative quantity of capsule 
contents equivalent to 250 milligrams of tetracycline hydrochloride to a 
250-milliliter volumetric flask. Add 50 milliliters of 0.1N hydrochloric 
acid and shake on a mechanical shaker for 5 minutes. Dilute to volume 
with water and filter through a fluted filter paper. Discard the first 
20 milliliters of filtrate and collect the next 20 milliliters.
    (iv) Tablets. Grind a representative number of tablets to a fine 
powder. Transfer an amount of the powder equivalent to 250 milligrams of 
tetracycline hydrochloride to a 250-milliliter volumetric flask. Add 50 
milliliters of 0.1N hydrochloric acid and shake on a mechanical shaker 
for 5 minutes. Dilute to volume with water and filter through a fluted 
filter paper. Discard the first 20 milliliters of filtrate and collect 
the next 20 milliliters.
    (v) Oral powders and suspensions. Proceed as described in paragraph 
(b) of this section.
    (2) Test procedure. Using a suitable spectrophotometer, determine 
the absorbance of the sample solution prepared as directed in paragraph 
(a)(1) of this section at 430 millimicrons using 0.02N hydrochloric acid 
as a blank. Then accurately dilute 1.0 milliliter of the sample solution 
to 100 milliliters with 0.02N hydrochloric acid and determine the 
absorbance of this solution at 356 millimicrons, using 0.02N 
hydrochloric acid as a blank.
    (3) Calculations.
    [GRAPHIC] [TIFF OMITTED] TR01JA93.036
    
where:
a430=Absorptivity (1%, 1 cm.) of sample at 430 millimicrons;
[GRAPHIC] [TIFF OMITTED] TR01JA93.037

For sterile dispensing containers, capsules, and tablets; 
absorptivity=Absorb-ance x 10; a356=Absorptivity (1%, 1 cm.) of sample 
at 356 millimicrons;
[GRAPHIC] [TIFF OMITTED] TR01JA93.038

For sterile dispensing containers, capsules and tablets; 
absorptivity=Absorb-ance x 1,000; 0.0019 x Absorbance ratio (A430/A356) 
observed with tetracycline;
195=Absorptivity (1%, 1 cm.) of anhydrotetracycline hydrochloride at 430 
millimicrons.

    (4) Evaluation. If the total anhydrotetracyclines content determined 
by the screening procedure described in paragraph (a) of this section 
exceeds 2 percent for bulks and 3 percent for injectables, tablets, and 
capsules, perform the determination for anhydrotetracyclines and 4-
epianhydrotetracycline described in paragraph (b) of this section. If 
the results of the test described in paragraph (a) of this section for 
total anhydrotetracyclines content are within the required limits in the 
case of bulks, injectables, tablets, and capsules, these results may be 
submitted in lieu of the results of the test for 4-
epianhydrotetracycline and that test as described in paragraph (b) of 
this section need not be performed.
    (b) Determination of anhydrotetracyclines content and 4-
epianhydrotetra-cycline content--(1) Apparatus and reagents--(i) 
Chromatographic tubes (15 millimeters ID  x  170 millimeters long having 
an outlet tube 4 millimeters ID  x  50 millimeters long).
    (ii) pH meter standardized at pH 7.0 and at pH 10.0.
    (iii) Diatomaceous earth, acid-washed (Celite 545 or equivalent).
    (iv) EDTA buffer. Dissolve 0.1 mole ethylenediaminetetraacetic acid 
disodium salt in 800 milliliters of water.

[[Page 405]]

Adjust to pH 7.8 with ammonium hydroxide, reagent grade, and dilute to 1 
liter with water.
    (v) Chloroform, spectrophotometric grade.
    (vi) Diluted ammonium hydroxide: Mix 1 volume of ammonium hydroxide, 
reagent grade, with 9 volumes of distilled water.
    (vii) 0.1N hydrochloric acid.
    (viii) 1.0N hydrochloric acid.
    (2) Preparation of support phase. Add 5 milliliters of EDTA buffer 
to 10 grams of diatomaceous earth and mix until the diatomaceous earth 
is uniformly moistened. It will no longer be free-flowing.
    (3) Preparation of sample solutions. Prepare the sample solutions as 
follows:
    (i) Tetracycline, tetracycline phosphate complex, and tetracycline 
hydrochloride bulk packaged for repacking or for use in the manufacture 
of another drug. Place an amount of sample equivalent to 250 milligrams 
of tetracycline hydrochloride into a 50-milliliter beaker and dissolve 
in 10 milliliters of 0.1N hydrochloric acid. Immediately adjust the pH 
to 7.8 with the diluted ammonium hydroxide, and if necessary, with 1.0N 
hydrochloric acid and 0.1N hydrochloric acid. Quantitatively transfer 
this solution to a 50-milliliter volumetric flask by rinsing the beaker 
with EDTA buffer, fill to volume with EDTA buffer and shake well. Use 
this solution without delay to prepare a column as directed in paragraph 
(b)(4) of this section.
    (ii) Capsules. Proceed as directed in paragraph (b)(3)(i) of this 
section, except pool the contents of a representative number of capsules 
and use an amount of the pooled capsule contents equivalent to 250 
milligrams of tetracycline hydrochloride.
    (iii) Tablets. Proceed as directed in paragraph (b)(3)(i) of this 
section, except grind tablets to a powder in a small mortar and use an 
amount of powder equivalent to 250 milligrams of tetracycline 
hydrochloride.
    (iv) Oral suspension and pediatric drops. Place 5 milliliters of 
oral suspension equivalent to 125 milligrams of tetracycline 
hydrochloride or 2 milliliters of pediatric drops equivalent to 200 
milligrams of tetracycline hydrochloride into a 50-milliliter beaker and 
add sufficient 0.1N hydrochloric acid to make 10 milliliters. Quickly 
adjust the pH to 7.8 with the diluted ammonium hydroxide, and if 
necessary, with 1N hydrochloric acid and 0.1N hydrochloric acid. 
Quantitatively transfer this solution to a 25-milliliter flask by 
rinsing the beaker with EDTA buffer, fill to volume with EDTA buffer, 
and shake well. Use this solution without delay to prepare a column as 
directed in paragraph (b)(4) of this section.
    (v) Oral powders. Reconstitute as directed in the labeling and 
proceed as directed in paragraph (b)(3)(iv) of this section.
    (vi) Sterile dispensing containers. Proceed as directed in paragraph 
(b)(3)(i) of this section.
    (4) Column preparation. Pack support phase into the chromatographic 
tube by increments and firmly tamp down each increment. Do not use any 
glass wool in the column outlet. Add enough support phase to the column 
to reach a height of 9 to 11 centimeters; then add 1 milliliter of 
sample solution to 1 gram of diatomaceous earth in a small beaker, and 
mix thoroughly. Pack the sample: diatomaceous earth mixture on top of 
the column. Dry wash the beaker with support phase and pack an 
additional 1-centimeter layer of support phase on top of the sample 
layer.
    (5) Column elution and fraction collection. Within 30 minutes after 
preparing the column, elute with chloroform. Collect 5 successive 
fractions of 5 milliliters, 5 milliliters, 10 milliliters, 10 
milliliters, and 5 milliliters. During elution, two clear separate 
yellow bands will appear on the column. The first band is 
anhydrotetracyclines and will almost always elute in the first 5-
milliliter fraction, but occasionally in the first and second 5-
milliliter fractions. The second band is 4-epianhydrotetracycline and 
will elute in the remaining fractions. Label the fraction or fractions 
containing the first yellow band anhydrotetracyclines. Label the 
fractions after the first yellow band 4-epianhydrotetracycline. 
Determine the absorbance of each fraction at a wavelength of 438 
nanometers using a suitable spectrophotometer equipped with a 1.0-
centimeter cell and chloroform as the blank. If necessary,

[[Page 406]]

make appropriate dilutions with choloroform to obtain a readable value.
    (6) Calculations--(i) Percent anhydrotetracyclines. Calculate the 
percent anhydrotetracyclines as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.039

where:
A=Absorbance of the sample solution at 438 nanometers;
b=Volume of fraction in milliliters;
c=Dilution factor of the fraction (for example, if 2 milliliters of the 
fraction are diluted to 10 milliliters for reading, c will be 5).
20.28=Absorptivity (1 milligram per milliliter, 1 centimeter) of 
anhydrotetra- cyclines in chloroform at 438 nanometers.
Total weight of anhydrotetracyclines in the sample=Sum of weights of 
anhydrotetracyclines in the fractions labeled anhydrotetracyclines  x  
Number of milliliters in the sample solution
Percent anhydrotetracyclines in tetracycline, tetracycline 
hydrochloride, tetracycline phosphate complex bulk packaged for 
repacking or for use in the manufacture of another drug =
[GRAPHIC] [TIFF OMITTED] TC01AP94.007

Percent anhydrotetracyclines in dosage forms=
[GRAPHIC] [TIFF OMITTED] TC01AP94.008

    (ii) Percent 4-epianhydrotetracycline. Calculate the percent 4-
epianhydrotetracycline as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.040

where:
A=Absorbance of the sample solution at 438 nanometers;
b=Volume of the fraction in milliliters;
c=Dilution factor of the fraction (for example, if 2 milliliters of the 
fraction are diluted to 10 milliliters for reading, c will be 5);
20.08=Absorptivity (1 milligram per milliliter, 1 centimeter) of 4-
epianhydrotetracycline in chloroform at 438 nanometers.
Total weight of 4-epianhydrotetracycline in the sample=Sum of weights of 
4-epianhydrotetracycline in the fractions labeled 4-
epianhydrotetracycline  x  Number of milliliters in the sample solution
Percent 4-epianhydrotetracycline in tetracycline, tetracycline 
hydrochloride, tetracycline phosphate complex bulk packaged for 
repacking or for use in the manufacture of another drug =
[GRAPHIC] [TIFF OMITTED] TC01AP94.009

Percent 4-epianhydrotetracycline in dosage forms=
[GRAPHIC] [TIFF OMITTED] TC01AP94.010

[39 FR 18944, May 30, 1974, as amended at 40 FR 22251, May 22, 1975; 43 
FR 11153, Mar. 17, 1978]



Sec. 436.310  Thin layer chromatography identity test for mitomycin.

    (a) Equipment--(1) Chromatography tank. A rectangular tank, 
approximately 9  x  9  x  3.5 inches, lined with filter paper and with a 
solvent trough on the bottom.

[[Page 407]]

    (2) Plates. Use 20 by 20 centimeter thin layer chromatography plates 
coated with silica gel G or equivalent, to a thickness of 250 microns.
    (b) Reagents--(1) Developing solvent. Mix n-butanol, glacial acetic 
acid, and water in volumetric proportions of 4:2:1, respectively.
    (2) Spray solution. Prepare a one-percent solution of ninhydrin in 
ethanol.
    (c) Preparation of spotting solutions. Prepare solutions of the 
sample and working standard, each containing 1 milligram of mitomycin 
per milliliter, in water.
    (d) Procedure. Pour the developing solvent into the solvent trough 
on the bottom of the tank and onto the paper lining the walls of the 
tank. Cover and seal the tank. Allow it to equilibrate for 30 minutes. 
Prepare a plate as follows: Apply spotting solutions on a line 2.5 
centimeters from the base of the silica gel plate and at points 2.0 
centimeters apart. Apply approximately 2 microliters of the working 
standard solution to points 1 and 3. When these spots are dry, apply 
approximately 2 microliters of sample solution to points 2 and 3. After 
all spots are thoroughly dry, place the silica gel plate into the trough 
in the chromatography tank. Cover and seal the tank tightly. Allow the 
solvent front to travel about 10 centimeters from the starting line. 
Remove the plate and allow it to air dry. After the plate is dry, spray 
lightly with the spray solution. Heat the plate in an oven at 110 deg. 
C. for 10-15 minutes. Mitomycin appears as a pink spot.
    (e) Evaluation. The sample and standard should have spots of 
corresponding Rf value (approximately 0.51), and standard and 
sample combined should appear as a single spot of corresponding Rf 
value.
[39 FR 18944, May 30, 1974, as amended at 49 FR 2242, Jan. 19, 1984]



Sec. 436.311  Thin layer chromatography identity test for amoxicillin.

    Using the sample solution prepared as described in the section for 
the antibiotic drug to be tested, proceed as described in paragraphs (a) 
through (e) of this section.
    (a) Equipment--(1) Chromatography tank. A rectangular tank, 
approximately 23 centimeters long, 23 centimeters high, and 9 
centimeters wide, equipped with a glass solvent trough in the bottom and 
a tight-fitting cover for the top. Line the inside walls of the tank 
with Whatman's 3MM chromatographic paper (0.33 millimeters) or 
equivalent.
    (2) Plates. Use 20- by 20-centimeter thin layer chromatography 
plates coated with Silica Gel G or equivalent to a thickness of 250 
microns.
    (b) Reagents--(1) Developing solvent. Mix methyl alcohol, 
chloroform, pyridine, and distilled water in volumetric proportions of 
90:80:10:30, respectively.
    (2) Spray solution. Dissolve 300 milligrams of ninhydrin in 100 
milliliters of ethyl alcohol.
    (c) Preparation of working standard. Weigh an amount of the 
amoxicillin working standard equivalent to 200 milligrams of amoxicillin 
into a 50-milliliter volumetric flask and bring to volume with 0.1N 
hydrochloric acid.
    (d) Procedure. Pour the developing solvent into the glass trough on 
the bottom of the tank and onto the paper lining the walls of the tank. 
Cover and seal the tank. Allow it to equilibrate for at least 2 hours. 
Spot duplicate plates by applying approximately 5 microliters each of 
standard and sample solutions on a line 1.5 centimeters from the base of 
the plate and at intervals of not less than 2.0 centimeters. All 
solutions must be spotted within 10 minutes of preparation. Place 
spotted plate in a desiccator until solvent has evaporated from spots. 
Place the plate into the glass trough at the bottom of the 
chromatography tank. Cover the tank. Allow the solvent to reach the 15-
centimeter scored mark, remove the plate from the tank and dry with a 
current of warm air until there is no detectable solvent odor. Apply the 
ninhydrin spray solution to the plate--do not saturate--and place 
immediately into an oven maintained at 110 deg. C for 15 minutes.
    (e) Evaluation. Measure the distance the solvent front traveled from 
the starting line and the distance the spots are from the starting line. 
Calculate the Rf value by dividing the latter by the former. 
Amoxicillin has an Rf value of about 0.53. The sample and 
standard

[[Page 408]]

should have spots of corresponding Rf values.
[39 FR 34032, Sept. 23, 1974; 48 FR 11427, Mar. 18, 1983, as amended at 
49 FR 2242, Jan. 19, 1984]



Sec. 436.312  Atomic absorption method for determining the zinc content of zinc bacitracin.

    (a) Equipment. An atomic absorbance spectrophotometer equipped with 
a zinc hollow-cathode discharge lamp, an air-acetylene flame, a 
nebulizer-burner system for introducing the sample solution into the 
flame, an optical dispersing device (such as a monochromator) for 
isolating a resonance line of zinc from others produced by the emission 
source, and a suitable radiation detector and recorder.
    (b) Preparation of working standard and sample solutions--(1) 
Workingstandard solutions. Prepare a standard stock solution containing 
10 milligrams of zinc per milliliter as follows: Weigh 3.11 grams of 
zinc oxide into a 250-milliliter volumetric flask, add 80 milliliters of 
1N HCl, warm to dissolve, cool to room temperature, and dilute to volume 
with water. Dilute aliquots of this standard stock solution with 0.001N 
HCl to obtain three working standard solutions containing respectively 
0.5, 1.5, and 2.5 micrograms of zinc per milliliter.
    (2) Sample solution. Accurately weigh approximately 200 milligrams 
of the sample into a 100-milliliter volumetric flask. Dissolve and 
dilute to volume with 0.01N HCl. Transfer a 2.0-milliliter aliquot of 
this solution to a 200-milliliter volumetric flask and dilute to volume 
with 0.001N HCl.
    (c) Procedure. Using 0.001N HCl as the blank, adjust the absorbance 
of the instrument to zero at a detection wavelength of 213.8 nanometers. 
Determine the absorbance of each standard solution and the sample 
solution at 213.8 nanometers.
    (d) Calculations. Plot the absorbance versus the concentration of 
each of the working standard solutions. Draw a straight response line of 
best fit through these points. Read the concentration of zinc in 
micrograms per milliliter corresponding to the absorbance of the sample 
solution. Calculate the percent zinc in the sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.041

where:
C=Concentration of zinc in the sample solution in micrograms per 
milliliter;
m=Percent moisture in the sample.
[40 FR 15088, Apr. 4, 1975]



Sec. 436.316  Determination of penicillin G content.

    (a) Reagents. The reagents are freshly prepared every three days and 
are of such quality that when used in this procedure with an authentic 
sample of penicillin G, not less than 97 percent of penicillin G is 
recovered.
    (1) Amyl acetate (iso-amyl acetate) solution. Saturate the amyl 
acetate (boiling range 138.5 deg. C--141.5 deg. C) with the N-
ethylpiperidine salt of penicillin G by adding 2 milligrams of the salt 
for each 1.0 milliliter of the solvent. Cool this solution to 0 deg. C--
8 deg. C and filter it through a sintered-glass filter immediately 
before use.
    (2) Acetone solution. Saturate reagent grade acetone with the N-
ethylpiperidine salt of penicillin G using 3 milligrams of salt for each 
1 milliliter of acetone. Cool this solution to 0 deg. C--8 deg. C and 
filter it through a sintered-glass filter immediately before use.
    (3) N-ethylpiperidine solution. N-ethylpiperidine (boiling range 
129.5 deg. C--131.0 deg. C) should be stored in brown bottles in a 
refrigerator. Dilute 1.0 milliliter of this reagent with 4.0 milliliters 
of amyl acetate. Saturate this solution with the N-ethylpiperidine salt 
of penicillin G, using about 3 milligrams of the salt for each 1.0 
milliliter of solution. Cool this solution to 0 deg. C--8 deg. C and 
filter it through a sintered-glass filter immediately before use.
    (4) Phosphoric acid solution. Prepare by dissolving 1.0 milliliter 
of reagent grade phosphoric acid (85 percent) in 4.0 milliliters of 
water. Cool to 0 deg. C--8 deg. C and shake before using.
    (5) Silica gel. Use dry silica gel (mesh size 6-16, Tyler standard). 
Place about

[[Page 409]]

0.5 gram of the silica gel in a micro filter funnel (approximately 10-
millimeter diameter) having a fritted-glass disc of medium porosity.
    (b) Procedure. Accurately weigh from 60 to 70 milligrams of the 
sample to be tested, except if penicillin G procaine is to be tested 
weigh 90 to 100 milligrams of sample, into a glass test tube or glass 
vial of approximately 10-milliliter capacity. Add 2.0 milliliters of 
water to dissolve or suspend (procaine) the penicillin and cool to 
0 deg. C--5 deg. C. Add 2.0 milliliters of amyl acetate solution and 0.5 
milliliter of phosphoric acid solution, stopper and shake the container 
vigorously for approximately 15 seconds. For penicillin G procaine, add 
a second 0.5-milliliter portion of phosphoric acid solution and shake 
vigorously. Centrifuge to obtain a clear separation of the two layers 
(approximately 20 seconds). If any penicillin procaine remains 
undissolved, add a third 0.5-milliliter portion of phosphoric acid 
solution, shake the container vigorously, and centrifuge. After 
centrifuging, remove as much of the amyl acetate layer as possible, 
usually about 1.7 milliliters to 1.8 milliliters, with a suitable 
hypodermic needle and syringe and place the portion removed into the 
filter funnel containing silica gel, described in paragraph (a)(5) of 
this section. Allow the amyl acetate to remain in contact with the 
silica gel for exactly 20 seconds, then apply suction and collect the 
filtrate in a small test tube placed in a suction flash surrounded by 
cracked ice. Pipet a 1.0-milliliter aliquot of the amyl acetate filtrate 
into a tared flat-bottom glass tube (approximately 15 x 50 millimeters) 
containing 1.0 milliliter of acetone solution and 0.5 milliliter of N-
ethylpiperidine solution. The time elapsing between acidification and 
the addition of the filtrate to the above reagents should not be more 
than 3 minutes. Place the glass tube containing the mixture into a large 
weighing bottle, stopper the bottle and allow to stand for not less than 
2 hours in a refrigerator at 0 deg. C--8 deg. C. Remove the liquid from 
the precipitate by means of a tared micro filter stick and wash with a 
total of 1.0 milliliter of acetone solution adding the latter by means 
of a hypodermic syringe equipped with a fine needle. Place the filter 
stick inside the glass tube, dry under vacuum at room temperature for 
not less than 1 hour, and weigh. (The N-ethylpiperidine penicillin G 
residues can be saved for saturating reagents).
    (c) Calculations. Calculate the percent penicillin G content as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.042

[42 FR 59857, Nov. 22, 1977]



Sec. 436.317  Solubility characteristic test for griseofulvin (ultramicrosize) tablets.

    (a) Apparatus--(1) Vessel. A cylindrical glass tank. The approximate 
dimensions are 40 centimeters in diameter and at least 23 centimeters in 
height.
    (2) Heating system. A 1,500-watt immersion heating element connected 
to a partial immersion, contact thermometer and an appropriate control 
relay.
    (3) Circulating system components. The circulating system consists 
of three different circulating devices:
    (i) Circulating pump of a centrifugal, immersion type. Tubing 
approximately 1 centimeter outside diameter and 46 centimeters in length 
is attached to the pump outlet producing a flow rate of approximately 
1,600 milliliters per minute when operated as described.
    (ii) A ``4-element stirrer'' consisting of a motor and a shaft 
approximately 45 centimeters long and 8 millimeters in diameter. The 
motor rotates the vertical shaft in a clockwise direction at 
approximately 180 revolutions per minute. There are 4 elements or sets 
of stirring blades on the shaft. One set, located at the bottom of the 
shaft, is a 3-bladed element of 2.5 centimeters

[[Page 410]]

overall radius with circular blades, 1.8 centimeters in diameter and 1 
to 2 millimeters in thickness, pitched at an angle of approximately 45 
degrees from the horizontal plane, so that fluid is propelled downward 
when the shaft is rotated in a clockwise direction. The three remaining 
sets of stirring blades have 4 blades each, symmetrically positioned 
about the shaft. Each set of blades is 3.2 centimeters in overall 
radius. Each blade is rectangular in shape, 2.4 centimeters in length, 
1.2 centimeters in height, and 1 to 2 millimeters in thickness. The four 
sets of blades are located at 5 centimeter intervals on the shaft, the 
top three being fixed in a staggered configuration.
    (iii) A rotating basket device consisting of a motor capable of 
constant speed of 100plus-minus5 revolutions per minute in a 
clockwise direction, a shaft, and a cylindrical basket. The shaft and 
the basket are fabricated from Type 316 stainless steel. The shaft is 6 
millimeters in diameter and approximately 30 centimeters in length. It 
must run true on the motor axis so that the basket rotates smoothly and 
without perceptible wobble. The basket consists of two parts, one of 
which, the top, is attached to the shaft. It is of solid metal except 
for a 2-millimeter round vent, and is fitted with three spring clips 
that allow the removal of the lower part, or the basket proper, to admit 
the test sample. The detachable part of the basket is fabricated of 
welded seam stainless steel, 40 mesh woven wire cloth, formed into a 
cylinder 3.66 centimeters high and 2.5 centimeters in diameter, with a 
narrow rim of sheet metal around the top.
    (4) Circulating system configuration. All three circulating devices 
are located in one half of the tank. In clockwise order they are the 
circulating pump, the rotating basket, and the 4-element stirrer. There 
is a distance of 12 to 13 centimeters between each of the three devices. 
The rotating basket shaft and the stirring shaft are located 9 to 10 
centimeters from the tank wall. The 4-element stirrer is positioned 1 to 
1.5 centimeters from the bottom of the tank. The rotating basket is 
fixed at 7 to 8 centimeters from the bottom. The circulating pump intake 
is located approximately 3 centimeters from the top of the fluid in the 
tank and 5 to 6 centimeters from the wall of the tank. The pump's outlet 
hose is held by a clamp so that hose makes a clockwise arc around the 
inside wall of the tank, descending to a point near the bottom of the 
tank and 5 to 6 centimeters from the wall, which is 180 degrees from the 
pump inlet.
    (b) Dissolution medium. Distilled water.
    (c) Procedure. Place 24 liters of dissolution medium into the vessel 
and maintain the temperature at 37plus-minus0.5 deg. C by 
means of the heater, circulating pump, and the 4-element stirrer. 
Withdraw a 25-milliliter portion of the dissolution medium as a sample-
blank solution. Place one tablet into the basket, and lower it into its 
proper position in the tank. Rotate the basket at 
100plus-minus5 revolutions per minute in a clockwise 
direction. After 60 minutes, withdraw a second 25-milliliter portion as 
the sample solution. Filter the sample-blank solution and the sample 
solution through water-washed glass wool, or an equivalent filter, 
discarding the first 10 to 15 milliliters of each filtrate. Determine 
the amount of griseofulvin dissolved as directed in paragraph (d)(2) of 
this section.
    (d) Griseofulvin assay--(1) Preparation of standard solution and 
standard-blank solution. Accurately weigh approximately 50 milligrams of 
griseofulvin working standard and place into a 100-milliliter volumetric 
flask. Dissolve and dilute to volume with methyl alcohol. Transfer 2.0 
milliliters of this solution to a 200-milliliter volumetric flask and 
dilute to volume with distilled water. This is the standard solution. 
Transfer a 2.0-milliliter portion of methyl alcohol to a 200-milliliter 
volumetric flask and dilute to volume with distilled water. This is the 
standard-blank solution. Filter the standard-blank solution and the 
standard solution through water-washed glass wool, or an equivalent 
filter, discarding the first 10 to 15 milliliters of each filtrate.
    (2) Procedure. Using a suitable spectrophotometer and distilled 
water as the blank, determine the absorbance of the four filtered 
solutions at the absorbance peak at approximately 295

[[Page 411]]

nanometers, using suitable spectrophotometer cells with a 1-centimeter 
light path. Determine the exact position of the absorbance peak for the 
particular instrument used.
    (3) Calculation. Determine the percentage of griseofulvin dissolved 
as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.043

where:
Au=Absorbance of the sample solution minus the absorbance of 
the sample-blank solution;
Ws=Weight of the working standard in milligrams;
V=Volume of the dissolution medium in liters;
As=Absorbance of the standard solution minus the absorbance 
of the standard-blank solution;
P=Labeled potency of the sample in milligrams of griseofulvin per 
tablet.

    (e) Evaluation. The tablet passes the solubility characteristic test 
if it dissolves to the extent of not less than 50 percent at 60 minutes. 
If the tablet fails to meet this requirement, repeat the test on five 
additional tablets. The batch passes the solubility characteristic test 
if not less than 5 of 6 tablets meet the requirement.
[40 FR 41522, Sept. 8, 1975; 40 FR 45426, Oct. 2, 1975]



Sec. 436.318  Continuous flow thin layer chromatography identity test.

    (a) Equipment--(1) Chromatography tank. A rectangular tank, 
approximately 23 centimeters long, 23 centimeters high, and 9 
centimeters wide equipped with a glass solvent trough in the bottom.
    (2) Plates. Use a 20  x  20 centimeter thin-layer chromatography 
plate coated with Silica Gel G or equivalent to a thickness of 250 
micrometers.
    (3) Cover. A stainless steel cover with a slot, measuring 21  x  0.6 
centimeters, cut in the front edge.
    (4) Supporting platform. A platform that can be placed in the bottom 
of the chromatography tank so that the solvent trough is elevated about 
3.75 centimeters.
    (b) Reagents--(1) Developing solvent. Mix chloroform, redistilled 
methanol and concentrated ammonium hydroxide in volumetric proportions 
of 25:60:30, respectively.
    (2) Spray solution. Dissolve 1 gram of ninhydrin in 100 milliliters 
of n- butanol and add 1 milliliter of pyridine.
    (c) Preparation of spotting solutions. Prepare solutions of the 
sample and working standard, each containing 6 milligrams of antibiotic 
to be tested per milliliter in distilled water.
    (d) Procedure. Prepare a plate as follows: On a line 2 centimeters 
from the base of the silica gel plate, and at intervals of 1 centimeter, 
spot 3 microliters each of the standard solution and the sample 
solution. In addition, prepare one spot composed of 3 microliters of the 
sample solution and 3 microliters of the standard solution. Place the 
supporting platform in the bottom of the tank and place the solvent 
trough on it, near the front of the tank. Place a piece of Whatman 3 MM 
filter paper or equivalent, measuring 20x3 centimeters and folded in 
half, lengthwise, over the front edge of the tank to form a cushion and 
drying wick for the plate. Place the plate in the solvent trough with 
the coated side toward the front of the tank and leaning against the 
filter paper at the top. Pour the developing solvent into the trough and 
bottom of the tank. Cover the tank. The plate should extend 
approximately 1 centimeter beyond the top of the tank and through the 
slot in the cover. Seal all the openings in the tank with masking tape, 
except where the plate leans against the filter paper. Remove the plate 
from the tank after 5.5 hours. Allow the plate to air dry and then heat 
it for 15 minutes at 110 deg. C in an oven. Remove the plate from the 
oven and immediately spray it with the spray solution. The compound 
appears as a pink spot.
    (e) Evaluation. The sample and standard should have traveled the 
same distance from the origin, and the standard and sample combined 
should appear as a single spot that has traveled the same distance as 
the sample and standard individually.
[40 FR 57797, Dec. 12, 1975]

[[Page 412]]



Sec. 436.319  Thin layer chromatography identity test for bacitracin and bacitracin zinc.

    (a) Equipment--(1) Chromatography tank. A rectangular tank 
approximately 23 centimeters long, 23 centimeters high, and 9 
centimeters wide, equipped with a glass solvent trough in the bottom and 
a tight-fitting cover for the top. Line the inside walls of the tank 
with Whatman 3MM chromatographic paper or equivalent.
    (2) Plates. Use a 20- by 20-centimeter thin layer chromatography 
plate coated with silica gel G or equivalent to a thickness of 250 
micrometers. Activate the plate by heating for 20 minutes at 110 deg. C. 
Allow to cool to room temperature and use immediately.
    (b) Reagents--(1) Developing solvent. Mix n- butanol, water, 
pyridine, glacial acetic acid, and ethyl alcohol in volumetric 
proportions of 60:10:6:15:5, respectively.
    (2) Spray solution. Dissolve 1 gram of ninhydrin in a mixture of 1 
milliliter of pyridine and sufficient n- butanol to make 100 
milliliters.
    (c) Preparation of spotting solutions. Prepare solutions of the 
sample and working standard, each containing 6.0 milligrams of 
bacitracin per milliliter in 1 percent disodium ethylenediamine 
tetraacetic acid in water.
    (d) Procedure. Pour the developing solvent into the glass trough on 
the bottom of the tank and onto the paper lining the walls of the tank. 
Cover and seal the tank. Allow it to equilibrate for at least 30 
minutes. Prepare a plate as follows: On a line 2.0 centimeters from the 
base of the silica gel plate, and at intervals of 2.0 centimeters, spot 
approximately 1.0 microliter of the standard solution to points 1 and 3. 
When these spots are dry, apply approximately 1.0 microliter of sample 
solution to points 2 and 3. After all spots are thoroughly dry, place 
the base of the silica gel plate directly into the glass trough in the 
chromatography tank. Cover and seal the tank. Allow the solvent front to 
travel approximately 13 centimeters from the starting line. Remove the 
plate from the tank, and allow it to air dry. After the plate is dry, 
spray lightly with the spray solution. The plate may take 1 hour or more 
to develop at room temperature. The development may be speeded up by 
warming the plate in a 110 deg. C oven.
    (e) Evaluation. The sample and standard should have spots of 
corresponding Rf value (approximately 0.26) and standard and 
sample combined should appear as a single spot of corresponding Rf 
value.
[42 FR 27228, May 27, 1977]



Sec. 436.320  Ferric chloride colorimetric assay.

    (a) Reagents. (1) 1N hydrochloric acid.
    (2) 0.01N hydrochloric acid.
    (3) Ferric chloride stock solution. Quickly weigh (very hygroscopic) 
5.0 grams of FeCl36H2O into a 100-
milliliter beaker. Add approximately 10 milliliters of 1N hydrochloric 
acid and stir to dissolve. Quantitatively transfer to a 50-milliliter 
glass-stoppered amber volumetric flask and make up to volume with water.
    (4) Ferric chloride working reagent. Pipette 10.0 milliliters of 
ferric chloride stock solution into a 2-liter volumetric flask, add 20 
milliliters 1N hydrochloric acid, and bring to volume with water. Check 
the pH; it should be between 2.0 and 2.1.
    (b) Standard solution. Accurately weigh approximately 50 milligrams 
of the working standard of the antibiotic to be tested and dissolve with 
25 milliliters of 0.1N hydrochloric acid. Quantitatively transfer to a 
250-milliliter volumetric flask and dilute to volume with distilled 
water. Keep in a glass-stoppered flask and store under refrigeration. 
Discard solution after 7 days.
    (c) Sample solution. Accurately weigh approximately 50 milligrams of 
the sample and dissolve with 25 milliliters of 0.1N hydrochloric acid. 
Quantitatively transfer to a 250-milliliter volumetric flask and dilute 
to volume with distilled water.
    (d) Procedure. Pipette exactly 10.0 milliliters of the standard 
solution and of the sample solution into separate test tubes. To each 
tube add exactly 10 milliliters of ferric chloride working reagent, mix, 
and allow to stand 15 minutes. Determine the absorbance of each solution 
at 490 nanometers in a suitable spectrophotometer against a blank 
prepared from 10.0 milliliters of

[[Page 413]]

0.01N hydrochloric acid and 10.0 milliliters of ferric chloride working 
reagent.
    (e) Estimation of potency. Calculate the potency as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.044
    
[43 FR 11154, Mar. 17, 1978; 43 FR 34456, Aug. 4, 1978]



Sec. 436.321  Griseofulvin gas liquid chromatography.

    (a) Equipment. Gas chromatograph equipped with an electronic 
integrator and with a flame ionization detector: Hewlett Packard 7600 or 
equivalent.
    (b) Reagents. (1) Chloroform, reagent grade.
    (2) Internal standard solution: Prepare a solution containing 1.0 
milligram of tetraphenylcyclopentadienone per milliliter in chloroform.
    (c) Typical conditions--(1) Column. 1.2 meters by 4 millimeters ID, 
glass, packed with 1 percent OV-17 on Gas Chrom Q (100/120 mesh), or 
equivalent.
    (2) Temperatures. Column 245 deg. C; detector 260 deg. C; injection 
port 260 deg. C.
    (3) Carrier gas. Helium approximately 60 millimeters per minute and 
40 pounds per square inch (1.7 kilograms per square centimeter).
    (4) Detector. Hydrogen flame ionization-hydrogen at 12 pounds per 
square inch (0.5 kilogram per square centimeter), air at 34 pounds per 
square inch (1.43 kilograms per square centimeter).
    (5) Sensitivity. Adjusted to obtain peak heights greater than 50 
percent full scale deflection.
    (d) Preparation of griseofulvin sample and working standard 
solutions. Accurately weigh approximately 40 milligrams of both the 
sample and the working standard into separate 25-milliliter volumetric 
flasks. Add sufficient internal standard solution to dissolve the 
contents of each flask with vigorous mixing and then dilute to volume 
with internal standard solution and mix. Proceed as directed in 
paragraph (e) of this section.
    (e) Procedure. Inject 1.0 microliter of this solution into the gas 
chromatograph. Use the typical conditions and materials listed in 
paragraphs (a), (b), and (c) of this section. The resolution of the 
peaks should be complete. The griseofulvin peak will elute before the 
internal standard peak. Calculate the griseofulvin content as directed 
in paragraph (f) of this section.
    (f) Calculations. Calculate the griseofulvin content of the sample 
as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.045

where:
Ru=Area of the griseofulvin sample peak (at a retention time 
equal to that observed for the griseofulvin standard)/Area of the 
internal standard peak;
Rs=Area of the griseofulvin working standard peak/Area of the 
internal standard peak;
Ws=Weight of the griseofulvin working standard in milligrams;
Wu=Weight of the sample in milligrams;
f=Potency of the griseofulvin working standard in micrograms per 
milligram.
[44 FR 20660, Apr. 6, 1979]



Sec. 436.322  High-pressure liquid chromatographic assay for anthracycline antibiotics.

    (a) Equipment. A suitable high-pressure liquid chromatograph, such 
as a Waters Associates Model 244 \1\ or equivalent equipped with:
---------------------------------------------------------------------------

    \1\ Available from Waters Associates, Inc., Maple St., Milford, 
Mass. 10757.
---------------------------------------------------------------------------

    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;

[[Page 414]]

    (4) A suitable recorder of at least 25.4 centimeter deflection;
    (5) A suitable integrator;
    (6) A 30-centimeter column having an inside diameter of 4.6 
millimeters and packed with a suitable reverse phase packing such as: 
Waters Associates, Micro-Bondapak C18.\1\
    (b) Reagents. (1) Solvent mixture: Water: acetonitrile (69:31).
    (2) Mobile phase: Water: acetonitrile (69:31) adjusted to pH 2 with 
phosphoric acid. Filter the mobile phase through a suitable glass fiber 
filter or equivalent that is capable of removing particulate 
contamination to 1 micron in diameter. Degas the mobile phase just prior 
to its introduction into the chromatograph pumping system.
    (3) Internal standard solution: Prepare a 2.0-milligram-per-
milliliter solution of 2-naphthalenesulfonic acid in the solvent 
mixture.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 1.5 milliliters per minute. Use a detector 
sensitivity setting that gives a peak height for the reference standard 
that is at least 50 percent of scale. The minimum between peaks must be 
no more than 2 millimeters above the initial baseline.
    (d) Procedure. Use the standard and sample solutions prepared as 
directed in the individual monographs for the drug being tested. Use the 
equipment, reagents, and operating conditions listed in paragraphs (a), 
(b), and (c) of this section. Inject 5 microliters of the standard 
solution into the chromatograph. Allow an elution time sufficient to 
obtain satisfactory separation of expected components (ordinarily this 
time is 20 minutes). After separation of the standard solution has been 
completed, inject 5 microliters of the sample solution into the 
chromatograph and repeat the procedure described for the standard 
solution. The elution order is: Void volume, internal standard, 
doxorubicin, dihydrodaunomycin, daunomycin, adriamycinone, 
dihydrodaunomycinone, bromodaunomycin, daunomycinone, and bis-
anhydrodaunomycinone.
    (e) Calculations. Calculate the anthracycline content as directed in 
the individual monograph for the drug being tested.
[43 FR 44836, Dec. 29, 1978]



Sec. 436.323  Continuous flow thin layer chromatography identity test for cefamandole nafate.

    (a) Equipment--(1) Chromatography tank. Use a rectangular tank 
approximately 23 centimeters long, 23 centimeters high, and 9 
centimeters wide equipped with a glass solvent trough in the bottom.
    (2) Plates. Use a 20 x 20 centimeter thin-layer chromatography plate 
coated with silica gel G or equivalent to a thickness of 250 
micrometers.
    (3) Cover. A stainless steel cover with a slot measuring 21 x 0.6 
centimeters, cut in the front edge.
    (4) Supporting platform. A platform that can be placed in the bottom 
of the chromatography tank so that the solvent trough is elevated about 
3.75 centimeters.
    (b) Reagents--(1) Developing solvent. Mix n- butanol, glacial acetic 
acid, and water in volumetric proportions of 4:1:1, respectively.
    (2) Spray solution. Mix starch iodide solution, glacial acetic acid, 
and 0.1 N iodine test solution, U.S.P. in volumetric proportions of 
50:3:1. Prepare the starch iodide solution by mixing starch iodide paste 
test solution, U.S.P. and water in volumetric proportions of 1:1.
    (c) Preparation of spotting solutions. Prepare solutions of the 
sample and working standard, each containing 1 milligram of cefamandole 
nafate per milliliter in distilled water.
    (d) Procedure. Prepare a plate as follows: On a line 2 centimeters 
from the base of the silica gel plate, and at intervals of 1 centimeter, 
spot 5 microliters each of the standard solution and the sample 
solution. In addition, prepare one spot composed of 5 microliters of the 
sample solution and 5 microliters of the standard solution. Place the 
supporting platform in the bottom of the tank and place the solvent 
trough on it, near the front of the tank. Place a piece of Whatman 3 MM 
filter paper or equivalent, measuring 20 x 3 centimeters and folded in 
half, lengthwise, over the front edge of the tank to form a cushion and 
drying

[[Page 415]]

wick for the plate. Place the plate in the solvent trough with the 
coated side toward the front of the tank and leaning against the filter 
paper at the top. Pour the developing solvent into the trough and bottom 
of the tank. Cover the tank. The plate should extend approximately 1 
centimeter beyond the top of the tank and through the slot in the cover. 
Seal all the openings in the tank with masking tape, except where the 
plate leans against the filter paper. Remove the plate from the tank 
after 4 hours. Allow the plate to air dry and then heat it in an oven 
for 15 minutes at 110 deg. C. Remove the plate from the oven and 
immediately spray it with the spray solution. The compound appears as a 
white spot on a purple background.
    (e) Evaluation. The sample and standard should have traveled the 
same distance from the origin, and the combined standard and sample 
should appear as a single spot that has traveled the same distance as 
the sample and standard individually.
[44 FR 20664, Apr. 6, 1979]



Sec. 436.324  Polarographic analysis of cefamandole.

    (a) Equipment--(1) Polarograph. Use a polarograph equipped with a 
dropping mercury indicating electrode, a platinum auxilliary electrode, 
and a saturated calomel reference electrode, such as Princeton Applied 
Research Model 174 1 or equivalent.
---------------------------------------------------------------------------

    1 Available from Princeton Applied Research Corporation, 
P.O. Box 2565, Princeton, NJ 08540.
---------------------------------------------------------------------------

    (2) X-Y plotter. Use a suitable X-Y plotter, such as Houston 
Omnigraphic Model 2200-3-3 2 or equivalent.
---------------------------------------------------------------------------

    2 Available from Houston Instrument, 8500 Cameron Road, 
Austin, TX 78753.
---------------------------------------------------------------------------

    (3) Nitrogen. Use a nitrogen tank equipped with a pressure-reducing 
regulator and a filter to remove traces of oxygen, such as an oxisorb 
filter 1 or equivalent.
    (b) Reagent. pH 2.3 Buffer: Dissolve 3.6 grams of dibasic sodium 
phosphate, 39.4 grams of citric acid, and 70.8 grams of potassium 
chloride in sufficient distilled water to make 1 liter.
    (c) Operating conditions--(1) Operating mode: Differential pulse.
    (2) Scan range: -0.3 volt to -1.05 volts.
    (3) Scan rate: -2 millivolts per second.
    (4) Sensitivity: 10 to 20 microamperes or equivalent to keep peak on 
scale.
    (5) Mercury drop time: 1 second per drop.
    (6) Modulation amplitude: 25 millivolts.
    (7) Display direction: +
    (8) Damping: None.
    (d) Preparation of sample and working standard solutions. Use the 
cefamandole lithium working standard. Accurately weigh approximately 12 
milligrams of sample or working standard into a 50-milliliter volumetric 
flask. Dissolve the sample or working standard in 4 milliliters of 
distilled water. Immediately prior to polarography, add 30 milliliters 
of pH 2.3 buffer, dilute to volume with distilled water, and mix.
    (e) Procedure. Transfer a portion of the sample or working standard 
solution to the polarographic cell. Pass a stream of nitrogen through 
the solution for 5 minutes to remove the dissolved oxygen. After 5 
minutes, disperse the nitrogen above the sample. Start the mercury 
dropping from the mercury dropping electrode, and, using the operating 
conditions described in paragraph (c) of this section, record the 
polarogram. Compare the polarogram of the sample to that of the working 
standard.
    (f) Calculations. Calculate the potency of cefamandole as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.046
    
where:
    A=The peak height of the sample;

[[Page 416]]

    B=The peak height of the working standard.

    The peak height is obtained from the polarogram by measuring the 
vertical distance from the peak to the baseline of the sample or working 
standard.
[44 FR 20664, Apr. 6, 1979, as amended at 47 FR 20756, May 14, 1982]



Sec. 436.325  High pressure liquid chromatography assay for vidarabine.

    (a) Equipment. A suitable high pressure liquid chromatograph, such 
as a Waters Associates Model 244 \1\ or equivalent, equipped with:
---------------------------------------------------------------------------

    1 Available from Waters Associates, Inc., Maple St., 
Milford, MA 10757.
---------------------------------------------------------------------------

    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (4) A suitable recorder of at least 25.4 centimeter deflection;
    (5) A 30-centimeter column having an inside diameter of 4 
millimeters and packed with a suitable octadecyl bonded silica phase 
packing such as Waters Associates, Micro-Bondapak C18.\1\
    (b) Mobile phase. (1) Transfer 2.2 grams of sodium dioctyl 
sulfosuccinate and 10 milliliters of glacial acetic acid to a 1-liter 
volumetric flask. Dissolve with 500 milliliters of methanol, dilute to 
volume with distilled water, and mix. Filter the mobile phase through a 
suitable glass fiber filter or equivalent that is capable of removing 
particulate contamination to 1 micron in diameter.
    (2) De-gas the mobile phase just before its introduction into the 
chromatograph pumping system.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 1.5 milliliters per minute. Use a detector 
sensitivity setting that gives a peak height for the reference standard 
that is at least 50 percent of scale. The minimum between peaks must be 
no more than 2 millimeters above the initial baseline.
    (d) Preparation of sample and working standard solutions. Accurately 
weigh approximately 24 milligrams of sample or working standard into a 
200-milliliter volumetric flask. Add about 150 milliliters of distilled 
water and heat on a steam bath for 10 minutes. Shake until all the 
powder is dissolved. Cool to room temperature and dilute to volume with 
distilled water.
    (e) Procedure. Using the equipment, mobile phase, and operating 
conditions listed in paragraphs (a), (b), and (c) of this section, 
inject 10 microliters of the sample or working standard solution 
prepared as directed in paragraph (d) of this section into the 
chromatograph. Allow an elution time sufficient to obtain satisfactory 
separation of expected components. The elution order is void volume, 9-
-D-arabinofuranosylhypoxanthine (if present), vidarabine, and 
adenine (if present).
    (f) Calculations. Calculate the vidarabine content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.047
    
where:
A=Area of the vidarabine sample peak (at a retention time equal to that 
observed for the standard);
B= Area of the standard peak;
Ws=Weight of standard in milligrams;
Wu=Weight of sample in milligrams; and
f=Potency of standard in micrograms per milligram.
[44 FR 30334, May 25, 1979, as amended at 47 FR 23708, June 1, 1982]



Sec. 436.326  Thin layer chromatographic identity test for cefoxitin sodium.

    Using the sample solution prepared as described in the section for 
the antibiotic drug to be tested, proceed as described in paragraphs 
(a), (b), (c), (d), and (e) of this section.
    (a) Equipment--(1) Chromatography tank. A rectangular tank, 
approximately 23 centimeters long, 23 centimeters high, and 9 
centimeters wide, equipped with a glass solvent trough in the bottom and 
a tight-fitting cover for the top. Line the inside walls of the tank 
with Whatman 3 MM, chromatographic paper or equivalent.
    (2) Plates. Use a 20 x 20 centimeter thin layer chromatography plate 
coated with silica gel G or equivalent to a thickness of 250 
micrometers.
    (b) Developing solvent. Mix ethyl acetate, pyridine, n-butanol, 
acetic acid,

[[Page 417]]

and water in volumetric proportions of 42:21:21:6:10, respectively.
    (c) Spray solution. Immediately before use, mix 100 milliliters of a 
1-percent ferric chloride solution in 1 percent hydrochloric acid with 
100 milliliters of a 1-percent potassium ferricyanide solution and 75 
milliliters of methanol.
    (d) Preparation of working standard solution. Prepare a solution 
containing approximately 2.5 milligrams per milliliter of cefoxitin 
working standard in distilled water.
    (e) Procedure. Pour the developing solvent into the glass trough on 
the bottom of the tank and onto the paper lining the walls of the tank. 
Cover and seal the tank. Allow it to equilibrate for 1 hour. Prepare a 
plate as follows: On a line 2 centimeters from the base of the silica 
gel plate, and at intervals of 2 centimeters, spot 10 microliters each 
of the standard solution and the sample solution. After all spots are 
thoroughly dry, place the silica gel plate directly into the glass 
trough. Cover and seal the tank. Allow the solvent front to travel about 
15 centimeters from the starting line. Remove the plate from the tank 
and heat it for 1 hour at 60 deg. C in a circulating air oven. Remove 
the plate from the oven and allow it to cool at room temperature. Apply 
the spray solution and allow it to air dry. After approximately 15 
minutes, the compound appears as a blue spot on a yellow-green 
background.
    (f) Evaluation. Measure the distance the solvent front traveled from 
the starting line and the distance the spots are from the starting line. 
Calculate the Rf value by dividing the latter by the former. 
The sample and standard should have spots of corresponding Rf 
values.
[44 FR 10373, Feb. 20, 1979, as amended at 49 FR 2242, Jan. 19, 1984]



Sec. 436.327  Thin layer chromatographic identity test for cyclacillin.

    (a) Equipment--(1) Chromatography tank. Use a rectangular tank 
approximately 23 x 23 x 9 centimeters, with a glass solvent trough on 
the bottom and a tight-fitting cover.
    (2) Plates. Use 20 x 20 centimeter thin layer chromatography plates 
coated with Silica Gel G or equivalent to a thickness of 250 microns.
    (b) Reagents--(1) Developing solvent. One percent ammonium formate 
aqueous solution.
    (2) Spray solution. Dilute starch iodide paste TS (U.S.P. XIX) with 
an equal volume of water. Mix diluted starch iodide paste, glacial 
acetic acid, and 0.1N iodine in volumetric proportions of 50:3:1, 
respectively.
    (c) Assay solutions--(1) Preparation of working standard solution. 
Accurately weigh an amount of cyclacillin working standard and dissolve 
the material with sufficient 0.1N sodium hydroxide to obtain a solution 
containing 1 milligram per milliliter. Allow the solution to stand for 
15 minutes before using.
    (2) Preparation of sample solution. Using the sample solution 
prepared as described in the section for the antibiotic to be tested, 
proceed as described in paragraphs (d) and (e) of this section.
    (d) Procedure. Pour the developing solvent into the glass trough on 
the bottom of the tank. Cover and seal the tank. Allow it to 
equilibrate. Prepare a plate as follows: On a line 2 centimeters from 
the base of the thin layer chromatography plate and at intervals of 2 
centimeters, spot 5 microliters each of the working standard solution 
and sample solution. Dry the spots thoroughly with a stream of dry air. 
Place the plate in the trough in the chromatography tank. Cover and seal 
the tank. Allow the solvent front to travel about 15 centimeters from 
the starting line and then remove the plate from the tank. Dry the plate 
by heating for 30 minutes at 80 deg. C in a circulating air oven. 
Visualize the spots by applying the spray solution.
    (e) Evaluation. Measure the distance the solvent front traveled from 
the starting line, and the distance the spots are from the starting 
line. Divide the latter by the former to calculate the Rf 
value. The sample and standard should appear as white spots against a 
blue background at an Rf of approximately 0.6. The test is 
satisfactory if the Rf value of the sample compares with that 
of the working standard.
[46 FR 2981, Jan. 13, 1981, as amended at 49 FR 2242, Jan. 19, 1984]

[[Page 418]]



Sec. 436.328  High pressure liquid chromatographic assay for sulfisoxazole acetyl content.

    (a) Equipment. A suitable high pressure liquid chromatograph, such 
as a Waters Associates Model 244 \1\ or equivalent equipped with:
---------------------------------------------------------------------------

    \1\ Available from: Waters Associates, Inc., Maple Street, Milford, 
MA 10757.
---------------------------------------------------------------------------

    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (4) A suitable recorder of at least 25.4 centimeter deflection;
    (5) A 30-centimeter column having an inside diameter of 4.0 
millimeters and packed with a suitable reverse phase packing such as: 
Waters Associates, Micro-Bondapak C18; \1\ and
    (6) A suitable integrator.
    (b) Reagents--(1) Mobile phase. Mix acetonitrile (high pressure 
liquid chromatography grade): water (40:60). Filter the mobile phase 
through a suitable glass fiber filter or equivalent which is capable of 
removing particulate contamination to 1 micron in diameter. De-gas the 
mobile phase just prior to its introduction into the chromatograph 
pumping system.
    (2) Internal standard solution. Dissolve 0.33 milligram of 
benzanilide per milliliter in acetonitrile (high pressure liquid 
chromatography grade). Filter the solution through a suitable glass 
fiber filter or equivalent which is capable of removing particulate 
contamination to 1 micron in diameter.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 1.2 milliliters per minute. Use a detector 
sensitivity setting that gives a peak height for reference standard that 
is at least 50 percent of scale. The minimum between peaks must be no 
more than 2 millimeters above the baseline.
    (d) Preparation of the working standard and sample solutions--(1) 
Working standard solution. Prepare a solution containing 1.0 milligram 
per milliliter of sulfisoxazole acetyl in the internal standard 
solution.
    (2) Sample solution. Reconstitute the sample as directed in the 
labeling. Allow to stand for 1 hour. Shake gently and transfer 5.0 
milliliters of the sample to a separatory funnel. Extract the suspension 
three times with 75-milliliter portions of chloroform. Collect the 
chloroform layers in a 250-milliliter volumetric flask. Dilute the flask 
to volume with chloroform and mix. Filter a portion of the solution 
through a suitable glass fiber filter or equivalent which is capable of 
removing particulate contamination to 1 micron in diameter. Transfer a 
4.0-milliliter aliquot of the filtrate into a 25-milliliter glass-
stoppered flask and evaporate to dryness under a stream of dry air. 
Dissolve the residue in 10.0 milliliters of the internal standard 
solution, stopper, and mix.
    (e) Procedure. Using the equipment, reagents, and operating 
conditions listed in paragraphs (a), (b), and (c) of this section, 
inject 5 microliters of sample or working standard solution prepared as 
described in paragraph (d) of this section, into the chromatograph. 
Allow an elution time sufficient to obtain satisfactory separation of 
expected components. The elution order is void volume, sulfisoxazole 
acetyl and benzanilide.
    (f) Calculations. Calculate the sulfisoxazole content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.048
    
where:
    A=Area of sample peak (at a retention time equal to that of the 
standard) divided by the area of the internal standard peak;
    B=Area of the standard peak divided by the area of the internal 
standard peak;

[[Page 419]]

    0.864=The molecular weight of sulfisoxazole divided by the molecular 
weight of sulfisoxazole acetyl.
[46 FR 2990, Jan. 13, 1981]



Sec. 436.329  High-pressure liquid chromatographic assay for meclocycline.

    (a) Equipment. A suitable high-pressure liquid chromatograph, such 
as a Waters Associates Model 244 \1\ or equivalent equipped with:
---------------------------------------------------------------------------

    \1\ Available from: Waters Associates, Inc., Maple St., Milford, MA 
10757.
---------------------------------------------------------------------------

    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 340 nanometers;
    (4) A suitable recorder of at least 25.4 centimeter deflection;
    (5) A suitable integrator;
    (6) A column approximately 25 centimeters in length having an inside 
diameter of approximately 4 millimeters and packed with a suitable 
reverse-phase packing such as: 10 micrometer silica gel particles bonded 
to octadecyl silane, Vydac 201 TP Reverse Phase \2\ or equivalent.
---------------------------------------------------------------------------

    \2\ Available from: The Separations Group, 16640 Spruce St., 
Hesperia, CA 92345.
---------------------------------------------------------------------------

    (b) Reagents--(1) 0.001M Ammonium (ethylenedinitrilo) tetraacetate. 
Moisten 293 milligrams of (ethylenedinitrilo) tetraacetic acid with 1 
milliliter of methanol and dissolve in 7 milliliters of concentrated 
ammonium hydroxide. Dilute to 900 milliliters with distilled water, 
adjust the pH to 6.6 with glacial acetic acid, and dilute to 1,000 
milliliters with distilled water.
    (2) Mobile phase. Mix 150 milliliters of tetrahydrofuran (high-
pressure liquid chromatography grade) with 850 milliliters of 0.001M 
ammonium (ethylenedinitrilo) tetraacetate. Filter the mobile phase 
through a suitable glass fiber filter or equivalent that is capable of 
removing particulate contamination to 1 micron in diameter. Degas the 
mobile phase just prior to its introduction into the chromatograph 
pumping system.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 0.8 milliliter per minute. Use a detector 
sensitivity setting that gives a peak height for the reference standard 
that is at least 50 percent of scale. The minimum between peaks must be 
no more than 2 millimeters above the initial baseline.
    (d) Preparation of sample and working standard solutions. Accurately 
weigh an amount of sample or working standard equivalent to 
approximately 25 milligrams of meclocycline into a 50-milliliter 
volumetric flask. Dissolve and dilute to volume with methanol and mix. 
Transfer exactly 3.0 milliliters of this solution to a 25-milliliter 
volumetric flask, dilute to volume with mobile phase, and mix.
    (e) Procedure. Using the equipment, reagents, and operating 
conditions listed in paragraphs (a), (b), and (c) of this section, 
inject 10 microliters of the sample or working standard solution 
prepared as described in paragraph (d) of this section into the 
chromatograph. Allow an elution time sufficient to obtain satisfactory 
separation of expected components. The elution order is void volume, 
oxytetracycline (if present), demeclocycline (if present), methacycline 
(if present), and meclocycline.
    (f) Calculations. Calculate the meclocycline content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.049
    
where:
A= Area or peak height of the sample peak (at a retention time equal to 
          that observed for the standard);
B= Area or peak height of the standard peak.
[46 FR 3836, Jan. 16, 1981]

[[Page 420]]



Sec. 436.330  Thin layer chromatographic identity test for bacampicillin.

    (a) Equipment--(1) Chromatography tank. Use a rectangular tank 
approximately 23  x  23  x  9 centimeters, with a glass solvent trough 
on the bottom and a tight-fitting cover, lined with Whatman's 3MM 
chromatographic paper (0.3 millimeter) or equivalent.
    (2) Plates. Use 20  x  20 centimeter thin layer chromatography 
plates coated with Silica Gel 60F 254 or equivalent to a thickness of 
250 microns.
    (b) Reagents--(1) Developing solvent. Mix methylene chloride, 
chloroform, and 95 percent ethyl alcohol in volumetric proportions of 
100:10:10, respectively.
    (2) Spray solution. Dissolve 1 gram of ninhydrin in 100 milliliters 
of n-butanol and add 1 milliliter of pyridine.
    (c) Spotting solutions--(1) Preparation of working standard 
solution. Dissolve and dilute a weighed amount of the bacampicillin 
hydrochloride working standard with sufficient 95 percent ethyl alcohol 
to obtain a solution containing 2 milligrams per milliliter.
    (2) Preparation of sample solution. Dissolve and dilute a weighed 
amount of the sample with sufficient 95 percent ethyl alcohol to obtain 
a solution containing 2 milligrams per milliliter. Proceed as described 
in paragraphs (d) and (e) of this section.
    (d) Procedure. Pour the developing solvent into the glass trough on 
the bottom of the tank and onto the paper lining the walls of the tank. 
Cover and seal the tank. Allow it to equilibrate for one hour. Prepare a 
plate as follows: On a line 2.5 centimeters from the base of the thin 
layer chromatography plate and at intervals of 2.0 centimeters, spot 5 
microliters of the working standard solution to positions 1 and 3. When 
these spots are dry, apply 5 microliters of the sample solution to 
points 2 and 3. After all the spots are thoroughly dry, place the plate 
into the trough in the bottom of the tank. Cover and tightly seal the 
tank, allow the solvent front to travel about 15 centimeters from the 
starting line (about 30 minutes) and then remove the plate from the 
tank. Air dry the plate. Visualize the spots by spraying with spray 
solution and heating in an oven at 100 deg. C for approximately 10 
minutes.
    (e) Evaluation. Measure the distance the solvent front traveled from 
the starting line, and the distance the spots are from the starting 
line. Divide the latter by the former to calculate the Rf 
value. Bacampicillin appears as a purple spot at an Rf value 
of approximately 0.52. The test is satisfactory if the Rf 
value of the sample compares with that of the working standard. The 
combined spot should appear as a single spot of corresponding Rf 
value.
[46 FR 25602, May 8, 1981, as amended at 49 FR 2242, Jan. 19, 1984]



Sec. 436.331  High-pressure liquid chromatographic assay for dactinomycin.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (4) A suitable recorder of at least 25.4-centimeter deflection;
    (5) A suitable integrator; and
    (6) A 30-centimeter column having an inside diameter of 4.0 
millimeters and packed with octadecyl silane chemically bonded to porous 
silica or ceramic microparticles, 5 micrometers to 10 micrometers in 
diameter, U.S.P. XX.
    (b) Mobile phase. Mix acetonitrile (high-pressure liquid 
chromatography grade): water (60:40). Filter the mobile phase through a 
suitable glass fiber filter or equivalent that is capable of removing 
particulate contamination to 1 micron in diameter. Degas the mobile 
phase just prior to its introduction into the chromatograph pumping 
system.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 2.5 milliliters per minute. Use a detector 
sensitivity setting that gives a peak height for the working standard 
that is at least 50 percent of scale. The minimum between peaks must be 
no more than 2 millimeters above the initial baseline.
    (d) Preparation of working standard and sample solutions--(1) 
Preparation of

[[Page 421]]

working standard solution. Prepare a solution containing 0.25 milligram 
per milliliter of dactinomycin in mobile phase.
    (2) Preparation of sample solution. Prepare the sample solution as 
described in the individual monograph for the drug being tested.
    (e) Procedure. Use the equipment, mobile phase, operating 
conditions, and working standard and sample solutions described in 
paragraphs (a), (b), (c), and (d) of this section, and proceed as 
directed in paragraph (e)(1) of this section.
    (1) System suitability test. Equilibrate and condition the column by 
passage of about 10 to 15 void volumes of mobile phase followed by two 
or more replicate injections of 10 microliters each of the working 
standard solution. Allow an elution time sufficient to obtain 
satisfactory separation of expected components after each injection. 
Record the peak responses and, calculate the relative standard deviation 
as described for system suitability tests in the U.S.P. XX General 
Chapter 621 chromatography. Proceed as directed in paragraph (e)(2) of 
this section if the minimum performance requirement for the relative 
standard deviation is not more than 1.0 percent. If the minimum 
performance requirement is not met, adjustment must be made to the 
system to obtain satisfactory operation before proceeding as described 
in paragraph (e)(2) of this section.
    (2) Determination of the chromatogram. Inject 10 microliters of the 
working standard solution into the chromatograph. Allow an elution time 
sufficient to obtain satisfactory separation of the expected components. 
After separation of the working standard solution has been completed, 
inject 10 microliters of the sample solution into the chromatograph and 
repeat the procedure described for the working standard solution.
    (f) Calculations. Calculate the dactinomycin content as described in 
the individual monograph for the drug being tested.
[49 FR 24017, June 11, 1984, as amended at 50 FR 5749, Feb. 12, 1985]



Sec. 436.332  High-pressure liquid chromatographic assay for moxalactam.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (4) A 30-centimeter column having an inside diameter of 4.0 
millimeters and packed with octadecyl silane chemically bonded to porous 
silica or ceramic microparticles, 5 to 10 micrometers in diameter, 
U.S.P. XX;
    (5) A suitable recorder of at least 25.4 centimeter deflection;
    (6) A suitable integrator.
    (b) Mobile phase. Mix 0.01M ammonium acetate:methanol (19:1). Filter 
the mobile phase through a suitable glass fiber filter or equivalent 
that is capable of removing particulate contamination to 1 micron in 
diameter. Degas the mobile phase just prior to its introduction into the 
chromatograph pumping system.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 0.5 milliliter per minute. Use a detector 
sensitivity setting that gives a peak height for the working standard 
that is at least 50 percent of scale.
    (d) Preparation of working standard solution. Transfer the contents 
of an ampoule of working standard to a tared weighing bottle. Place the 
unstoppered weighing bottle in a desiccator containing a saturated 
aqueous solution of potassium carbonate to provide an atmosphere of 42 
percent relative humidity. Allow the moisture content of the working 
standard to equilibrate for 16 hours. Determine the moisture content as 
described in Sec. 436.201 of this chapter. Equilibrated standard 
material must be kept in a closed weighing bottle and used within 36 
hours of equilibration. Dissolve approximately 50 milligrams of the 
working standard, accurately weighed and corrected for moisture, with 
sufficient distilled water to obtain a solution containing 0.5 milligram 
of

[[Page 422]]

moxalactam per milliliter. Use the prepared solution immediately.
    (e) Preparation of sample solution. Mix contents of vial thoroughly. 
Dissolve an accurately weighed portion of approximately 50 milligrams of 
sample with distilled water to obtain a concentration of 0.5 milligram 
per milliliter (estimated); also, reconstitute the sample as directed in 
the labeling. Then using a suitable hypodermic needle and syringe, 
remove all of the withdrawable contents if it is represented as a single 
dose container; or, if the labeling specifies the amount of potency in a 
given volume of the resultant preparation, remove an accurately measured 
representative portion from each container. Further dilute an aliquot of 
this solution with distilled water to obtain a concentration of 0.5 
milligram per milliliter (estimated). Use the prepared solution 
immediately.
    (f) Procedure. Using the equipment, reagents, and operating 
conditions as listed in paragraphs (a), (b), and (c) of this section, 
inject 5 microliters of the working standard solution into the 
chromatograph. Allow an elution time sufficient to obtain satisfactory 
separation of the expected components. After separation of the working 
standard solution has been completed, inject 5 microliters of the sample 
solution into the chromatograph and repeat the procedure described for 
the working standard solution.
    (g) Calculations. (1) Calculate the moxalactam content in micrograms 
per milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.050

where:
Ru=Sum of the areas of the moxalactam sample R-isomer and the 
          S-isomer peaks;
Rs=Sum of the areas of the moxalactam working standard R-
          isomer and the S-isomer peaks;
Wu=Weight of the sample in milligrams;
Ws=Weight of the moxalactam working standard in milligrams;
P=Potency of the moxalactam working standard in micrograms per 
          milligram, corrected for moisture.

    (2) Calculate the moxalactam content of the vial as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.051
    
where:
Ru=Sum of the areas of the moxalactam R-isomer and the S-
          isomer peaks;
Rs=Sum of the areas of the moxalactam working standard R-
          isomer and the S-isomer peaks;
Ws=Weight of the moxalactam working standard in milligrams;
P=Potency of the moxalactam working standard in micrograms per 
          milligram, corrected for moisture;
d=Dilution factor.

    (3) Calculate the ratio of R-isomer to S-isomer as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.052
    
[46 FR 61069, Dec. 15, 1981]



Sec. 436.333  Thin layer chromatographic identity test for moxalactam.

    (a) Equipment--(1) Chromatography tank. A rectangular tank, 
approximately 23 centimeters long, 23 centimeters high, and 9 
centimeters wide, equipped with a glass solvent trough in the bottom and 
a tight-fitting cover for the top. Line the inside walls of the tank 
with Whatman 3MM chromatographic paper or equivalent.
    (2) Plates. Use a 20 x 20 centimeter thin layer chromotagraphy plate 
coated with silca gel G or equivalent to a thickness of 250 micrometers.
    (b) Developing solvent. Mix ethyl acetate, glacial acetic acid, 
acetonitrile, and water in volumetric proportions of 42:14:14:18, 
respectively.
    (c) Preparation of spotting solutions. Prepare solutions of the 
sample and working standard, each containing 10 milligrams per 
milliliter of moxalactam in distilled water.

[[Page 423]]

    (d) Procedure. Pour the developing solvent into the glass trough on 
the bottom of the tank and onto the paper lining the walls of the tank. 
Cover and seal the tank. Allow it to equilibrate for 1 hour. Prepare a 
plate as follows: On a line 2 centimeters from the base of the silica 
gel plate, and at intervals of 2 centimeters, spot 10 microliters each 
of the standard solution and the sample solution. After all spots are 
thoroughly dry, place the silica gel plate directly into the glass 
trough. Cover and seal the tank. Allow the solvent front to travel about 
15 centimeters from the starting line. Remove the plate from the tank 
and air dry. Expose the plate to iodine vapors for 40 minutes. 
Immediately circumscribe all spots using a suitable marker.
    (e) Evaluation. Measure the distance the solvent front traveled from 
the starting line and the distance the spots are from the starting line. 
Calculate the Rf value by dividing the latter by the former. 
The sample and standard should have spots of corresponding Rf 
values and intensity.
[46 FR 61070, Dec. 15, 1981, as amended at 49 FR 2242, Jan. 19, 1984]



Sec. 436.334  High-pressure liquid chromatographic assay for piperacillin.

    (a) Equipment. A high-pressure liquid chromatograph equipped with:
    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (4) A suitable recorder of at least 25.4-centimeter deflection;
    (5) A suitable integrator;
    (6) A 25-centimeter column having an inside diameter of 4.6 
millimeters and packed with octadecyl silane chemically bonded to porous 
silica or ceramic microparticles, 5 to 10 micrometers in diameter 
(United States Pharmacopeia XX).
    (b) Reagents. (1) 0.2M monobasic sodium phosphate: Dissolve 27.60 
grams of monobasic sodium phosphate with sufficient water to make 1,000 
milliliters.
    (2) 10 percent tetrabutylammonium hydroxide in water.
    (3) Ampicillin-piperacillin solution: Dissolve and dilute 25 
milligrams of ampicillin and 5 milligrams of piperacillin monohydrate 
with sufficient mobile phase to obtain 100 milliliters, and mix.
    (c) Mobile phase. Methanol:water:0.2M monobasic sodium phosphate:10 
percent tetrabutylammonium hydroxide (450 : 447 : 100:3) adjusted to pH 
5.5 plus-minus0.02 with phosphoric acid. The concentration of 
reagents may be varied to obtain acceptable operation of the system. De-
gas the mobile phase just prior to its introduction into the 
chromatograph pumping system.
    (d) Preparation of working standard and sample solutions--(1) 
Working standard solution. Place approximately 20 milligrams of the 
working standard, accurately weighed, into a 50-milliliter volumetric 
flask. Add 25 to 30 milliliters of mobile phase. Shake until dissolved. 
Dilute to volume with mobile phase.
    (2) Sample solution--(i) Micrograms per milligram. Place 
approximately 20 milligrams of the sample, accurately weighed, into a 
50-milliliter volumetric flask. Add 25 to 30 milliliters of mobile 
phase. Shake until dissolved. Dilute to volume with mobile phase.
    (ii) Milligrams per vial. Reconstitute as directed in the labeling. 
Withdraw the total contents and dilute with mobile phase to a 
concentration of 0.4 milligram of piperacillin per milliliter.
    (e) Procedure. Use the equipment, reagents, mobile phase, and 
working standard and sample solutions described in paragraphs (a), (b), 
(c), and (d) of this section and proceed as directed in paragraph (e) of 
this section.
    (1) Systems suitability test. Chromatograph three replicate samples 
of ampicillin-piperacillin solution as directed in paragraph (e)(2) of 
this section. Allow an elution time sufficient to obtain satisfactory 
separation of expected components after each injection. Record the peak 
responses and calculate the resolution factor as described for system 
suitability tests in the United States Pharmacopeia XX General Chapter 
621 for gas chromatography. The resolution factor between ampicillin and 
piperacillin is not

[[Page 424]]

less than 15. If the resolution factor does not meet this limit, 
adjustments must be made to the system to obtain satisfactory operation 
before proceeding as described in paragraph (e)(2) of this section.
    (2) Determination of the chromatogram. Operate the high-pressure 
liquid chromatograph at ambient temperature at a flow rate of one 
milliliter per minute. Use a detector sensitivity setting that gives a 
peak height for the reference standard that is at least 50 percent of 
scale. Purge the column with mobile phase until a steady baseline is 
established. Inject 10 microliters of the working standard solution into 
the chromatograph. Allow an elution time sufficient to obtain separation 
of the expected components. After separation of the working standard 
solution has been completed, inject 10 microliters of the sample 
solution into the chromatograph and repeat the procedure described for 
the working standard solution.
    (f) Calculations--(1) Calculate the piperacillin content in 
micrograms per milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.053

where:
A=Area of the sample peak (at a retention time equal to that observed 
          for the standard);
B=Area of the standard peak.

    (2) Calculate the piperacillin content in grams per vial as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.054
    
where:
A=Area of the sample peak (at a retention time equal to that observed 
          for the standard);
B=Area of the standard peak;
d=Dilution factor.
[47 FR 15768, Apr. 13, 1982; 47 FR 33493, Aug. 3, 1982]



Sec. 436.335  High-pressure liquid chromatographic assay for chloramphenicol palmitate.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 280 nanometers;
    (4) A suitable recorder of at least 25.4-centimeter deflection;
    (5) A suitable integrator; and
    (6) A 30-centimeter column having an inside diameter of 4.0 
millimeters and packed with octadecyl silane chemically bonded to porous 
silica or ceramic microparticles, 5 to 10 micrometers in diameter, 
U.S.P. XX.
    (b) Mobile phase. Mix methanol:water:glacial acetic acid (170:30:1). 
Degas the mobile phase just prior to its introduction into the 
chromatograph pumping system.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 2.0 milliliters per minute. Use a detector 
sensitivity setting that gives a peak height for the reference standard 
that is at least 50 percent of scale. The minimum between peaks must be 
no more than 2 millimeters above the initial baseline.
    (d) Preparation of sample and working standard solutions. Accurately 
weigh approximately 65 milligrams of sample

[[Page 425]]

or chloramphenicol palmitate working standard each into a 50-milliliter 
volumetric flask. Add approximately 35 milliliters of methanol and 1 
milliliter of glacial acetic acid. Place in an ultrasonic bath for 10 
minutes and dilute to volume with methanol.
    (e) Procedure. Using the equipment, mobile phase, and operating 
conditions listed in paragraphs (a), (b), and (c) of this section, 
inject 10 microliters of the working standard solution into the 
chromatograph. Allow an elution time sufficient to obtain satisfactory 
separation of expected components. After separation of the working 
standard solution has been completed, inject 10 microliters of the 
sample solution into the chromatograph and repeat the procedure 
described for the working standard solution.
    (f) Calculations. Calculate the chloramphenicol content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.055
    
where:
A=Area of chloramphenicol palmitate sample peak (at a retention time 
equal to that observed for the standard);
B=Area of the working standard peak;
Ws=Weight of standard in milligrams;
Wu=Weight of sample in milligrams; and
f=Micrograms of chloramphenicol activity per milligram of 
chloramphenicol palmitate working standard.
[49 FR 6091, Feb. 17, 1984]



Sec. 436.336  Thin layer chromatographic identity test for azlocillin.

    (a) Equipment--(1) Chromatography tank. A rectangular tank, 
approximately 23 centimeters long, 23 centimeters high, and 9 
centimeters wide, equipped with a glass solvent trough in the bottom and 
a tight-fitting cover for the top.
    (2) Iodine vapor chamber. A rectangular tank approximately 23 
centimeters long, 23 centimeters high, and 9 centimeters wide, with a 
suitable cover, containing iodine crystals.
    (3) Plates. Use 20 x 20 centimeter thin layer chromatography plates 
coated with Silica Gel G or equivalent to a thickness of 250 microns.
    (b) Reagents--(1) Buffer. Dissolve 9.078 grams of potassium 
phosphate, monobasic (KH2PO4) in sufficient 
distilled water to make 1,000 milliliters (solution A). Dissolve 17.88 
grams of sodium phosphate, dibasic, heptahydrate 
(Na2HPO4.7H2 O) in sufficient distilled 
water to make 1,000 milliliters (solution B). Place 12.1 milliliters of 
solution B into a 100-milliliter volumetric flask and dilute to volume 
with solution A.
    (2) Developing solvent. Place 50 milliliters of n-butyl acetate, 9 
milliliters of n-butanol, 25 milliliters of glacial acetic acid, and 15 
milliliters of buffer into a separatory funnel. Shake well and allow the 
layers to separate. Discard the lower phase and use the upper phase as 
the developing solvent.
    (c) Preparation of spotting solutions. Prepare solutions of the 
sample and working standard, each containing 20 milligrams of azlocillin 
per milliliter in distilled water.
    (d) Procedure. Pour developing solvent into the glass trough on the 
bottom of the chromatography tank to a depth of about 1 centimeter. Use 
the chamber immediately. Prepare plate as follows: Apply spotting 
solutions on a line 2.5 centimeters from the base of the silica gel 
plate and at points 2.0 centimeters apart. Apply approximately 10 
microliters of the working standard solution to points 1 and 3. When 
these spots are dry, apply approximately 10 microliters of sample 
solution to points 2 and 3. Place spotted plate in a desiccator until 
solvent has evaporated from spots. Place the plate into the glass trough 
at the bottom of the chromatography tank. Cover the tank. Allow the 
solvent to travel about 15 centimeters from the starting line. Remove 
the plate from the tank and allow to air dry. Warm the iodine vapor 
chamber to vaporize the iodine crystals and place the dry plate in the 
iodine vapor chamber until the spots are visible, usually about 10 
minutes.
    (e) Evaluation. Measure the distance the solvent front traveled from 
the starting line and the distance the spots are from the starting line. 
Calculate the Rf value by dividing the latter by the former. 
The azlocillin sample and the standard should have spots of 
corresponding Rf values (approximately 0.4), and standard and 
sample combined should appear as a single spot for

[[Page 426]]

azlocillin. The penicilloate and penilloate of azlocillin as well as 
ampicillin appear as additional spots with Rf values of 
approximately 0.15, 0.3, and 0.25, reectively.
[47 FR 53348, Nov. 26, 1982]



Sec. 436.337  High-pressure liquid chromatographic assay for cephradine.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 8 millimeters;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (4) A suitable recorder that is compatible with the detector output;
    (5) A suitable integrator (optional); and
    (6) A 25-centimeter column having an inside diameter of 4.6 
millimeters and packed with octadecyl silane chemically bonded to porous 
silica or ceramic microparticles, 10 micrometers in diameter, U.S.P. XX.
    (b) Reagents. (1) 4 percent glacial acetic acid.
    (2) 3.86 percent sodium acetate.
    (c) Mobile phase. 4 percent glacial acetic acid:3.86 percent sodium 
acetate:methanol:distilled water (3:15:200:782). Filter the mobile phase 
through a suitable glass fiber filter or equivalent that is capable of 
removing particulate contamination to 1 micron in diameter. Degas the 
mobile phase prior to its introduction into the chromatograph pumping 
system. The distilled water:methanol ratio may be varied to obtain 
acceptable operation of the system.
    (d) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 1.2 milliliters per minute. Use a detector 
sensitivity setting that gives a peak height for the cephradine in the 
cephradine working standard that is about 75 percent of full scale.
    (e) Preparation of working standard and sample solutions--(1) 
Preparation of cephradine working standard solution. Place an accurately 
weighed portion of the cephradine working standard into a suitably sized 
container. Add 5.0 milliliters of distilled water and place in an 
ultrasonic bath to facilitate dissolution. Dilute with a sufficient 
amount of mobile phase to obtain a solution containing 0.8 milligram of 
cephradine activity per milliliter.
    (2) Preparation of cephalexin working standard solution. Dissolve an 
accurately weighed portion of the cephalexin working standard with 
mobile phase to obtain a solution containing 0.02 milligram of 
cephalexin activity per milliliter. Place in an untrasonic bath to 
facilitate dissolution.
    (3) Preparation of sample solutions--(i) Product not packaged for 
dispensing (micrograms of cephradine per milligram). Dissolve an 
accurately weighed portion of the sample with mobile phase to obtain a 
solution containing 0.8 milligram per milliliter. Place in an ultrasonic 
bath to facilitate dissolution. Using this sample solution, proceed as 
directed in paragraph (f)(1) of this section.
    (ii) Product packaged for dispensing. Determine both micrograms of 
cephradine per milligram of the sample and milligrams of cephradine per 
container. Use separate containers for preparation of each sample 
solution as described in paragraphs (e)(3)(ii) (a) and (b) of this 
section.
    (a) Micrograms of cephradine per milligram. Dissolve an accurately 
weighed portion of the sample with mobile phase to obtain a solution 
containing 0.8 milligram per milliliter. Place in an ultrasonic bath to 
facilitate dissolution. Using this sample solution, proceed as directed 
in paragraph (f)(1) of this section.
    (b) Milligrams of cephradine per container. Reconstitute the sample 
as directed in the labeling. Then, using a suitable hypodermic needle 
and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency in a given volume of the resultant preparation, 
remove an accurately measured representative portion from each 
container. Dilute the solution thus obtained with mobile phase to obtain 
a solution containing 0.8 milligram per milliliter. Using this sample 
solution,

[[Page 427]]

proceed as directed in paragraph (f)(1) of this section.
    (f) Procedure--(1) Cephradine content. Using the equipment, 
reagents, mobile phase, and operating conditions as listed in paragraphs 
(a), (b), (c), and (d) of this section, inject 10 microliters of the 
cephradine working standard solution into the chromatograph. Allow an 
elution time sufficient to obtain satisfactory separation of the 
expected components. After separation of the working standard solution 
has been completed, inject 10 microliters of the sample solution 
prepared as described in paragraph (e)(3)(i) of this section into the 
chromatograph and repeat the procedure described for the working 
standard solution. The elution order is void volume, cephalexin, and 
cephradine. If the sample is packaged for dispensing, repeat the 
procedure for each sample solution prepared as described in paragraphs 
(e)(3)(ii) (a) and (b) of this section.
    (2) Cephalexin content. Proceed as directed in paragraph (f)(1) of 
this section, except:
    (i) Use a detector sensitivity setting that gives a peak height for 
the cephalexin in the cephalexin working standard that is about 75 
percent of full scale; and
    (ii) Use the cephalexin working standard in lieu of the cephradine 
working standard.
    (g) Calculations. (1) Calculate the micrograms of cephradine per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.011

where:
Au=Area of the cephradine peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cephradine peak in the chromatogram of the 
          cephradine working standard;
Ps=Cephradine activity in the cephradine working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of sample solution; 
          and
m= Percent moisture content of the sample.

    (2) Calculate the cephradine content of the vial as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.012
    
where:
Au=Area of the cephradine peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the chephradine peak in the Chromatogram of the 
          cephradine working standard;
Ps=Cephradine activity in the cephradine working standard 
          solution in micrograms per milliliter;
Cs=Milligrams of the standard per milliliter; and
d=Dilution factor of the sample.

    (3) Calculate the percent cephalexin content of the sample as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.056

where:
Aa=Area of the cephalexin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
Ab=Area of the cephalexin peak in the chromatogram of the 
          cephalexin working standard;
Wb=Milligrams of cephalexin per milliliter of cephalexin 
          working standard solution;
Wu=Milligrams of cephradine per milliliter of sample 
          solution;
Pb=Micrograms of cephalexin per milligram of cephalexin 
          working standard; and
m=Percent moisture content of the sample.
[49 FR 47483, Dec. 5, 1984]



Sec. 436.338  High-pressure liquid chromatographic assay for cefoperazone.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (4) A suitable recorder of at least 25.4 centimeter deflection;
    (5) A suitable integrator;
    (6) A 30-centimeter column having an inside diameter of 4.0 
millimeters and

[[Page 428]]

packed with octadecyl silane chemically bonded to porous silica or 
ceramic microparticles, 5 to 10 micrometers in diameter, United States 
Pharmacopeia XX.
    (b) Mobile phase. Mix 1.2 milliliters 1M triethylammonium acetate, 
2.8 milliliters 1M acetic acid, and 120 milliliters acetonitrile in a 
one liter flask and dilute to volume with distilled water. Filter the 
mobile phase through a suitable glass fiber filter or equivalent that is 
capable of removing particulate contamination to 1 micron in diameter. 
Degas the mobile phase just prior to its introduction into the 
chromatographic pumping system.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 2.0 milliliters per minute. Use a detector 
sensitivity setting that gives a peak height for the working standard 
that is at least 50 percent of scale.
    (d) Preparation of working standard solution. Dissolve approximately 
40 milligrams of working standard, accurately weighed, with mobile phase 
to obtain a solution containing 0.16 milligram of cefoperazone activity 
per milliliter.
    (e) Preparation of sample solutions--(1) Product not packaged for 
dispensing (micrograms of cefoperazone per milligram). Dissolve an 
accurately weighed portion of the sample with sufficient mobile phase to 
obtain a solution containing 0.16 milligram of cefoperazone activity per 
milliliter. Using this sample solution, proceed as directed in paragraph 
(f) of this section.
    (2) Product packaged for dispensing. Determine both micrograms of 
cefoperazone per milligram of the sample and milligrams of cefoperazone 
per container. Use separate containers for preparation of each sample 
solution as described in paragraph (e)(2)(i) and (ii) of this section.
    (i) Micrograms of cefoperazone per milligram. Dissolve and 
accurately weighed portion of the sample with sufficient mobile phase to 
obtain a solution containing 0.16 milligram of cefoperazone activity per 
milliliter. Using this sample solution, proceed as directed in paragraph 
(f) of this section.
    (ii) Milligrams of cefoperazone per container. Reconstitute the 
sample as directed in the labeling. Then using a suitable hypodermic 
needle and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency in a given volume of the resultant preparation, 
remove an accurately measured representative portion from each 
container. Further dilute and aliquot of this solution with mobile phase 
to a concentration of 0.16 milligram of cefoperazone activity per 
milliliter. Using this sample solution, proceed as directed in paragraph 
(f) of this section.
    (f) Procedure. Using the equipment, reagents, and operating 
conditions as listed in paragraphs (a), (b), and (c) of this section, 
inject 10 microliters of the working standard solution into the 
chromatograph. Allow an elution time sufficient to obtain satisfactory 
separation of the expected components. After separation of the working 
standard solution has been completed, inject 10 microliters of the 
sample solution prepared as described in paragraph (e)(1) of this 
section into the chromatograph and repeat the procedure described for 
the working standard solution. If the sample is packaged for dispensing, 
repeat the procedure for each sample solution prepared as described in 
paragraphs (e)(2)(i) and (ii) of this section.
    (g) Calculations--(1) Calculate the micrograms of cefoperazone per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.011

where:
Au=Area of the cefoperazone sample peak (at a retention time 
          equal to that observed for the standard);
As=Area of the cefoperazone working standard peak;
Ps=Cefoperazone activity in the cefoperazone working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of sample solution; 
          and
m= Percent moisture content of the sample.

    (2) Calculate the cefoperazone content of the vial as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.012
    

[[Page 429]]


where:
Au=Area of the cefoperazone sample peak (at a retention time 
          equal to that observed for the standard);
As=Area of the cefoperazone working standard peak;
Ps=Cefoperazone activity in the cefoperazone working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.
[48 FR 789, Jan. 7, 1983; 48 FR 7439, Feb. 22, 1983; 48 FR 28250, June 
21, 1983]



Sec. 436.339  High-pressure liquid chromatographic assay for bleomycin fractions.

    (a) Equipment. A high-pressure liquid chromatograph equipped with:
    (1) Two solvent pumps;
    (2) A solvent programmer;
    (3) A low dead volume cell 8 to 20 microliters;
    (4) A light path length of 1 centimeter;
    (5) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (6) A suitable recorder;
    (7) A suitable integrator; and
    (8) A suitable-sized column approximately 25 centimeters in length 
having an inside diameter of 4.6 millimeters and packed with octadecyl 
silane chemically bonded to porous silica or ceramic microparticles, 5 
to 10 micrometers in diameter, USP XX.
    (b) Reagents--(1) 0.005M 1-pentanesulfonic acid in 0.5 percent 
acetic acid adjusted to pH 4.3 with concentrated ammonium hydroxide. 
Filter and degas before using.
    (2) Methanol, spectrophotometric grade. Filter and degas before 
using.
    (3) Mobile phase. Adjust the solvent programmer for linear gradient 
development starting with a mixture of 0.005M 1-pentanesulfonic 
acid:methanol (9:1) and ending with a mixture of 0.005M 1-
pentanesulfonic acid:methanol (6:4) in 1 hour at a flow rate of 1.2 
milliliters per minute. Minor flow rate and gradient changes can be made 
as necessary depending on column and instrument conditions. Disodium 
ethylenediaminetetraacetic acid USP at a concentration of 0.005M may be 
added to the mobile phase if necessary for satisfactory performance.
    (c) Preparation of sample solution. Reconstitute the vial with 6 
milliliters of deaerated water.
    (d) Procedure. Using the equipment and reagents listed in paragraphs 
(a) and (b) of this section, start pumping the mobile solvent at the 
initial conditions. Inject 10 microliters of the sample solution into 
the chromatograph and begin the linear gradient pumping program. After 
the final mobile phase conditions are reached (1 hour) continue to pump 
the solvent mixture for an additional 20 minutes or until the 
demethylbleomycin A2 is eluted. The elution order is void 
volume, bleomycinic acid, bleomycin A2, bleomycin 
A5, bleomycin B2, bleomycin B4, and 
demethylbleomycin A2.
    (e) Calculations. Calculate the percentage of each bleomycin by 
comparing its peak area contribution to that of the total response of 
all the bleomycins.
[48 FR 51912, Nov. 15, 1983]



Sec. 436.340  High-pressure liquid chromatographic assay for tetracycline hydrochloride content and 4-epitetracycline hydrochloride content.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (4) A suitable recorder of at least 25.4-centimeter deflection;
    (5) A suitable integrator; and
    (6) A 30-centimeter column having an inside diameter of 4.0 
millimeters and packed with octadecyl silane chemically bonded to porous 
silica or ceramic microparticles.
    (b) Mobile phase. Dissolve 0.55 gram of monobasic ammonium phosphate 
in 900 milliliters of water. Adjust the pH to 1.8 with concentrated 
phosphoric acid and dilute to 1 liter with water. Mix 800 milliliters of 
this solution with 200 milliliters of methanol. Filter the mobile

[[Page 430]]

phase through a suitable glass fiber filter that is capable of removing 
particulate contamination to 1 micron in diameter. Degas the mobile 
phase just prior to its introduction into the chromatography pumping 
system.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 1.0 milliliter per minute. Use a detector 
sensitivity setting that gives a peak height for the 4-epitetracycline 
peak that is at least 50 percent of scale.
    (d) Preparation of working standard and sample solutions--(1) 
Working standard solution. Accurately weigh approximately 18 milligrams 
of the tetracycline hydrochloride working standard into a 50-milliliter 
volumetric flask. Into the same flask, accurately weigh approximately 38 
milligrams of the 4-epitetracycline working standard. Dissolve and 
dilute to volume with a methanol:water mixture (7:18).
    (2) Sample solution. Reconstitute the sample as directed in the 
labeling. Transfer 10.0 milliliters of the reconstituted sample into a 
50-milliliter volumetric flask and dilute to volume with a 
methanol:water mixture (7:18).
    (e) Procedure. Using the equipment, reagents, and operating 
conditions as listed in paragraphs (a), (b), and (c) of this section, 
inject 10 microliters of the working standard solution into the 
chromatograph. Allow an elution time sufficient to obtain separation of 
the expected components. After separation of the working standard 
solution has been completed, inject 10 microliters of the sample 
solution into the chromatograph and repeat the procedure described for 
the working standard solution. The elution order is 4-epitetracycline 
followed by tetracycline.
    (f) Calculations. Calculate the tetracycline hydrochloride and 4-
epitetracycline hydrochloride content as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.057

where:
A1=Area of the tetracycline sample peak (at a retention time 
          equal to that observed for tetracycline in the tetracycline 
          working standard);
A2=Area of the tetracycline peak in the tetracycline working 
          standard;
A3=Area of the 4-epitetracycline sample peak (at a retention 
          time equal to that observed for the 4-epitetracycline peak in 
          the 4-epitetracycline working standard);
A4=Area of the 4-epitetracycline peak in the 4-
          epitetracycline working standard;
Wt=Milligrams of the tetracycline working standard;
We=Milligrams of the 4-epitetracycline working standard;
B=Percent tetracycline hydrochloride in the tetracycline working 
          standard;
C=Percent tetracycline hydrochloride in the 4-epitetracycline working 
          standard;
D=Percent 4-epitetracycline hydrochloride in the 4-epitetracycline 
          working standard; and
E=Percent 4-epitetracycline hydrochloride in the tetracycline working 
          standard.
[48 FR 51290, Nov. 8, 1983]



Sec. 436.341  High-pressure liquid chromatographic assay for plicamycin.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 280 nanometers;

[[Page 431]]

    (4) A suitable recorder of at least 25.4-centimeter deflection;
    (5) A suitable integrator; and
    (6) A 25-centimeter column having an inside diameter of 4.6 
millimeters and packed with octadecyl silane chemically bonded to porous 
silica or ceramic microparticles, 5 micrometers to 10 micrometers in 
diameter, U.S.P. XX.
    (b) Reagents--(1) 0.01M phosphoric acid.
    (2) Mobile phase. Mix acetonitrile (high-pressure liquid 
chromatography grade):0.01M phosphoric acid (350:650). Filter the mobile 
phase through a suitable glass fiber filter or equivalent that is 
capable of removing particulate contamination to 1 micron in diameter. 
Degas the mobile phase just prior to its introduction into the 
chromatograph pumping system.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 1.0 milliliter per minute. Use a detector 
sensitivity setting that gives a peak height for the working standard 
that is at least 50 percent of scale.
    (d) Preparation of working standard and sample solutions--(1) 
Preparation of working standard solution. Place approximately 5 
milligrams of the plicamycin working standard, accurately weighed, into 
a 50-milliliter, amber volumetric flask and dilute to volume with mobile 
phase and mix.
    (2) Preparation of sample solution. Prepare the sample solution as 
described in the individual monograph for the drug being tested.
    (e) Procedure. Use the equipment, reagents, operating conditions, 
and working standard and sample solutions described in paragraphs (a), 
(b), (c), and (d) of this section, and proceed as directed in paragraph 
(e)(1) of this section.
    (1) System suitability test. Equilibrate and condition the column by 
passage of about 10 to 15 void volumes of mobile phase followed by two 
or more replicate injections of the working standard solution. Allow an 
elution time sufficient to obtain satisfactory separation of expected 
components after each injection. Record the peak responses and calculate 
the relative standard deviation as described for system suitability 
tests in the U.S.P. XX General Chapter 621 chromatography. Proceed as 
directed in paragraph (e)(2) of this section if the minimum performance 
requirement for the relative standard deviation is not more than 2.0 
percent. If the minimum performance requirement is not met, adjustment 
must be made to the system to obtain satisfactory operation before 
proceeding as described in paragraph (e)(2) of this section.
    (2) Determination of the chromatogram. Inject 10 microliters of the 
working standard solution into the chromatograph. Allow an elution time 
sufficient to obtain satisfactory separation of expected components. 
After separation of the working standard has been completed, inject 10 
microliters of the sample solution into the chromatograph and repeat the 
procedure described for the working standard solution.
    (f) Calculations. Calculate the plicamycin content as described in 
the individual monograph for the drug being tested.
[49 FR 24017, June 11, 1984, as amended at 50 FR 5749, Feb. 12, 1985]



Sec. 436.342  High-pressure liquid chromatographic assay for cefazolin.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (4) A suitable recorder of at least 25.4 centimeter deflection; and
    (5) A 30-centimeter column having an inside diameter of 4.0 
millimeters and packed with octadecyl silane chemically bonded to porous 
silica or ceramic microparticles, 5 to 10 micrometers in diameter, USP 
XX.
    (b) Reagents--(1) Buffer solution, pH 3.6. Transfer 0.9 gram of 
sodium phosphate, dibasic USP and 1.298 grams of citric acid USP to a 1-
liter volumetric flask. Dissolve and dilute to volume with distilled 
water and mix.
    (2) Buffer solution, pH 7.0. Transfer 5.68 grams of sodium 
phosphate, dibasic

[[Page 432]]

USP and 3.63 grams of potassium phosphate monobasic to a 1-liter 
volumetric flask. Dissolve and dilute to volume with distilled water and 
mix.
    (3) Mobile phase. Mix buffer solution, pH 3.6: acetonitrile (9:1). 
Filter through a suitable glass fiber filter or equivalent that is 
capable of removing particulate contamination to 1 micron in diameter. 
Degas the mobile phase just prior to its introduction into the 
chromatograph pumping system.
    (4) Internal standard solution. Transfer 1.2 grams of salicylic acid 
to a 200-milliliter volumetric flask. Dissolve in 10 milliliters of 
methyl alcohol, dilute to volume with buffer solution, pH 7.0, and mix.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 2 milliliters per minute. Use a detector 
sensitivity setting that gives a peak height for the working standard 
that is at least 50 percent of scale. The minimum between peaks must be 
no more than 2 millimeters above the initial baseline.
    (d) Preparation of working standard and sample solutions--(1) 
Working standard solution. Place approximately 50 milligrams of 
cefazolin working standard, accurately weighed, into a 50-milliliter 
volumetric flask. Dissolve and dilute to volume with buffer solution, pH 
7.0, and mix. Transfer 4.0 milliliters of this solution to a 200-
milliliter volumetric flask, add 5.0 milliliters of internal standard 
solution, dilute to volume with buffer solution, pH 7.0, and mix.
    (2) Sample solution. Place approximately 50 milligrams of the 
sample, accurately weighed, into a 50-milliliter volumetric flask. 
Dissolve and dilute to volume with buffer solution, pH 7.0, and mix. 
Transfer 4.0 milliliters of this solution to a 200-milliliter volumetric 
flask, add 5.0 milliliters of internal standard solution, dilute to 
volume with buffer solution, pH 7.0, and mix.
    (e) Procedure. Using the equipment, mobile phase, and operating 
conditions listed in paragraphs (a), (b), and (c) of this section, 
inject 10 microliters of the working standard solution prepared as 
directed in paragraph (d)(1) of this section into the chromatograph. 
After separation of the working standard solution has been completed, 
inject 10 microliters of the sample solution prepared as described in 
paragraph (d)(2) of this section into the chromatograph and repeat the 
procedure described for the working standard solution. Allow an elution 
time sufficient to obtain satisfactory separation of the expected 
components. The elution order is void volume, salicylic acid and 
cefazolin.
    (f) Calculation. Calculate the micrograms of cefazolin per milligram 
of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.058

where:
Ru = Area of the cefazolin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard) /Area of internal standard peak;
Rs = Area of the cefazolin peak in the chromatogram of the 
          cefazolin working standard/Area of internal standard peak;
Ps = Cefazolin activity in the cefazolin working standard 
          solution in micrograms per milliliter;
Cu = Milligrams of sample per milliliter of sample solution; 
          and
m = Percent moisture content of the sample.
[48 FR 33478, July 22, 1983; 48 FR 34947, Aug. 2, 1983]



Sec. 436.343  High-pressure liquid chromatographic assay for cefuroxime.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (4) A suitable recorder of at least 25.4 centimeter deflection;
    (5) A suitable integrator; and
    (6) A 15-centimeter column having an inside diameter of 4.6 
millimeters and packed with hexyl silane chemically bonded to porous 
silica or ceramic microparticles, 5 micrometers in diameter.
    (b) Reagents--(1) Acetate buffer, pH 3.4. Place 50 milliliters of 
0.1M sodium acetate into a 1,000-milliliter volumetric flask and dilute 
to volume with 0.1M acetic acid. Mix.

[[Page 433]]

    (2) Mobile phase. Mix 0.1M acetate buffer, pH 3.4:acetonitrile 
(10:1). Filter the mobile phase through a suitable glass fiber filter or 
equivalent that is capable of removing particulate contamination to 1 
micron in diameter. Degas the mobile phase just prior to its 
introduction into the chromatograph pumping system.
    (3) Internal standard solution. Prepare a 1.5 milligram per 
milliliter solution of orcinol monohydrate in water.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 2.0 milliliters per minute. Use a detector 
sensitivity setting that gives a peak height for the working standard 
that is at least 50 percent of scale.
    (d) Preparation of working standard and sample solutions--(1) 
Preparation of working standard solution. Dissolve an accurately weighed 
portion of the cefuroxime working standard with sufficient distilled 
water to obtain a stock solution containing 1.0 milligram of cefuroxime 
per milliliter. Immediately transfer 5.0 milliliters of the stock 
solution to a 100-milliliter volumetric flask, add 20.0 milliliters of 
internal standard solution and dilute to 100 milliliters with distilled 
water and mix. Store the solution in a refrigerator and use within 6 
hours.
    (2) Preparation of sample solutions--(i) Product not packaged for 
dispensing (micrograms of cefuroxime per milligram). Dissolve an 
accurately weighed portion of the sample with sufficient distilled water 
to obtain a stock solution containing 1.0 milligram of cefuroxime per 
milliliter. Immediately transfer 5.0 milliliters of the stock solution 
to a 100-milliliter volumetric flask, add 20.0 milliliters of internal 
standard solution and dilute to 100 milliliters with distilled water and 
mix. Store the solution in a refrigerator and use within 6 hours. Using 
this sample solution, proceed as directed in paragraph (e) of this 
section.
    (ii) Product packaged for dispensing. Determine both micrograms of 
cefuroxime per milligram of the sample and milligrams of cefuroxime per 
container. Use separate containers for preparation of each sample 
solution as described in paragraphs (d)(2)(ii) (a) and (b) of this 
section.
    (a) Micrograms of cefuroxime per milligram. Dissolve an accurately 
weighed portion of the sample with sufficient distilled water to obtain 
a stock solution containing 1.0 milligram of cefuroxime per milliliter. 
Immediately transfer 5.0 milliliters of the stock solution to a 100-
milliliter volumetric flask, add 20.0 milliliters of internal standard 
solution and dilute to 100 milliliters with distilled water and mix. 
Store the solution in a refrigerator and use within 6 hours. Using this 
sample solution, proceed as directed in paragraph (e) of this section.
    (b) Milligrams of cefuroxime per container. Reconstitute the sample 
as directed in the labeling. Then using a suitable hypodermic needle and 
syringe, remove all of the withdrawable contents if it is represented as 
a single-dose container; or, if the labeling specifies the amount of 
potency in a given volume of the resultant preparation, remove an 
accurately measured representative portion from each container. Dilute 
the solution thus obtained with distilled water to obtain a stock 
solution of 1.0 milligram per milliliter. Immediately transfer 5.0 
milliliters of the stock solution to a 100-milliliter volumetric flask, 
add 20.0 milliliters of internal standard solution and dilute to 100 
milliliters with distilled water and mix. Store the solution in a 
refrigerator and use within 6 hours. Using this sample solution, proceed 
as directed in paragraph (e) of this section.
    (e) Procedure. Using the equipment, reagents, and operating 
conditions as listed in paragraphs (a), (b), and (c) of this section, 
inject 10 microliters of the working standard solution into the 
chromatograph. Allow an elution time sufficient to obtain satisfactory 
separation of the expected components. After separation of the working 
standard solution has been completed, inject 10 microliters of the 
sample solution prepared as described in paragraph (d)(2)(i) of this 
section into the chromatograph and repeat the procedure described for 
the working standard solution. If the sample is packaged for dispensing, 
repeat the procedure for each sample solution prepared as described in 
paragraphs (d)(2)(ii)(a) and (d)(2)(ii)(b) of this section.

[[Page 434]]

    (f) Calculations. (1) Calculate the micrograms of cefuroxime per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.059

where:
Ru=Area of the cefuroxime peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard)/Area of internal standard peak;
Rs=Area of the cefuroxime peak in the chromatogram of the 
          cefuroxime working standard/Area of internal standard peak;
Ps=Cefuroxime activity in the cefuroxime working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of sample solution; 
          and
m=Percent moisture content of the sample.

    (2) Calculate the cefuroxime content of the vial as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.060
    
where:
Ru=Area of the cefuroxime peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard)/Area of internal standard peak;
Rs=Area of the cefuroxime peak in the chromatogram of the 
          cefuroxime working standard/Area of internal standard peak;
Ps=Cefuroxime activity in the cefuroxime working standard 
          solution in micrograms per milliliter; and
d= Dilution factor of the sample.
[48 FR 38460, Aug. 24, 1983; 48 FR 40704, Sept. 9, 1983]



Sec. 436.344  Thin layer chromatographic identity test for cefuroxime.

    (a) Equipment--(1) Chromatography tank. Use a rectangular tank 
approximately 23 x 23 x 9 centimeters, with a glass solvent trough on 
the bottom and a tight-fitting cover. Line the inside walls of the tank 
with Whatman 3MM chromatographic paper or equivalent.
    (2) Plates. Use 20  x  20 centimeter thin layer chromatography 
plates coated with Silica Gel F or equivalent to a thickness of 250 
microns.
    (b) Developing solvent. Mix chloroform, methanol, and formic acid in 
volumetric proportions of 90:16:4, respectively.
    (c) Preparation of the spotting solutions. Dissolve approximately 
200 milligrams each of the working standard and sample in 5 milliliters 
of a 50 percent aqueous acetone solution.
    (d) Procedure. Pour the developing solvent into the glass trough at 
the bottom of the chromatography tank. Cover and seal the tank. Allow it 
to equilibrate for 1 hour. Prepare a plate as follows: On a line 2 
centimeters from the base of the plate, and at intervals of 2 
centimeters, spot 5 microliters each of the sample and working standard 
solutions. After all spots are thoroughly dry, place the plate directly 
into the glass trough of the chromatography tank. Cover and seal the 
tank tightly. Allow the solvent front to travel a minimum of 15 
centimeters from the starting line. Remove the plate from the tank and 
allow it to air dry. Observe under ultraviolet light (254 nanometers).
    (e) Evaluation. Measure the distance the solvent front traveled from 
the starting line and the distance the spots are from the starting line. 
Calculate the Rf value by dividing the latter by the former. 
The sample and standard should have spots of corresponding Rf 
values.
[48 FR 38461, Aug. 24, 1983]



Sec. 436.345  High-pressure liquid chromatographic assay for ceftizoxime.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (4) A suitable recorder of at least 25.4 centimeter deflection;
    (5) A suitable integrator; and
    (6) A 30-centimeter column having an inside diameter of 4.0 
millimeters and packed with octadecyl silane chemically bonded to porous 
silica or ceramic microparticles, 5 to 10 micrometers in diameter, USP 
XX.
    (b) Reagents--(1) pH 3.6 buffer solution. Transfer 2.31 grams of 
sodium phosphate diabasic dodecahydrate and 1.42 grams of citric acid 
monohydrate to a

[[Page 435]]

1-liter volumetric flask. Dissolve and dilute to volume with distilled 
water.
    (2) pH 7.0 buffer solution. Transfer 14.33 grams of sodium phosphate 
dibasic dodecahydrate and 3.63 grams of potassium phosphate monobasic to 
a 1-liter volumetric flask. Dissolve and dilute to volume with distilled 
water.
    (3) Mobile phase. Mix pH 3.6 buffer solution:acetonitrile (9:1). 
Filter the mobile phase through a suitable glass fiber filter or 
equivalent that is capable of removing particulate contamination to 1 
micron in diameter. Degas the mobile phase just prior to its 
introduction into the chromatograph pumping system.
    (4) Internal standard solution. Place 1.2 grams of salicyclic acid 
in a 200-milliliter volumetric flask. Dissolve in 10 milliliters of 
methyl alcohol, dilute to volume with pH 7.0 buffer solution and mix.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 2.0 milliliters per minute. Use a detector 
sensitivity setting that gives a peak height for the working standard 
that is at least 50 percent of scale.
    (d) Preparation of working standard solution. Dissolve an accurately 
weighed portion of the ceftizoxime working standard with sufficient pH 
7.0 buffer solution to obtain a solution containing 1,000 micrograms of 
ceftizoxime activity per milliliter. Transfer 2.0 milliliters of this 
solution to a 100-milliliter volumetric flask, add 5.0 milliliters of 
internal standard solution, dilute to volume with pH 7.0 buffer solution 
and mix.
    (e) Preparation of sample solutions--(1) Product not packaged for 
dispensing (micrograms of ceftizoxime per milligram). Dissolve an 
accurately weighed portion of the sample with sufficient pH 7.0 buffer 
solution to obtain a concentration of 1.0 milligram per milliliter. 
Transfer 2.0 milliliters of this solution to a 100-milliliter volumetric 
flask, add 5.0 milliliters of internal standard solution, dilute to 
volume with pH 7.0 buffer solution and mix. Using this sample solution, 
proceed as directed in paragraph (f) of this section.
    (2) Product packaged for dispensing. Determine both micrograms of 
ceftizoxime per milligram of the sample and milligrams of ceftizoxime 
per container. Use separate containers for preparation of each sample 
solution as described in paragraphs (e)(2) (i) and (ii) of this section.
    (i) Micrograms of ceftizoxime per milligram. Dissolve an accurately 
weighed portion of the sample with sufficient pH 7.0 buffer solution to 
obtain a concentration of 1.0 milligram of ceftizoxime per milliliter. 
Transfer 2.0 milliliters of this solution to a 100-milliliter volumetric 
flask, add 5.0 milliliters of internal standard solution, dilute to 
volume with pH 7.0 buffer solution and mix. Using this sample solution, 
proceed as directed in paragraph (f) of this section.
    (ii) Milligrams of ceftizoxime per container. Reconstitute the 
sample as directed in the labeling. Then using a suitable hypodermic 
needle and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency is a given volume of the resultant preparation, 
remove an accurately measured representative portion from each 
container. Further dilute an aliquot of the solution thus obtained with 
sufficient pH 7.0 buffer solution to obtain a concentration of 1.0 
milligram per milliliter. Transfer 2.0 milliliters of this solution to a 
100-milliliter volumetric flask, add 5.0 milliliters of internal 
standard solution, dilute to volume with pH 7.0 buffer solution and mix. 
Using this sample solution, proceed as directed in paragraph (f) of this 
section.
    (f) Procedure. Using the equipment, reagents, and operating 
conditions as listed in paragraphs (a), (b), and (c) of this section, 
inject 10 microliters of the working standard solution into the 
chromatograph. Allow an elution time sufficient to obtain satisfactory 
separation of the expected components. The elution order is void volume, 
ceftizoxime, and internal standard. After separation of the working 
standard solution has been completed, inject 10 microliters of the 
sample solution prepared as described in paragraph (e)(1) of this 
section into the chromatograph and repeat the procedure described for 
the working standard solution. If the sample is packaged for

[[Page 436]]

dispensing, repeat the procedure for each sample solution prepared as 
described in paragraphs (e)(2) (i) and (ii) of this section.
    (g) Calculations--(1) Calculate the micrograms of ceftizoxime per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.061

where:

Ru=Area of the ceftizoxime peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard)/Area of internal standard peak;
Rs=Area of the ceftizoxime peak in the chromatogram of the 
          ceftizoxime working standard/Area of internal standard peak;
Ps=Ceftizoxime activity in the ceftizoxime working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of sample solution; 
          and
m=Percent moisture content of the sample.
[GRAPHIC] [TIFF OMITTED] TR01JA93.062

where:
Ru=Area of the ceftizoxime peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard)/Area of internal standard peak;
Rs=Area of the ceftizoxime peak in the chromatogram of the 
          ceftizoxime working standard/Area of internal standard peak;
Ps=Ceftizoxime activity in the ceftizoxime working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.
[48 FR 46270, Oct. 12, 1983; 48 FR 49656, Oct. 27, 1983]



Sec. 436.346  High-pressure liquid chromatographic assay for cyclosporine.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A suitable pump capable of reproducibly delivering a liquid to a 
pressure of 4,500 pounds per square inch and a flow rate of at least 5 
milliliters per minute;
    (2) A suitable ultraviolet detection system operating at a 
wavelength of 210 nanometers;
    (3) A suitable recorder;
    (4) A suitable integrator;
    (5) An oven or water bath capable of maintaining the column at an 
operating temperature of 70 deg. C;
    (6) A steel capillary tube, 1 meter in length, having an inside 
diameter of 0.25 millimeter. This tube is inserted between the injection 
system and the chromatographic column and is equilibrated to 70 deg. C; 
and
    (7) A sample injection valve on which the loop determines the sample 
size.
    (b) Columns. The chromatographic column is packed with 
microparticulate (3 to 10 micrometers in diameter) reversed phase 
packing materials that exhibit some degree of polarity such as the 
hydrocarbon bonded silicas with dimethyl, trimethyl, or octyl groups. 
Connect a saturation column gravity packed with similarly bonded silica 
particles 40 to 60 microns in diameter to the inlet of the analytical 
column.
    (c) Mobile phase. Mix acetonitrile, water, methanol, and o-
phosphoric acid (900:525:75:0.075 by volume). Degas by passing through a 
0.5-micrometer filter with vacuum and ultrasonicate for no less than 2 
minutes before use. The mobile phase may be sparged perceptibly with 
helium through a 2-micrometer metal filter for the duration of the 
analysis. Adjust the ratio of acetonitrile to aqueous buffer as 
necessary to obtain satisfactory retention of the peaks.
    (d) Operating conditions. Perform the assay at a constant operating 
temperature of 70 deg. C with a typical flow rate of 2.0 milliliters per 
minute. Use a detector sensitivity setting that gives a peak height for 
the working standard that is at least 50 percent of scale with a typical 
chart speed of 2.5 millimeters per minute. Obtain chromatograms for 
performance parameters at a chart speed of not less than 25 millimeters 
per minute to allow a more accurate measurement of peak geometry.
    (e) Preparation of working standard and sample solutions. Prepare 
the working standard and sample solutions as directed in the individual 
monographs for cyclosporine.
    (f) Systems suitability. Equilibrate and condition the column by 
passage of about 10 to 15 void volumes of mobile phase followed by about 
5 injections of not less than 10 microliters each of working standard 
solution. Proceed

[[Page 437]]

with the analysis when the following minimum performance requirements 
have been met or exceeded.
    (1) Capacity ratio factor. Calculate the capacity ratio (k) of the 
cyclosporine peak as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.063

where:
t=Retention time of solute; and
tm=Retention time of solvent or unretained substance.


The capacity ratio is satisfactory if it is not less than 3 or not more 
than 10.
    (2) Coefficient of variation. The coefficient of variation of at 
least five replicate injections is less than 1 percent.
    (3) Efficiency. Calculate the efficiency (n) as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.013
    
where:
t=Retention time of solute; and
W0.5=Peak width at half height. Both t and W0.5 
          must be measured in the same units.


The efficiency is satisfactory if it is greater than 1,500 theoretical 
plates when assaying cyclosporine and greater than 700 theoretical 
plates when assaying finished dosage forms.
    (4) Asymmetry factor. Calculate the asymmetry factor (As) 
as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.064

where:
W0.1=Horizontal distance measured from a point on the 
          cyclosporine peak ascent 10 percent above the baseline to an 
          intercept with the cyclosporine peak descent; and
f=Horizontal distance from point of 10 percent ascent above the baseline 
          of the cyclosporine peak to point of maximum peak height.

The asymmetry factor is satisfactory if it is not more than 1.5.
    (5) Resolution. Calculate the resolution (Rs) as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.065
    
where:
t=Retention time of solute; and the subscripts i and j designate two 
          different peaks and where tj is larger than 
          ti; and
W=Width of peak at baseline as determined by extrapolating the relative 
          straight sides to the baseline. Both t and W must be measured 
          in the same units.


Resolution between the cyclosporine peak and any other peak must be at 
least 1.1.
    (g) Procedure. Using the equipment, columns, mobile phase, operating 
conditions and the working standard and sample solutions listed in 
paragraphs (a), (b), (c), (d), and (e) of this section, inject 20 
microliters of the working standard solution into the chromatograph. 
Allow an elution time sufficient to obtain satisfactory separation of 
expected components. After separation of the working standard solution 
has been completed, inject 20 microliters of the sample solution into 
the chromatograph and repeat the procedure described for the working 
standard solution.
    (h) Calculations. Calculate the cyclosporine content of cyclosporine 
and its dosage forms as directed in the individual monographs.
[49 FR 22631, May 31, 1984; 49 FR 27489, July 5, 1984]



Sec. 436.347  High-pressure liquid chromatographic assay for cefoxitin.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (4) A suitable recorder of at least 25.4 centimeter deflection;
    (5) A suitable integrator; and
    (6) A 30-centimeter column having an inside diameter of 4.0 
millimeters and packed with octadecyl silane chemically bonded to porous 
silica or ceramic microparticles, 5 micrometers to 10 micrometers in 
diameter, U.S.P. XX.
    (b) Reagents--(1) One percent potassium phosphate buffer, pH 6.0. 
Prepare as described in Sec. 436.101(a)(1).

[[Page 438]]

    (2) Mobile phase. Mix distilled water:glacial acetic 
acid:acetonitrile (800:10:190). Filter the mobile phase through a 
suitable glass fiber filter or equivalent that is capable of removing 
particulate contamination to 1 micron in diameter. Degas the mobile 
phase just prior to its introduction into the chromatograph pumping 
system.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 1.0 milliliter per minute. Use a detector 
sensitivity setting that gives a peak height for the working standard 
that is at least 50 percent of scale. The minimum between peaks must be 
no more than 2 millimeters above the baseline.
    (d) Preparation of working standard and sample solutions. Use the 
working standard and sample solutions prepared as described in the 
individual monographs for the drug being tested.
    (e) Procedure. Using the equipment, reagents, and operating 
conditions as described in paragraphs (a), (b), and (c) of this section, 
inject 10 microliters of the working standard solution into the 
chromatograph. Allow an elution time sufficient to obtain separation of 
the expected components. After separation of the working standard 
solution has been completed, inject 10 microliters of the sample 
solution into the chromatograph and repeat the procedure described for 
the working standard solution.
    (f) Calculations. Calculate the cefoxitin content as described in 
the individual monographs for the drug being tested.
[49 FR 47827, Dec. 7, 1984]



Sec. 436.348  High-pressure liquid chromatographic assay for ceforanide.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A low dead volume cell 8 to 20 microliters;
    (2) A light path length of 1 centimeter;
    (3) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (4) A suitable recorder of at least 25.4-centimeter deflection;
    (5) A suitable integrator; and
    (6) A 30-centimeter column having an inside diameter of 4.0 
millimeters and packed wth octadecyl silane chemically bonded to porous 
silica or ceramic microparticles, 5 micrometers to 10 micrometers in 
diameter, U.S.P. XX. A particular column used for analysis of ceforanide 
should not be used for the analysis of other drugs.--
    (b) Mobile phase. Mix 18.0 milliliters of 10 percent aqueous 
tetrabutylammonium hydroxide and 8.56 milliliters of 11N potassium 
hydroxide. Add the mixture to approximately 700 milliliters of distilled 
water. Add 200 milliliters of reagent grade methanol. Adjust the pH of 
the mixture to pH 7.0 with concentrated phosphoric acid and dilute to 
1,000 milliliters with distilled water. Prepare fresh daily. Filter the 
mobile phase through a suitable glass fiber filter or equivalent which 
is capable of removing particulate contamination to 1 micron in 
diameter. Degas the mobile phase just prior to its introduction into the 
chromatograph pumping system.
    (c) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 1 milliliter per minute. Use a detector 
sensitivity setting that gives a peak height for the working standard 
that is at least 50 percent of scale.
    (d) Preparation of working standard and sample solutions--(1) 
Preparation of working standard solution. Prepare a solution containing 
1,000 micrograms of ceforanide activity per milliliter in mobile phase. 
Inject working standard solution within 5 minutes after dissolution.
    (2) Preparation of sample solution. Prepare the sample solution as 
directed in the individual monograph for the drug being tested. Inject 
sample solution within 5 minutes after dissolution.
    (e) Procedure. Use the equipment, mobile phase, operating 
conditions, and working standard and sample solutions described in 
paragraphs (a), (b), (c), and (d) of this section, and proceed as 
directed in paragraph (e)(1) of this section.
    (1) System suitability test. Equilibrate and condition the column by 
passage of about 10 to 15 void volumes of mobile

[[Page 439]]

phase followed by three replicate injections of 10 microliters each of 
the working standard solution. Allow an elution time sufficient to 
obtain satisfactory separation of expected components after each 
injection. Record the peak responses and calculate the tailing factor, 
efficiency of the column, coefficient of variation, and capacity factor 
as described for system suitability tests in the U.S.P. XX General 
Chapter 621 chromatography. Proceed as directed in paragraph (e)(2) of 
this section if the following minimum performance requirements have been 
met:
    (i) Tailing factor. The tailing factor is satisfactory if it is not 
more than 1.2;
    (ii) Efficiency of the column. The efficiency of the column is 
satisfactory if it is greater than 1,900 theoretical plates;
    (iii) Coefficient of variation. The coefficient of variation of at 
least three replicate injections is satisfactory if it is not more than 
1.5 percent; and
    (iv) Capacity factor. The capacity factor is satisfactory if it is 
not less than 1.8 and not more than 5.

If the minimum performance requirements are not met, adjustments must be 
made to the system to obtain satisfactory operation before proceeding as 
described in paragraph (e)(2) of this section.
    (2) Determination of the chromatogram. Inject 10 microliters of the 
working standard solution into the chromatograph. Allow an elution time 
sufficient to obtain satisfactory separation of the expected components. 
After separation of the working standard solution has been completed, 
inject 10 microliters of the sample solution into the chromatograph and 
repeat the procedure described for the working standard solution.
    (f) Calculations. Calculate the ceforanide content as directed in 
the individual monograph for the drug being tested.
[49 FR 25846, June 25, 1984; 49 FR 34347, Aug. 30, 1984]



Sec. 436.349  High-pressure liquid chromatographic assay for L-lysine in ceforanide for injection.

    (a) Equipment. A suitable high-pressure liquid chromatograph 
equipped with:
    (1) A suitable pump capable of reproducibly delivering a liquid to a 
pressure of 5,000 pounds per square inch;
    (2) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers;
    (3) A suitable recorder;
    (4) A suitable integrator; and
    (5) A 25-centimeter column having an inside diameter of 4.6 
millimeters and packed with octadecyl silane chemically bonded to porous 
silica or ceramic microparticles, 5 micrometers to 10 micrometers in 
diameter, U.S.P. XX.
    (b) Reagents--(1) 2,4-Dinitrofluorobenzene solution. Weigh 
accurately approximately 760 milligrams of 2,4-dinitrofluorobenzene into 
a 50-milliliter volumetric flask. Dissolve and dilute to volume with 
absolute ethyl alcohol.
    (2) Tris (hydroxymethyl) aminomethane (THAM) solution. Weigh 
accurately approximately 1.44 grams of THAM into a 100-milliliter 
volumetric flask. Dissolve and dilute to volume with distilled water.
    (c) Mobile phase. Mix methanol and water (62:38), and adjust to pH 
3.0 with glacial acetic acid.
    (d) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of 1.5 milliliters per minute. Use a detector 
sensitivity setting that gives a peak height for the standard that is at 
least 50 percent of scale with a typical chart speed of 0.2 inch per 
minute.
    (e) Preparation of standard and sample solutions--(1) Preparation of 
standard solution. Weigh accurately approximately 36 milligrams of L-
lysine used as the standard into a 100-milliliter volumetric flask. 
Dissolve and dilute to volume with distilled water. Transfer 2.0 
milliliters of the L-lysine solution into a 10-milliliter volumetric 
flask, add 2.0 milliliters of THAM solution and 3.0 milliliters of 2,4-
dinitrofluorobenzene solution. Cap tightly and mix well. Place the flask 
in a 50 deg. C water bath for 30 minutes. Remove from water bath, allow 
the flask to cool to room temperature, and dilute to volume with 
methanol. Mix well.
    (2) Preparation of sample solution. Weigh accurately approximately 
150

[[Page 440]]

milligrams of the sample, ceforanide for injection, into a 100-
milliliter volumetric flask. Dissolve and dilute to volume with 
distilled water. Transfer 2.0 milliliters of the sample solution into a 
10-milliliter volumetric flask, add 2.0 milliliters of THAM solution and 
3.0 milliliters of 2,4-dinitrofluorobenzene solution. Cap tightly and 
mix well. Place the flask in a 50 deg. C water bath for 30 minutes. 
Remove from water bath, allow the flask to cool to room temperature, and 
dilute to volume with methanol. Mix well.
    (f) Procedure. Use the equipment, reagents, mobile phase, operating 
conditions, and standard and sample solutions described in paragraphs 
(a), (b), (c), (d), and (e) of this section, and proceed as directed in 
paragraph (f)(1) of this section.
    (1) System suitability test. Equilibrate and condition the column by 
passage of about 10 to 15 void volumes of mobile phase followed by three 
replicate injections of 20 microliters each of the standard solution. 
Allow an elution time sufficient to obtain satisfactory separation of 
the expected components after each injection. Record the peak responses 
and calculate the resolution factor, tailing factor, efficiency of the 
column, coefficient of variation, and capacity factor as described for 
system suitability tests in the U.S.P. XX General Chapter 621 
chromatography. Proceed as directed in paragraph (f)(2) of this section 
if the following minimum performance requirements have been met:
    (i) Resolution factor. The resolution factor between the peak for 
derivatized L-lysine and from the peak for the dinitrofluorobenzene 
derivatizing reagent is satisfactory if it is not less than 4.5;
    (ii) Tailing factor. The tailing factor is satisfactory if it is not 
more than 1.3;
    (iii) Efficiency of the column. The efficiency of the column is 
satisfactory if it is greater than 1,500 theoretical plates;
    (iv) Coefficient of variation. The coefficient of variation of at 
least three replicate injections is satisfactory if it is not more than 
1.5 percent; and
    (v) Capacity factor. The capacity factor is satisfactory if it is 
not less than 4 and not more than 6.

If the minimum performance requirements are not met, adjustments must be 
made to the system to obtain satisfactory operation before proceeding as 
described in paragraph (f)(2) of this section.
    (2) Determination of the chromatogram. Inject 20 microliters of the 
standard solution into the chromatograph. Allow an elution time 
sufficient to obtain satisfactory separation of the expected components. 
After separation of the standard solution is completed, inject 20 
microliters of the sample solution into the chromatograph and repeat the 
procedure described for the standard solution.
    (g) Calculations. Calculate the percent of L-lysine per milligram of 
ceforanide for injection as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.066

where:
Au = Area of the L-lysine peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As = Area of the L-lysine peak in the chromatogram of the L-
          lysine standard:
Ps = L-lysine content in the L-lysine standard solution in 
          micrograms per milliliter; and
Cu = Milligrams of sample per milliliter of sample solution.
[49 FR 25846, June 25, 1984; 49 FR 34347, Aug. 30, 1984; 49 FR 40006, 
Oct. 12, 1984]



Sec. 436.350  High-performance liquid chromatographic assay for cefonicid.

    (a) Apparatus. A suitable high-performance liquid chromatograph 
equipped with:
    (1) A suitable detection system specified in the monograph for the 
drug being tested;
    (2) A suitable recording device of at least 25-centimeter 
deflection;
    (3) A suitable chromatographic data managing system; and
    (4) An analytical column, 3 to 30 centimeters long, packed with a 
material as defined in the monograph for the drug being tested; and if 
specified in

[[Page 441]]

that monograph, the inlet of this column may be connected to a guard 
column 3 to 5 centimeters in length, packed with the same material of 40 
to 60 micrometers particle size.
    (b) Procedure. Perform the assay and calculate the drug content 
using the temperature, instrumental conditions, and calculations 
specified in the monograph for the drug being tested with a flow rate 
not to exceed 2.0 milliliters per minute. Use a detector sensitivity 
setting that gives a peak height for the working standard that is at 
least 50 percent of scale with typical chart speed of not less than 2.5 
millimeters per minute. Use the apparatus described in paragraph (a) of 
this section; and the reagents and working standard and sample solutions 
described in the monograph for the drug being tested. Equilibrate and 
condition the column by passage of 10 to 15 void volumes of mobile phase 
followed by five replicate injections of the same volume (between 10 and 
20 microliters) of the working standard solution. Allow an operating 
time sufficiently long to obtain satisfactory separation and elution of 
the expected components after each injection. Record the peak responses 
and calculate the prescribed system suitability requirements as follows:
    (c) System suitability test. Using the apparatus and procedure 
described in this section, test the chromatographic system for assay as 
follows:
    (1) Tailing factor. Calculate the tailing factor (T), from distances 
measured along the horizontal line at 5 percent of the peak height above 
the baseline, as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.067

where:
W0.05 = Width of peak at 5 percent height; and
f = Horizontal distance from point of ascent to a point coincident with 
          maximum peak height.

    (2) Efficiency of the column. Calculate the number of theoretical 
plates (n) of the column as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.014

where:
n=Efficiency, as number of theoretical plates for column;
tR=Retention time of solute; and
wh=Peak width at half-height.

    (3) Resolution factor. Calculate the resolution factor (R), between 
desacetyl cefonicid and cefonicid, as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.068

where:
t1=Retention time of desacetyl cefonicid;
t2=Retention time of cefonicid; and
w1 and w2=Widths of the bases of the corresponding 
          peaks obtained by extrapolating the relatively straight sides 
          of the peaks to the baseline.

    (4) Coefficient of variation (relative standard deviation). 
Calculate the coefficient of variation (SR in percent) as 
follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.015

where:
X is the mean of N individual measurements of Xi.


If the complete operating system meets the system suitability 
requirements of the monograph for the drug being tested, proceed as 
described in paragraph (b) of this section, using the sample solution in 
lieu of the working standard solution.
[49 FR 34347, Aug. 30, 1984, as amended at 49 FR 44460, Nov. 7, 1984; 50 
FR 29209, July 18, 1985]



Sec. 436.351  High-performance liquid chromatographic assay for amoxicillin and clavulanic acid.

    (a) Apparatus. A suitable high-performance liquid chromatograph 
equipped with:
    (1) A suitable detection system specified in the monograph for the 
drug being tested;
    (2) A suitable recording device of at least 25-centimeter 
deflection;
    (3) A suitable chromatographic data managing system; and

[[Page 442]]

    (4) An analytical column, 10 to 30 centimeters long, packed with a 
material as defined in the monograph for the drug being tested; and if 
specified in that monograph, the inlet of this column may be connected 
to a guard column, 3 to 5 centimeters in length, packed with the same 
material of 40 to 60 micrometers particle size.
    (b) Procedure. Perform the assay and calculate the drug content 
using the temperature, instrumental conditions, and calculations 
specified in the monograph for the drug being tested with a flow rate 
not to exceed 2.0 milliliters per minute. Use a detector sensitivity 
setting that gives a peak height for the working standard that is at 
least 50 percent of scale with typical chart speed of not less than 2.5 
millimeters per minute. Use the apparatus described in paragraph (a) of 
this section; and the reagents and working standard and sample solutions 
described in the monograph for the drug being tested. Equilibrate and 
condition the column by passage of 10 to 15 void volumes of mobile phase 
followed by five replicate injections of the same volume (between 10 and 
20 microliters) of the working standard solution. Allow an operating 
time sufficiently long to obtain satisfactory separation and elution of 
the expected components after each injection. The retention times for 
amoxicillin and clavulanic acid are about 2.1 and 1.0 minutes, 
respectively, under these prescribed conditions. Record the peak 
responses and calculate the prescribed system suitability requirements 
as follows:
    (c) System suitability test. Using the apparatus and procedure 
described in this section, test the chromatographic system for assay as 
follows:
    (1) Tailing factors for the amoxicillin and clavulanic acid peaks. 
Calculate the tailing factors (T), from distances measured along the 
horizontal line at 5 percent of the peak height above the baseline, as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.069

where:
W0.05=Width of peak at 5 percent height; and
f=Horizontal distance from point of ascent to a point coincident with 
          maximum peak height.

    (2) Efficiency of the column. Calculate the number of theoretical 
plates (n) of the column as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.016

where:
n=Efficiency, as number of theoretical plates for column;
tR=Retention time of amoxicillin or clavulanic acid peaks; 
          and
wh=Corresponding peak width at half-height.

    (3) Resolution factor. Calculate the resolution factor (R) as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.070

where:
t1=Retention time of amoxicillin peak;
t2=Retention time of clavulanic acid peak; and
w1 and w2=Widths of the bases of the corresponding 
          peaks obtained by extrapolating the relatively straight sides 
          of the peaks to the baseline.

    (4) Coefficient of variation (Relative standard deviation). 
Calculate the coefficient of variation (SR in percent) as 
follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.017

where:
X is the mean of N individual measurements of Xi.


If the complete operating system meets the system suitability 
requirements of the monograph for the drug being tested, proceed as 
described in paragraph (b) of this section, using the sample solution in 
lieu of the working standard solution.
[49 FR 39671, Oct. 10, 1984]



Sec. 436.352  High-performance liquid chromatographic assay for determining clavam-2-carboxylate content in clavulanate potassium.

    (a) Apparatus. A suitable high-performance liquid chromatograph 
equipped with:

[[Page 443]]

    (1) A suitable detection system specified in the monograph for the 
drug being tested;
    (2) A suitable recording device of at least 25-centimeter 
deflection;
    (3) A suitable chromatographic data managing system; and
    (4) An analytical column, approximately 30 centimeters in length, 
packed with a material as defined in the monograph for the drug being 
tested.
    (b) Procedure. Perform the assay and calculate the drug content 
using the temperature, instrumental conditions, and calculations 
specified in the monograph for the drug being tested with a flow rate 
not to exceed 0.5 milliliter per minute. Use a detector sensitivity 
setting that gives a peak height for the working standard that is at 
least 50 percent of scale with typical chart speed of not less than 2.5 
millimeters per minute. Use the apparatus described in paragraph (a) of 
this section; and the mobile phase and working standard and sample 
solutions described in the monograph for the drug being tested. 
Equilibrate and condition the column by passage of 10 to 15 void volumes 
of mobile phase followed by five replicate injections of the same volume 
(between 10 and 20 microliters) of the working standard solution. Allow 
an operating time sufficiently long to obtain satisfactory separation 
and elution of the expected components after each injection. The 
retention times for clavam-2-carboxylic acid and clavulanic acid are 
about 10 and 14 minutes, respectively, under these prescribed 
conditions. The sample solution should be injected at least in duplicate 
and an average should be taken. For each such series of samples 
injected, two injections of standard should be made, one before and one 
after the sample series, and an average should be taken. Record the peak 
responses and calculate the prescribed system suitability requirements 
as follows:
    (c) System suitability test. Using the apparatus and procedure 
described in this section, test the chromatographic system for assay as 
follows:
    (1) Tailing factor. Calculate the tailing factor (T), from distances 
measured along the horizontal line at 5 percent of the peak height above 
the baseline, as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.071

where:
W0.05=Width of peak at 5 percent height; and
f=Horizontal distance from point of ascent to a point coincident with 
          maximum peak height.

    (2) Efficiency of the column. Calculate the number of theoretical 
plates (n) of the column as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.018

where:
n=Efficiency, as number of theoretical plates for column;
tR=Retention time of clavam-2-carboxylic acid peak; and
wh=Corresponding peak width at half-height.

    (3) Resolution factor. Calculate the resolution factor (R) as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.072

where:
t1=Retention time of clavam-2-carboxylic acid peak;
t2=Retention time of clavulanic acid peak; and
w1 and w2=Widths of the bases of the corresponding 
          peaks obtained by extrapolating the relatively straight sides 
          of the peaks to the baseline.

    (4) Coefficient of variation (Relative standard deviation). 
Calculate the coefficient of variation (SR

[[Page 444]]

[GRAPHIC] [TIFF OMITTED] TC01AP94.019


where:
X is the mean of N individual measurements of Xi

If the complete operating system meets the system suitability 
requirements of the monograph for the drug being tested, proceed as 
described in paragraph (b) of this section, using the sample solution in 
lieu of the working standard solution.
[49 FR 39671, Oct. 10, 1984]



Sec. 436.353  High-performance liquid chromatographic assay for amdinocillin.

    (a) Apparatus. A suitable high-performance liquid chromatograph 
equipped with:
    (1) A suitable detection system specified in the monograph for the 
drug being tested;
    (2) A suitable recording device of at least 25-centimeter 
deflection;
    (3) A suitable chromatographic data managing system; and
    (4) An analytical column, 3 to 30 centimeters long, packed with a 
material as defined in the monograph for the drug being tested; and if 
specified in that monograph, the inlet of this column may be connected 
to a guard column, 3 to 5 centimeters in length, packed with the same 
material of 40 to 60 micrometers particle size.
    (b) Procedure. Perform the assay and calculate the drug content 
using the temperature, instrumental conditions, and calculations 
specified in the monograph for the drug being tested with a flow rate 
not to exceed 2.0 milliliters per minute. Use a detector sensitivity 
setting that gives a peak height for the working standard that is at 
least 50 percent of scale with typical chart speed of not less than 2.5 
millimeters per minute. Use the apparatus described in paragraph (a) of 
this section; and the reagents and working standard and sample solutions 
described in the monograph for the drug being tested. Equilibrate and 
condition the column by passage of 10 to 15 void volumes of mobile phase 
followed by five replicate injections of the same volume (between 10 and 
20 microliters) of the working standard solution. Allow an operating 
time sufficiently long to obtain satisfactory separation and elution of 
the expected components after each injection. Record the peak responses 
and calculate the prescribed system suitability requirements as 
described for the system suitability test in paragraph (c) of this 
section.
    (c) System suitability test. Using the apparatus and procedure 
described in this section, test the chromatographic system for assay as 
follows:
    (1) Tailing factor. Calculate the tailing factor (T), from distances 
measured along the horizontal line at 5 percent of the peak height above 
the baseline, as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.073

where:
W0.05=Width of peak at 5 percent height; and
f=Horizontal distance from point of ascent to a point coincident with 
          maximum peak height.

    (2) Efficiency of the column. Calculate the number of theoretical 
plates (n) of the column by either of the following formulas:
[GRAPHIC] [TIFF OMITTED] TC01AP94.020

where:
n=Efficiency, as number of theoretical plates for column;
tR=Retention time of solute;
wh=Peak width at half-height; and
W=Width of the base of the peak obtained by extrapolating the relatively 
          straight sides of the peak to the baseline.


[[Page 445]]


    (3) Resolution factor. Calculate the resolution factor (R) as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.074

where:
tRj=Retention time for a solute eluting after i (tRj 
          is larger than tRi);
tRi=Retention time for any solute;
wi=Width of peak at baseline for any solute; and
wj=Width of peak at baseline for any solute eluting after i.

    (4) Coefficient of variation (relative standard deviation). 
Calculate the coefficient of variation (SR
[GRAPHIC] [TIFF OMITTED] TC01AP94.021

where:
X is the mean of N individual measurements of Xi.

    If the complete operating system meets the system suitability 
requirements of the monograph for the drug being tested, proceed as 
described in paragraph (b) of this section, using the sample solution in 
lieu of the working standard solution.
[50 FR 7764, Feb. 26, 1985; 50 FR 10220, Mar. 14, 1985; 50 FR 18243, 
Apr. 30, 1985]



Sec. 436.354  High-performance liquid chromatographic assay for ceftriaxone.

    (a) Apparatus. A suitable high-performance liquid chromatograph 
equipped with:
    (1) A suitable detection system specified in the monograph for the 
drug being tested;
    (2) A suitable recording device of at least 25-centimeter 
deflection;
    (3) A suitable chromatographic data managing system; and
    (4) An analytical column, 3 to 30 centimeters long, packed with a 
material as defined in the monograph for the drug being tested.
    (b) Procedure. Perform the assay at the temperature specified in the 
monograph for the drug being tested with a flow rate not to exceed 2.0 
milliliters per minute, Use a detector sensitivity setting that gives a 
peak height for the working standard that is at least 50 percent of 
scale. Use the apparatus described in paragraph (a) of this section; and 
also, use the system suitability requirements, reagents, working 
standard, test and sample solutions, and calculations as directed in the 
individual monograph for the drug being tested. Equilibrate and 
condition the column by passage of 10 to 15 void volumes of mobile phase 
followed by five replicate injections of 20 microliters each of the test 
solution. Allow an operating time sufficiently long to obtain 
satisfactory separation and elution of the expected components after 
each injection. Record the peak responses and calculate the prescribed 
system suitability requirements as described for the system suitability 
test in paragraph (c) of this section.
    (c) System suitability test. Using the apparatus and procedure 
described in this section, test the chromatographic system for assay as 
follows:
    (1) Capacity factor. Calculate the capacity factor (k) as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.075
    
where:
tR=Retention time of solute; and
tM=Retention time of solvent or unretained substance.

    (2) Resolution. Calculate the resolution (R) as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.076
    
where:
tRj=Retention time for a solute eluting after i (tRj 
          is larger than tRi);
tRi=Retention time for any solute;
wi=Width of peak at baseline for any solute; and
wj=Width of peak at baseline for any solute eluting after i.

    (3) Asymmetry factor. Calculate the asymmetry factor (As
    [GRAPHIC] [TIFF OMITTED] TR01JA93.077
    
where:
a=Horizontal distance from point of ascent to point of maximum peak 
          height; and

[[Page 446]]

b=Horizontal distance from point of maximum peak height to point of 
          descent.

    (4) Efficiency of the column. Calculate the efficiency of the column 
(reduced plate height) (hr
    (i)
    [GRAPHIC] [TIFF OMITTED] TR01JA93.078
    
    (ii)
    [GRAPHIC] [TIFF OMITTED] TR01JA93.079
    
    (iii)
    [GRAPHIC] [TIFF OMITTED] TR01JA93.080
    
where:
n=Efficiency, as number of theoretical plates for column;
tR=Retention time of solute;
wh=Peak width at half-height;
h=Efficiency, as height equivalent to one theoretical plate;
L=Length of column; and
dp=Average diameter of particle in column.

    (5) Coefficient of variation (relative standard deviation). 
Calculate the coefficient of variation (SR
[GRAPHIC] [TIFF OMITTED] TC01AP94.022

where:
X is the mean of N individual measurements of Xi.

The complete operating system is acceptable for assay if it meets 
the system suitability requirements of the monograph for the drug being 
tested. If the complete operating system is acceptable, proceed as 
described in paragraph (b) of this section using the sample solution in 
lieu of the test solution. Calculate the drug content as described in 
the individual monograph for the drug being tested.
[50 FR 9999, Mar. 13, 1985]



Sec. 436.355  High-performance liquid chromatographic assay for ticarcillin-clavulanic acid.

    (a) Equipment. A suitable high-performance liquid chromatograph 
equipped with:
    (1) A suitable detection system specified in the monograph for the 
drug being testing;
    (2) A suitable recording device of at least 25-centimeter 
deflection;
    (3) A suitable chromatographic data managing system; and
    (4) An analytical column, 10 to 30 centimeters long, packed with a 
material as defined in the monograph for the drug being tested; and if 
specified in that monograph, the inlet of this column may be connected 
to a guard column, 3 to 5 centimeters in length, packed with the same 
material of 40 to 60 micrometers particle size.
    (b) Procedure. Perform the assay and calculate the drug content 
using the temperature, instrumental conditions, and calculations 
specified in the monograph for the drug being tested with a flow rate 
not to exceed 2.0 milliliters per minute. Use a detector sensitivity 
setting that gives a peak height for the working standard that is at 
least 50 percent of scale with typical chart speed of not less than 2.5 
millimeters per minute. Use the equipment described in paragraph (a) of 
this section; and the reagents and working standard and sample solutions 
described in the monograph for the drug being tested. Equilibrate and 
condition the column by passage of 10 to 15 void volumes of mobile phase 
followed by five replicate injections of the same volume (between 10 and 
20 microliters) of the working standard solution. Allow an operating 
time sufficiently long to obtain satisfactory separation and elution of 
the expected components after each injection. The clavulanic acid peak 
is sharp and the chromatograms of standard and sample solutions show 
baseline separations between it and any neighboring peaks. The retention 
times for clavulanic acid and ticarcillin are approximately 3 minutes 
and 14 minutes, respectively. Record the peak responses and calculate 
the prescribed system suitability requirements described for the system 
suitability test in paragraph (c) of this section.

[[Page 447]]

    (c) System suitability test. Using the equipment and procedure 
described in this section, test the chromatographic system for assay as 
follows:
    (1) Tailing factors for the ticarcillin and clavulanic acid peaks. 
Calculate the tailing factors (T), from distances measured along the 
horizontal line at 5 percent of the peak height above the baseline, as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.081

where:
W0.05=Width of peak at 5 percent height; and
f=Horizontal distance from point of ascent to a point coincident with 
          maximum peak height.

    (2) Efficiency of the column. Calculate the number of theoretical 
plates (n) of the column as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.082

where:
n=Efficiency, as number of theoretical plates for column;
tR=Retention time of ticarcillin or clavulanic acid peaks; 
          and
wh=Corresponding peak width at half-height.

    (3) Resolution factor. Calculate the resolution factor (R) as 
follows
[GRAPHIC] [TIFF OMITTED] TR01JA93.083

where:
t1=Retention time of clavulanic acid peak;
t2=Retention time of ticarcillin peak; and
w1 and w2=Widths of the bases of the corresponding 
          peaks obtained by extrapolating the relatively straight sides 
          of the peaks to the baseline.

When using the method to assay clavulanic acid alone, the resolution 
factor is not applicable.
    (4) Coefficient of variation (Relative standard deviation). 
Calculate the coefficient of variation (SR
[GRAPHIC] [TIFF OMITTED] TC01AP94.023

where:
X is the mean of N individual measurements of Xi.


If the complete operating system meets the system suitability 
requirements of the monograph for the drug being tested, proceed as 
described in paragraph (b) of this section, using the sample solution in 
lieu of the working standard solution.
[50 FR 33517, Aug. 20, 1985; 50 FR 43384, Oct. 25, 1985]



Sec. 436.356  High-performance liquid chromatographic assay for ceftazidime.

    (a) Equipment. A suitable high-performance liquid chromatograph 
equipped with:
    (1) A suitable detection system specified in the monograph for the 
drug being tested;
    (2) A suitable recording device of at least 25-centimeter 
deflection;
    (3) A suitable chromatographic data managing system; and
    (4) An analytical column, 3 to 30 centimeters long, packed with a 
material as defined in the monograph for the drug being tested; and if 
specified in that monograph, the inlet of this column may be connected 
to a guard column, 3 to 5 centimeters in length, packed with the same 
material of 40 to 60 micrometers particle size.
    (b) Procedure. Perform the assay and calculate the drug content 
using the temperature, instrumental conditions, flow rate, and 
calculations specified in the monograph for the drug being tested. Use a 
detector sensitivity setting that gives a peak height for the working 
standard that is at least 50 percent of scale with typical chart speed 
of not less than 2.5 millimeters per minute.

[[Page 448]]

Use the equipment described in paragraph (a) of this section. Use the 
reagents, working standard solution, and sample solution described in 
the monograph for the drug being tested. Equilibrate and condition the 
column by passage of 10 to 15 void volumes of mobile phase followed by 
five replicate injections of the same volume (between 10 and 20 
microliters) of the working standard solution for the system suitability 
test. Allow an operating time sufficiently long to obtain satisfactory 
separation and elution of the expected components after each injection. 
Record the peak responses and calculate the prescribed system 
suitability requirements described for the system suitability test in 
paragraph (c) of this section.
    (c) System suitability test. Select the system suitability 
requirements specified in the monograph for the drug being tested. Then, 
using the equipment and procedure described in this section, test the 
chromatographic system for assay as follows:
    (1) Tailing factor. Calculate the tailing factor (T), from distances 
measured along the horizontal line at 5 percent of the peak height above 
the baseline, as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.084

where:
W0.05=Width of peak at 5 percent height; and
f=Horizontal distance from point of ascent to a point coincident with 
          maximum peak height.

    (2) Efficiency of the column. Calculate the number of theoretical 
plates (n) of the column as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.085

where:
n=Efficiency, as number of theoretical plates for column;
tR=Retention time of solute; and
wh=Peak width at half-height.

    (3) Resolution. Calculate the resolution (R) as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.086
    
where:
tRj=Retention time of a solute eluting after i(tRj 
          is larger than tRi);
tRi=Retention time of any solute;
wi=Width of peak at baseline measured by extrapolating the 
          relatively straight sides to the baseline of any solute; and
wj=Width of peak at baseline measured by extrapolating the 
          relatively straight sides to the baseline of any solute 
          eluting after i.

    (4) Coefficient of variation (relative standard deviation). 
Calculate the coefficient of variation (SR in percent) as 
follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.024

where:
X is the mean of N individual measurements of Xi.

If the complete operating system meets the system suitability 
requirements of the monograph for the drug being tested, proceed as 
described in paragraph (b) of this section, except alternate injections 
of the working standard solution with injections of the sample solution.
[50 FR 48397, Nov. 25, 1985]



Sec. 436.357  Atomic absorption test for sodium carbonate content.

    (a) Equipment. A suitable atomic absorbance spectrophotometer 
equipped with:
    (1) A suitable sodium hollow-cathode discharge lamp;
    (2) An oxidizing air-acetylene flame;
    (3) A nebulizer-burner system;
    (4) An optical dispersing device capable of isolating a resonance 
line of sodium from other wavelengths produced by the emission source; 
and
    (5) A suitable radiation detector.
    (b) Ionization buffer. Dissolve and dilute 19.07 grams of potassium 
chloride in distilled water to 1,000 milliliters.
    (c) Preparation of reference standard and sample solutions--(1) 
Reference

[[Page 449]]

standard solution. Accurately weigh approximately 140 milligrams of 
sodium chloride, which has been previously dried for 40 to 50 minutes at 
a temperature of 500 to 650  deg. C. Dissolve and dilute with sufficient 
distilled water to obtain a stock solution containing 5.5 micrograms of 
sodium per milliliter. Mix 10 milliliters of the stock solution with 10 
milliliters of ionization buffer and dilute the mixture with distilled 
water to obtain a solution containing 0.55 microgram of sodium per 
milliliter.
    (2) Sample solution. Dilute the stock sample solution, prepared as 
directed in the monograph for the drug being tested, with distilled 
water to obtain a solution containing 5.5 micrograms of sodium per 
milliliter (estimated). Mix 10 milliliters of this solution with 10 
milliliters of ionization buffer and dilute the mixture with distilled 
water to obtain a solution containing 0.55 microgram of sodium per 
milliliter (estimated).
    (3) Procedure. Determine the atomic absorbance of the reference 
standard and sample solutions at a wavelength of 589 nanometers, using 
the atomic absorbance spectrophotometer and a reagent blank prepared by 
diluting 10 milliliters of ionization buffer to 100 milliliters with 
distilled water.
    (d) Calculations. Calculate the percent sodium carbonate (S) as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.087

where:
Au=Absorbance of sodium in the sample solution;
As=Absorbance of sodium in the reference standard solution;
Ps=Sodium concentration in the reference standard solution in 
          micrograms per milliliter; and
Cu=Milligrams of sample per milliliter of sample solution.
[50 FR 48398, Nov. 25, 1985, as amended at 54 FR 20785, May 15, 1989]



Sec. 436.358  High-performance liquid chromatographic assay for pyridine.

    (a) Equipment. A suitable high-performance liquid chromatograph 
equipped with:
    (1) A suitable detection system specified in the monograph for the 
drug being tested;
    (2) A suitable recording device of at least 25-centimeter 
deflection;
    (3) A suitable chromatographic data managing system; and
    (4) An analytical column, 15 to 25 centimeters long, packed with a 
material as defined in the monograph for the drug being tested; and if 
specified in that monograph, the inlet of this column may be connected 
to a guard column, 3 to 5 centimeters in length, packed with the same 
material of 40 to 60 micrometers particle size.
    (b) Procedure. Perform the assay and calculate the pyridine content 
using the temperature, instrumental conditions, flow rate, and 
calculations specified in the monograph for the drug being tested. Use a 
detector sensitivity setting that gives a peak height for the working 
standard that is at least 25 percent of scale with typical chart speed 
of not less than 2.5 millimeters per minute. Use the equipment described 
in paragraph (a) of this section. Use the reagents, working standard 
solution, and sample solution described in the monograph for the drug 
being tested. Equilibrate and condition the column by passage of 10 to 
15 void volumes of mobile phase followed for the system suitability test 
by five replicate injections of the same volume (between 10 and 20 
microliters) of the system suitability test solution. Allow an operating 
time sufficiently long to obtain satisfactory separation and elution of 
the expected components after each injection. Record the peak responses 
and calculate the prescribed system suitability requirements described 
for the system suitability test in paragraph (c) of this section.
    (c) System suitability test. Select the system suitability 
requirements specified in the monograph for the drug being tested. Then, 
using the equipment and procedure described in this section, test the 
chromatographic system for assay as follows:
    (1) Tailing factor. Calculate the tailing factor for the pyridine 
peak (T), from distances measured along the horizontal line at 5 percent 
of the peak height above the baseline, as follows:

[[Page 450]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.088


where:
W0.05=Width of peak at 5 percent height; and
f=Horizontal distance from point of ascent to a point coincident with 
          maximum peak height.

    (2) Resolution. Calculate the resolution (R) as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.089
    
where:
tRj=Retention time of t-butyl ceftazidime;
tRi=Retention time of pyridine;
wi=Width of pyridine peak at the baseline measured by 
          extrapolating the relatively straight sides to the baseline; 
          and
wj=Width of t-butyl ceftazidime peak at the baseline measured 
          by extrapolating the relatively straight sides to the 
          baseline.

    (3) Coefficient of variation (relative standard deviation). 
Calculate the coefficient of variation for the pyridine peak (SR 
in percent ) as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.025

where:
X is the mean of N individual measurements of Xi.

If the complete operating system meets the system suitability 
requirements of the monograph for the drug being tested, proceed as 
described in paragraph (b) of this section, except alternate injections 
of the working standard solution with injections of the sample solution.
[50 FR 48398, Nov. 25, 1985; 50 FR 53308, Dec. 31, 1985]



Sec. 436.360  Gel permeation chromatographic assay for high molecular weight polymer.

    (a) Equipment. A suitable gel permeation chromatograph equipped 
with.
    (1) A suitable detection system specified in the monograph for the 
drug being tested;
    (2) A suitable recording device of at least 25-centimeter 
deflection;
    (3) A suitable chromatographic data managing system; and
    (4) An analytical column, 50 centimeters long and 9 millimeters 
internal diameter, packed with a material as defined in the monograph 
for the drug being tested.
    (b) Procedure. Perform the assay and calculate the high molecular 
weight polymer content using the temperature, instrumental conditions, 
and calculations specified in the monograph for the drug being tested. 
Use a detector sensitivity setting that gives a peak height for the 
working standard that is at least 10 percent of scale with a typical 
chart speed of not less than 2.5 millimeters per minute. Use the 
equipment described in paragraph (a) of this section. Use the reagents, 
working standard solution, and sample solution described in the 
monograph for the drug being tested. Equilibrate and condition the 
column by passage of mobile phase for not less than 18 hours, removing 
any voids that may form at the top of the column, followed by five 
replicate injections of the same volume (100 microliters) of the blue 
dextran system suitability test solution. Allow an operating time 
sufficiently long to obtain satisfactory separation and elution of the 
expected components after each injection. Record the peak responses and 
calculate the prescribed system suitability requirements described for 
the system suitability test in paragraph (c) of this section.
    (c) System suitability test. Select the system suitability 
requirements specified in the monograph for the drug being tested. Then, 
using the equipment and procedure described in this section, test the 
chromatographic system for assay as follows:
    (1) Tailing factor. Calculate the tailing factor (T), from distances 
measured along the horizontal line at 5 percent of the peak height above 
the baseline, as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.090

where:
W0.05=Width of peak at 5 percent height; and
f=Horizontal distance from point of ascent to

[[Page 451]]

          a point coincident with maximum peak height.

    (2) Efficiency of the column. Calculate the number of theoretical 
plates (n) of the column as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.091

where:
n=Efficiency, as number of theoretical plates for column;
tR=Retention time of solute; and
wh=Peak width at half-height.

    (3) Coefficient of variation (relative standard deviation). 
Calculate the coefficient of variation (SR in percent) as 
follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.026

where:
X is the mean of N individual measurements of Xi.


If the complete operating system meets the system suitability 
requirements of the monograph for the drug being tested, proceed as 
described in paragraph (b) of this section, using two injections of the 
same volume (100 microliters) of the working standard solution followed 
by one injection of the same volume (100 microliters) of the sample 
solution.
[50 FR 48398, Nov. 25, 1985; 51 FR 1367, Jan. 13, 1986]



Sec. 436.361  High-performance liquid chromatographic assay for aztreonam.

    (a) Equipment. A suitable high-performance liquid chromatograph 
equipped with:
    (1) A suitable detection system specified in the monograph for the 
drug being tested;
    (2) A suitable recording device of at least 25-centimeter 
deflection;
    (3) A suitable chromatographic data managing system; and
    (4) An analytical column, 3 to 30 centimeters long, packed with a 
material as defined in the monograph for the drug being tested; and if 
specified in that monograph, the inlet of this column may be connected 
to a guard column, 3 to 5 centimeters in length, packed with the same 
material of 40 to 60 micrometers particle size.
    (b) Procedure. Perform the assay and calculate the drug content 
using the temperature, instrumental conditions, flow rate, and 
calculations specified in the monograph for the drug being tested. Use a 
detector sensitivity setting that gives a peak height for the working 
standard that is at least 50 percent of scale with typical chart speed 
of not less than 2.5 millimeters per minute. Use the equipment described 
in paragraph (a) of this section. Use the reagents, working standard 
solution, and sample solution described in the monograph for the drug 
being tested. Equilibrate and condition the column by passage of 10 to 
15 void volumes of mobile phase followed by five replicate injections of 
the same volume (between 10 and 20 microliters) of the working standard 
solution. Allow an operating time sufficiently long to obtain 
satisfactory separation and elution of the expected components after 
each injection. Record the peak responses and calculate the prescribed 
system suitability requirements described for the system suitability 
test in paragraph (c) of this section.
    (c) System suitability test. Select the system suitability 
requirements specified in the monograph for the drug being tested. Then, 
using the equipment and procedure described in this section, test the 
chromatographic system for assay as follows:
    (1) Tailing factor. Calculate the tailing factor (T), from distances 
measured along the horizontal line at 5 percent of the peak height above 
the baseline, as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.092

where:
W0.05=Width of peak at 5 percent height; and
f=Horizontal distance from point of ascent to a point coincident with 
          maximum peak height.

    (2) Efficiency of the column. Calculate the number of theoretical 
plates (n) of the column as follows:

[[Page 452]]

[GRAPHIC] [TIFF OMITTED] TC01AP94.027


where:
n=Efficiency, as number of theoretical plates for column;
tR=Retention time of solute; and
wh=Peak width at half-height.

    (3) Resolution. Calculate the resolution (R) as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.093
    
where:
tRj=Retention time of a solute eluting after i (tRj 
          is larger than tRi);
tRi=Retention time of any solute;
wi=Width of peak at baseline of any solute; and
wj=Width of peak at baseline of any solute eluting after i.

    (4) Coefficient of variation (relative standard deviation). 
Calculate the coefficient of variation (SR in percent ) as 
follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.028

where:
X is the mean of N individual measurements of Xi.

If the complete operating system meets the system suitability 
requirements of the monograph for the drug being tested, proceed as 
described in paragraph (b) of this section, except alternate injections 
of the working standard solution with injections of the sample solution.
[52 FR 4611, Feb. 13, 1987; 52 FR 8550, Mar. 18, 1987]



Sec. 436.362  Thin-layer chromatographic test for free erythromycin content in erythromycin estolate bulk.

    (a) Equipment--(1) Chromatography tank. A rectangular tank 
approximately 23 centimeters long, 23 centimeters high, and 9 
centimeters wide, equipped with a glass solvent trough in the bottom and 
a tight-fitting cover for the top.
    (2) Plates. Use a 20- by 20-centimeter precoated silica gel 60 F-254 
thin-layer chromatography plate. Before using, place the plate in an 
unlined developing chamber containing approximately 100 milliliters of 
anhydrous methanol and allow the solvent front to travel to the top of 
the plate, marking the direction of travel. Remove the plate and allow 
to drip dry. Store in a dry place.
    (b) Reagents--(1) Developing solvent. Mix 15 milliliters of 
chloroform and 85 milliliters of anhydrous methanol. Use fresh 
developing solvent for each test.
    (2) Spray solution. Dissolve 150 milligrams of xanthydrol in a 
mixture of 7.5 milliliters of glacial acetic acid and 92.5 milliliters 
of 37 percent hydrochloric acid.
    (c) Preparation of spotting solutions--(1) Sample solution. Prepare 
a solution of the sample in anhydrous methanol to contain 10 milligrams 
per milliliter.

    Note:  It is advisable to prepare the sample and standard solutions 
immediately before spotting to minimize the possibility of degradation 
in solution.)

    (2) Standard solution. Prepare a solution of erythromycin base 
reference standard in anhydrous methanol to contain 1 milligram per 
milliliter. Weigh 99.5, 99.0, and 97.0 milligrams of erythromycin 
estolate (propionyl erythromycin lauryl sulfate) reference standard and 
transfer to separate 10-milliliter volumetric flasks. To these flasks 
add 0.5, 1.0, and 3.0 milliliters, respectively, of the 1-milligram-per-
milliliter solution of erythromycin base reference standard and dilute 
to volume with anhydrous methanol. These solutions contain, 
respectively, 0.5 percent, 1.0 percent, and 3.0 percent erythromycin 
base in erythromycin estolate. Prepare a solution of erythromycin 
estolate reference standard in anhydrous methanol to contain 10 
milligrams per milliliter. Prepare a solution of erythromycin base 
reference standard in anhydrous methanol to contain 0.1 milligram per 
milliliter.
    (d) Procedure. Pour 100 milliliters of developing solvent into the 
glass trough on the bottom of the unlined chromatography tank. Cover and 
seal the tank. Allow it to equilibrate while the plate is being 
prepared. Prepare a

[[Page 453]]

plate as follows: On a line 2.0 centimeters from the base of the thin-
layer plate, apply 1.0 microliter of each of the following solutions:
    (1) 10-milligrams-per-milliliter solution of erythromycin estolate 
reference standard, equivalent to 10 micrograms of erythromycin 
estolate;
    (2) 0.5 percent base-in-estolate solution, equivalent to 0.05 
microgram of base and 9.95 micrograms of estolate;
    (3) 1.0 percent base-in-estolate solution, equivalent to 0.10 
microgram of base and 9.90 micrograms of estolate;
    (4) 3.0 percent base-in-estolate solution, equivalent to 0.30 
microgram of base and 9.70 micrograms of estolate;
    (5) 0.1-milligram-per-milliliter solution of erythromycin base 
reference standard, equivalent to 0.1 microgram of erythromycin base; 
and
    (6) Sample solution, equivalent to 10 micrograms of erythromycin 
estolate. Allow the spots to dry. Place the plate directly in the 
chromatograph tank. Cover and seal the tank. Allow the solvent front to 
travel a distance of 7 centimeters (about 27 minutes). Remove the plate 
from the tank, and allow it to air dry under a hood. With the plate 
still under the hood, spray uniformly with the spray solution. Heat the 
sprayed plate in an oven at 100  deg.C for 5 minutes. (CAUTION: Avoid 
exposure to the acid fumes while removing the plate from the oven.)
    (e) Evaluation. Erythromycin base and erythromycin estolate appear 
as reddish-violet spots on the sprayed and heated plate. Better 
visualization of the erythromycin base spots may be gained by viewing 
the plate under long-wavelength (366 nanometers) ultraviolet light, 
erythromycin base appearing as dark spots on a yellow-green fluorescent 
background. Erythromycin base has an Rfvalue of about 0.3. 
Erythromycin estolate has an Rf value of about 0.7. Compare 
the size and intensity of any erythromycin base spots in the sample lane 
with the erythromycin base spots in the erythromycin base reference 
standard lane and in the 0.5 percent, 1.0 percent, and 3.0 percent base-
in-estolate lanes, and report the percentage of erythromycin base (free 
erythromycin) in the sample. For a more accurate determination of free 
erythromycin content, it may be necessary to repeat the test using a 
different set of standards.
[53 FR 1919, Jan. 25, 1988]



Sec. 436.363  High-performance liquid chromatographic assay for cefmenoxime.

    (a) Apparatus. A suitable high-performance liquid chromatograph 
equipped with:
    (1) A suitable detection system specified in the monograph for the 
drug being tested;
    (2) A suitable recording device of at least 18-centimeter 
deflection;
    (3) A suitable chromatographic data managing system; and
    (4) An analytical column, 3 to 30 centimeters long, packed with a 
material as defined in the monograph for the drug being tested; and if 
specified in that monograph, the inlet of this column may be connected 
to a guard column, 3 to 5 centimeters in length, packed with the same 
material of 30 to 60 micrometers particle size.
    (b) Procedure. Perform the assay and calculate the drug content 
using the temperature, instrumental conditions, and calculations 
specified in the monograph for the drug being tested with a flow rate 
not to exceed 2.0 milliliters per minute. Use a detector sensitivity 
setting that gives a peak height for the working standard that is at 
least 50 percent of scale with typical chart speed of not less than 2.5 
millimeters per minute. Use the apparatus described in paragraph (a) of 
this section; and the reagents and working standard and sample solutions 
described in the monograph for the drug being tested. Equilibrate and 
condition the column by passage of 10 to 15 void volumes of mobile phase 
followed by 5 replicate injections of the same volume (between 10 and 20 
microliters) of the working standard solution. Allow an operating time 
sufficiently long to obtain satisfactory separation and elution of the 
expected components after each injection. Record the peak responses and 
calculate the prescribed system suitability requirements described for 
the system suitability test in paragraph (c) of this section.
    (c) System suitability test. Using the apparatus and procedure 
described in

[[Page 454]]

this section, test the chromatographic system for assay as follows:
    (1) Tailing factor. Calculate the tailing factor (T), from distances 
measured along the horizontal line at 5 percent of the peak height above 
the baseline, as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.094

where:
W0.05=Width of peak at 5 percent height; and
f=Horizontal distance from point of ascent to a point coincident with 
          maximum peak height.

    (2) Efficiency of the column. Calculate the number of theoretical 
plates (n) of the column as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.029

where:
n=Efficiency, as number of theoretical plates for column;
tR=Retention time of solute; and
wh=Peak width at half-height.

    (3) Resolution. Calculate the resolution (R) as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.095
    
where:
tRJ=Retention time of a solute eluting after i 
          (tRJ is larger than tRi);
tRi=Retention time of any solute;
wi=Width of peak at baseline of any solute; and
wJ=Width of peak at baseline of any solute eluting after i.

    (4) Coefficient of variation (Relative standard deviation).
    Calculate the coefficient of variation (SR) in percent) 
as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.381

where:
X is the mean of N individual measurements of Xi. If the 
          complete operating system meets the system suitability 
          requirements of the monograph for the drug being tested, 
          proceed as described in paragraph (b) of this section, using 
          the sample solution in lieu of the working standard solution.
[53 FR 13401, Apr. 25, 1988; 53 FR 19368, May 27, 1988]



Sec. 436.364  Atomic absorption test for sodium carbonate content of cefmenoxime hydrochloride for injection.

    (a) Apparatus. A suitable atomic absorbance spectrophotometer 
equipped with:
    (1) A suitable sodium hollow-cathode discharge lamp;
    (2) An oxidizing air-acetylene flame;
    (3) A nebulizer-burner system;
    (4) An optical dispersing device capable of isolating a resonance 
line of sodium from other wavelengths produced by the emission source; 
and
    (5) A suitable radiation detector.
    (b) Reagents. Ionization buffer: Dissolve 19.07 grams of potassium 
chloride in distilled water and dilute to 1,000 milliliters.
    (c) Preparation of reference standard and sample solutions--(1) 
Reference standard solution. Accurately weigh approximately 140 
milligrams of sodium chloride which has been previously dired for 40 to 
50 minutes at a temperature of 500 to 650  deg. C. Dissolve and dilute 
with sufficient distilled water to obtain a stock solution containing 
5.5 micrograms of sodium per milliliter. Mix 10 milliliters of the stock 
solution with 10 milliliters of ionization buffer and dilute the mixture 
with distilled water to obtain a solution containing 0.55 microgram of 
sodium per milliliter.
    (2) Sample solution. Dilute the sample solution used in 
Sec. 442.222(b)(1)(ii)(B)(1) of this chapter, with sufficient distilled 
water to obtain a stock solution containing 5.5 micrograms of sodium per 
milliliter (estimated). Mix 10 milliliters of the stock solution with 10 
milliliters of ionization buffer and dilute the mixture with distilled 
water to obtain a solution containing 0.55 microgram of sodium per 
milliliter (estimated).
    (3) Procedure. Determine the atomic absorbance of the reference 
standard and sample solutions at a wavelength

[[Page 455]]

of 589 nanometers, using the atomic absorbance spectrophotometer and a 
reagent blank prepared by diluting 10 milliliters of ionization buffer 
to 100 milliliters with distilled water.
    (d) Calculations. Calculate the percent sodium carbonate as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.096
    
where:
Au=Absorbance of sodium in the sample solution;
As=Absorbance of sodium in the reference standard solution;
Ps=Milligrams of sodium chloride per milliliter of the 
          reference standard solution;
Cu=Milligrams of sample per milliliter of sample solution; 
          and
d= Dilution factor of the sample.
[53 FR 13401, Apr. 25, 1988]



Sec. 436.365  Thin layer chromatographic identity test for rifampin.

    (a) Equipment--(1) Chromatography tank. Use a rectangular tank 
approximately 23 x 23 x 9 centimeters, with a glass solvent trough on 
the bottom and a tight-fitting cover, lined with Whatman 3MM 
chromatographic paper or equivalent.
    (2) Plates. Use 20 x 20 centimeter thin layer chromatography plates 
coated with silica gel 60 F-254 or equivalent to a thickness of 250 
microns.
    (b) Developing solvent. Mix chloroform and methanol in volumetric 
proportions of 90:10, respectively.
    (c) Spotting solutions--(1) Preparation of working standard 
solution. Dissolve approximately 50 milligrams of rifampin working 
standard in 5 milliliters of chloroform.
    (2) Preparation of sample solution. Dissolve the contents of a 
sample vial in 60 milliliters of chloroform.
    (d) Procedure. Pour the developing solvent into the glass trough on 
the bottom of the tank and onto the paper lining the walls of the tank. 
Cover and seal the tank. Allow it to equilibrate. Prepare a plate as 
follows: On a line 2.5 centimeters from the base of the thin layer 
chromatography plate and at intervals of 2.0 centimeters, spot 3 
microliters of the working standard solution to points 1 and 3. When 
these spots are dry, apply 3 microliters of the sample solution to 
points 2 and 3. After all the spots are thoroughly dry, place the plate 
into the trough in the bottom of the tank. Cover and tightly seal the 
tank. Allow the solvent front to travel about 7 centimeters from the 
starting line. Remove the plate from the tank and air dry.
    (e) Evaluation. Measure the distance the solvent front traveled from 
the starting line, and the distance the red spots are from the starting 
line. Divide the latter by the former to calculate the Rf 
value.
[54 FR 38375, Sept. 18, 1989; 54 FR 42886, Oct. 18, 1989]



Sec. 436.366  High-performance liquid chromatography assay for determining chromatographic purity of vancomycin.

    (a) Apparatus. A suitable high-performance liquid chromatograph 
equipped with:
    (1) A suitable ultraviolet detection system operating at a 
wavelength of 254 nanometers or preferably 280 nanometers;
    (2) A suitable recording device of at least 25-centimeter 
deflection;
    (3) A suitable chromatographic data managing system; and
    (4) A 25-centimeter analytical column having an inside diameter of 
4.6 millimeters and packed with octadecyl silane chemically bonded to 
porous silica or ceramic microparticles; 5 micrometers in diameter.
    (b) Reagents--(1) 0.2 percent triethylammonium phosphate buffer. To 
2,000 milliliters of distilled water, either add 4 milliliters of 
triethylamine or 4 grams of triethylammonium chloride. Adjust the pH to 
3.2 with phosphoric acid.
    (2) Sample solvents. (i) Vancomycin hydrochloride: Mobile Phase A.

[[Page 456]]

    (ii) Vancomycin base: 5 milliliters Mobile Phase A; add 0.1N HCl 
dropwise with swirling until sample dissolves: dilute to volume with 
Mobile Phase A.
    (c) Mobile Phases--(1) Mobile Phase A. Add 70 milliliters of 
acetonitrile and 10 milliliters of tetrahydrofuran to 920 milliliters of 
0.2 percent triethylammonium phosphate buffer and mix well. Filter the 
mobile phase through a suitable glass fiber filter or equivalent that is 
capable of removing particulate contamination to 1 micron in diameter. 
Degas the mobile phase, briefly, just prior to its introduction into the 
chromatographic pumping system.
    (2) Mobile Phase B. Add 290 milliliters of acetonitrile and 10 
milliliters of tetrahydofuran to 700 milliliters of 0.2 percent 
triethylammonium phosphate buffer and mix well. Filter the mobile phase 
through a suitable glass fiber filter or equivalent that is capable of 
removing particulate contamination to 1 micron in diameter. Degas the 
mobile phase, briefly, just prior to its introduction into the 
chromatographic pumping system.
    (d) Operating conditions. Perform the assay at ambient temperature 
with a typical flow rate of about 2.0 milliliters per minute. Use a 
detector sensitivity setting that gives a peak height for the main peak 
(Vancomycin B) that is at least 50 percent of scale. The run time is 30 
minutes per injection and the gradient conditions are as follows: (0, 
12, 12.5, 8, 0, 2)

                                                                        
------------------------------------------------------------------------
                          Mobile     Mobile                             
    Time (minutes)       phase A    phase B       Gradient condition    
                        (percent)  (percent)                            
------------------------------------------------------------------------
0.....................        100          0  Initial conditions.       
12....................        100          0  Isocratic region.         
20....................          0        100  Linear ramp.              
22....................          0        100  Isocratic region.         
23....................        100          0  Return to initial.        
30....................        100          0  Reequilibration.          
------------------------------------------------------------------------

    (e) Preparation of resolution and sample solutions--(1) Resolution 
solution. Prepare a solution of vancomycin hydrochloride reference 
standard in water containing 0.5 milligram per milliliter. Heat at 65 
deg.C for 24 hours and allow to cool. This procedure generates two 
desamido-vancomycin isomers. The first desamido isomer elutes during the 
isocratic period and before the vancomycin B peak; the second desamido 
isomer elutes during the gradient ramp and is used to demonstrate the 
effective performance of this stage.
    (2) Sample preparation. In a volumetric flask either dissolve a 
representative sample or dilute a representative portion with sample 
solvent to give a sample preparation containing approximately 10 
milligrams per milliliter. Pipet 2 milliliters of this sample solution 
into a separate 50-milliliter volumetric flask and dilute to volume with 
sample solvent to give a diluted sample preparation containing 
approximately 0.4 milligram per milliliter.
    (f) Procedure. Optimize chromatographic conditions under isocratic 
conditions by equilibrating the system while pumping 100 percent mobile 
phase A through the column. Inject 20 microliters of the resolution 
solution onto the column and record the chromatogram. Adjust the 
acetonitrile concentration of mobile phase A as needed to provide a 
retention time for vancomycin B of 7.5 to 10.5 minutes. Use the 
resolution solution to perform the system suitability tests. The elution 
order is resolution compound 1, vancomycin B, resolution compound 2. 
Return the system to the initial gradient operating conditions. 
Separately inject 20 milliliters of each diluted (0.4 milligram per 
milliliter) and concentrated (10 milligrams per milliliter) sample 
solution onto the column and record each chromatogram.
    (g) System suitability test. Using the resolution solution described 
in paragraph (e)(1) of this section, test the performance of the 
chromatographic system as follows:
    (1) Asymmetry factor. Calculate the asymmetry factor (A), measured 
at a point that is 10 percent of the vancomycin B peak height from the 
baseline, as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.097

where:
    a=Horizontal distance from point of ascent to point of maximum peak 
height; and
    b=Horizontal distance from point of maximum peak height to point of 
descent.

[[Page 457]]

    The asymmetry factor (Ar) is satisfactory if it is not 
less than 0.8 and not more than 1.8.

    (2) Efficiency of the column. From the number of theoretical plates 
(n) calculated as described in Sec. 436.216(c)(2) calculate the reduced 
plate height (hr) for the vancomycin B peak as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.098

where:
    L=Length of the column in centimeters;
    n=Number of theoretical plates; and
    dp=Average diameter of the particles in the column in 
micrometers.

The absolute efficiency (hr) is satisfactory if it is not 
more than 40 for the vancomycin B peak in the resolution solution.

    (3) Resolution. The resolution (R) between the vancomycin B peak and 
the peak for resolution compound 1 is not less than 3.0. Resolution 
compound 2 is eluted between 3 and 6 minutes after the start of the 
period when the percentage of mobile phase B is increasing from 0 
percent to 100 percent.
    (4) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent) of five replicate 
injections of the resolution solution is calculated as described in 
Sec. 436.216(c)(4) is satisfactory if it is not more than 2.0 percent.
    (5) Capacity factor (k). Calculate the capacity factor (k) for 
vancomycin B as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.099

where:
    tr=Retention time of solute; and
    tm=Retention time of solvent or unretained substance, 
calculated as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.100

where:
    D=Column diameter in centimeters;
    L=Column length in centimeters;
    0.75=Average total column porosity; and
    F=Flow rate in milliliters per minute.

The capacity factor (k) for vancomycin B is satisfactory if it is not 
less than 2.6 and not more than 3.3.
    When the system suitability requirements have been met, then proceed 
as described in paragraph (f) of this section. Alternate chromatographic 
conditions are acceptable provided that the system suitability 
parameters are met. However, the sample preparation described in 
paragraph (e)(2) of this section should not be changed.

    (h) Calculations. (1) Calculate the percentage of vancomycin B in 
the specimen as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.101

where:
    AB=Area of the vancomycin B peak in the dilute (0.4 
milligram per milliliter) sample solution; and
    ATotal=Area of the vancomycin B peak in the dilute (0.4 
milligram per milliliter) solution+[Area of the total related substances 
peaks (exclude the area of the vancomycin B peak) in the concentrated 
solution (10 milligrams per milliliter) divided by 25].

    (2) Calculate the percentage of each other peak as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.102
    
where:
    Ai=Area of any given peak, other than the main peak in 
the concentrated solution (10 milligrams per milliliter); and
    ATotal=Area of the vancomycin B peak in the dilute (0.4 
milligram per milliliter) solution+[Area of the total related substances 
peaks (exclude the area of the vancomycin B peak) in the concentrated 
solution (10 milligrams per milliliter) divided by 25].
[54 FR 20383, May 11, 1989; 54 FR 25849, June 20, 1989]



Sec. 436.367  Thin-layer chromatographic identity test for cephalexin hydrochloride.

    (a) Equipment--(1) Chromatography tank. Use a rectangular tank 
approximately 23  x  23  x  9 centimeters, with a glass solvent trough 
in the bottom and a tight-fitting cover. Line the inside walls of the 
tank with Whatman 3 MM chromatographic paper or equivalent.
    (2) Plates. Use 20  x  20 centimeter thin layer chromatographic 
plates coated with silica gel 60F-254 or equivalent to a thickness of 
250 microns.
    (b) Developing solvent. Mix ethylacetate, acetonitrile, water and 
glacial acetic acid in volumetric proportions of 42:14:18:14, 
respectively.

[[Page 458]]

    (c) Preparation of the spotting solutions. Prepare a solution of the 
sample containing 25 milligrams per milliliter of cephalexin 
hydrochloride in water. Prepare a solution of cephalexin monohydrate 
reference material at a concentration of 25 milligrams per milliliter. 
Add water and 0.1N hydrochloric acid in a dropwise mode until the 
material is completely dissolved.
    (d) Procedure. Pour the developing solvent into the glass trough at 
the bottom of the chromatography tank. Cover and seal the tank. Allow it 
to equilibrate for 1 hour. Prepare a plate as follows: On a line 2 
centimeters from the base of the plate, and at intervals of 2 
centimeters, spot approximately 5 microliters of the standard solution 
to points 1 and 3 and approximately 5 microliters of the sample solution 
to point 2. After all spots are thoroughly dry, place the plate directly 
into the glass trough of the chromatography tank. Cover and seal the 
tank. Allow the solvent front to travel approximately 15 centimeters 
from the starting line. Remove the plate from the tank and allow it to 
air dry.
    (e) Evaluation. View the dry plate under ultraviolet light (254 
nanometers). Measure the distance the solvent front traveled from the 
starting line and the distance the spots are from the starting line. 
Calculate the Rf value by dividing the latter by the former. 
The sample and standard should have spots of corresponding Rf 
values of approximately 0.35.
[54 FR 48860, Nov. 28, 1989; 54 FR 51816, Dec. 18, 1989]



Sec. 436.368  Thin layer chromatographic identity test for cefprozil.

    (a) Equipment--(1) Chromatography tank. Use a glass rectangular tank 
approximately 23 x 23 x 9 centimeters lined with filter paper and 
equipped with a tight-fitting cover.
    (2) Plates. Use 20 x 20 centimeter thin layer chromatography plates 
coated with silica gel GF to a thickness of 250 microns.
    (b) Reagents--(1) Diluent. Mix 0.1N HCl and acetone in volumetric 
proportions of 1:4.
    (2) Developing solvent. Mix n-butanol, glacial acetic acid and water 
in volumetric proportions of 60:20:20.
    (3) Detection reagent. Iodine vapor.
    (c) Assay solutions--(1) Reference standard solution. Dissolve 50 
milligrams of cefprozil (Z) reference standard in 10 milliliters of 
diluent.
    (2) Sample solution. Place an amount of sample containing 
approximately 50 milligrams of cefprozil in a 20-milliliter glass 
stoppered vial. Add 10 milliliters of diluent. Shake for 5 minutes and 
allow the solids to settle.
    (d) Procedure. Pour a suitable quantity of the developing solvent 
into a glass, chromatographic tank lined with filter paper and allow to 
equilibrate for 1 hour. On a line 2 centimeters from the bottom edge of 
the plate, spot 10 microliters each of the reference solution and sample 
solution. Draw a line indicating the distance to which the developing 
solvent must travel at a point 12 centimeters from the bottom edge of 
the plate. Place the plate in the tank and allow the solvent to migrate 
to the finishing line. Remove the plate and air dry in a fume hood. 
Place the dried plate in a chamber containing iodine vapors.
    (e) Evaluation. Measure the distance the solvent front traveled from 
the starting line, and the distance the spots are from the starting 
line. Divide the latter by the former to calculate the Rf 
value. The identity test is positive if the sample solution produces a 
yellow spot at the same Rf value and has the same appearance 
as the spot obtained for the reference solution. The Rf value 
for cefprozil (Z) is approximately 0.45. Cefprozil (E), has an 
Rf value of approximately 0.47.Cefprozil (Z) is ``absent'' if 
the above test is performed and no spots, which correspond to those from 
the reference solution, are obtained for the sample.
[58 FR 26660, May 4, 1993]



Sec. 436.369  Thin layer chromatography test for free N-isobutylpiperidone content in rifabutin.

    (a) Equipment--(1) Chromatography tank. A rectangular tank, 
approximately 23 X 23 X 9 centimeters, with a glass solvent trough on 
the bottom and a tight-fitting cover.
    (2) Iodine vapor chamber. A rectangular tank, approximately 23 X 23 
X 9 centimeters, with a suitable cover, containing iodine crystals.

[[Page 459]]

    (3) Plates. Use 20 X 20 centimeter thin layer chromatography plates 
coated with silica gel 60F 254 or equivalent to a thickness of 250 
microns.
    (b) Reagents--(1) Developing solvent. Mix petroleum ether (b.p. 60 
to 80  deg. C) and acetone in volumetric proportions of 100:30, 
respectively.
    (2) Spray solution. Prepare a 1 percent solution of soluble starch 
in water (containing 0.01 percent mercuric iodide).
    (c) Preparation of spotting solutions--(1) Sample solution. Prepare 
a solution of the rifabutin sample in 1:1 chloroform/methanol to contain 
10 milligrams per milliliter.
    (2) Standard solution. Prepare a solution of N-isobutylpiperidone 
standard in 1:1 chloroform/methanol to contain 1 milligram per 
milliliter. Transfer aliquots of 0.5, 1.0, 2.0, 5.0, and 10.0 
milliliters into separate 100-milliliter volumetric flasks and dilute to 
volume with 1:1 chloroform/methanol. These solutions contain, 
respectively, the equivalent of 0.05, 0.1, 0.2, 0.5, and 1.0 percent of 
N-isobutylpiperidone.
    (d) Procedure. Pour 100 milliliters of developing solvent into the 
glass trough on the bottom of the unlined chromatography tank. Cover and 
seal the tank. Allow it to equilibrate while the plate is being 
prepared. Prepare a plate as follows: on a line 2.0 centimeters from the 
base of the thin layer chromatography plate, and at intervals of 2.0 
centimeters, apply 10 microliters of each of the standard solutions and 
the sample solution prepared as directed above. After the spots are 
thoroughly dry, place the plate into the trough in the bottom of the 
tank. Cover and tightly seal the tank, allow the solvent front to travel 
about 15 centimeters from the starting line and then remove the plate 
from the tank. Air dry the plate. Warm the iodine vapor chamber to 
vaporize the iodine crystals and place the dry plate in the iodine vapor 
chamber until the spots are visible (usually about 5 minutes). Remove 
the plate from the iodine vapor chamber and spray with 1 percent starch 
solution.
    (e) Evaluation. Measure the distance the solvent front traveled from 
the starting line and the distance the spots are from the starting line. 
Calculate the Rf value by dividing the latter by the former. 
N-isobutylpiperidone has an Rf value of about 0.3. Rifabutin 
has an Rf value of about 0.1. Compare the size and intensity 
of any N-isobutylpiperidone spots in the sample lane with the N-
isobutylpiperidone spots in the standard lanes, and report the 
percentage of N-isobutylpiperidone in the sample.
[59 FR 40806, Aug. 10, 1994]



Sec. 436.370  Spectrophotometric identity test for rifabutin capsules.

    (a) Equipment. A suitable spectrophotometer capable of recording the 
ultraviolet spectrum in the 200 to 400 nanometer range, using suitable 
quartz cells of 1 centimeter pathlength.
    (b) Preparation of working standard and sample solution--(1) Working 
standard solution. Suspend approximately 200 milligrams of rifabutin 
working standard in 20 milliliters of methanol and sonicate for 
approximately 5 minutes. Filter the resulting solution through a 
suitable 0.5 micrometer filter. Transfer a 2-milliliter aliquot of the 
filtered solution to a 100-milliliter volumetric flask and fill to 
volume with methanol. Further dilute with methanol to obtain a solution 
containing 20 micrograms of rifabutin activity per milliliter.
    (2) Sample solution. Empty and combine the contents of five 
capsules. Suspend a quantity of the capsule contents equivalent to 200 
milligrams of rifabutin in 20 milliliters of methanol. Sonicate for 
about 5 minutes and then filter through an appropriate 0.5 micrometer 
filter. Transfer a 2-milliliter aliquot to a 100-milliliter volumetric 
flask and dilute to volume with methanol. Further dilute with methanol 
to obtain a solution containing 20 micrograms of rifabutin activity per 
milliliter (estimated).
    (c) Procedure. Using a suitable spectrophotometer equipped with 1.0 
centimeter cells and methanol as the blank, determine the absorbance 
spectra of the working standard and sample solutions over the 
ultraviolet range of 250 to 300 nanometers.
    (d) Evaluation. Compare the spectrum of the sample to that of the 
working standard. The identity of the rifabutin capsules is confirmed by 
quantitative comparison of the two spectra with an

[[Page 460]]

absorbance maximum being observed at about 275 nanometers.
[59 FR 40807, Aug. 10, 1994]



     Subpart G--Chemical Tests for Nonantibiotic Active Ingredients



Sec. 436.400  Thin layer chromatographic identity test for iodochlorhydroxyquin.

    (a) Equipment--(1) Chromatography tank. A rectangular tank, 
approximately 9  x  9  x  3.5 inches with a glass solvent trough on the 
bottom.
    (2) Plates. Use 20  x  20 centimeter thin layer chromatography 
plates coated with Silica Gel G or equivalent to a thickness of 250 
microns.
    (b) Developing solvent. Mix benzene and methanol in volumetric 
proportions of 90:10.
    (c) Preparation of spotting solutions--(1) Sample solution. Use the 
sample solution prepared as described in the section for the particular 
product to be tested.
    (2) Reference solution. Prepare a solution containing 0.5 milligram 
of iodochlorhydroxyquin U.S.P. reference standard per milliliter in 
acetone.
    (d) Procedure. Pour developing solvent into the glass trough on the 
bottom of the chromatography tank. Cover and seal the tank. Allow it to 
equilibrate for 1 hour. Spot a plate as follows: Apply approximately 10 
microliters each of the sample solution and of the reference solution on 
a line 2.0 centimeters from the base of the silica gel plate and at 
intervals of not less than 2.0 centimeters. After all spots are 
thoroughly dry, place the silica gel plate directly into the glass 
trough of the chromatography tank. Cover and reseal the tank. Allow the 
solvent front to travel about 15 centimeters from the starting line, 
remove the plate from the tank, and allow to air dry. Examine under a 
strong source of ultraviolet light. The sample and standard are visible 
as dark spots.
    (e) Evaluation. Measure the distance the solvent front traveled from 
the starting line and the distance the spots are from the starting line. 
Calculate the Rf value by dividing the latter by the former. 
The sample and standard should have spots of corresponding Rf 
values (0.55 to 0.60).



          Subpart H--Tests for Specific Antibiotic Dosage Forms



Sec. 436.500  Penicillin in oil and wax.

    (a) Potency. Proceed as directed in Sec. 440.80a(b)(1) of this 
chapter except paragraph (b)(1)(ix) thereof and, in lieu of the 
directions in Sec. 440.80a(b)(1)(iv) of this chapter prepare sample as 
follows: Liquefy the sample by warming, thoroughly mix, and withdraw 1.0 
milliliter using a sterile syringe equipped with an 18-gauge needle. 
Transfer to a separatory funnel containing approximately 50 milliliters 
of peroxide-free ether. Shake the separatory funnel vigorously to bring 
about complete mixing of the material with the ether. Shake with a 25-
milliliter portion of 1 percent phosphate buffer at pH 6.0. Remove the 
buffer layer and repeat the extraction with three 25-milliliter 
quantites of buffer. Combine the extracts and make the proper estimated 
dilutions in 1 percent phosphate buffer at pH 6.0. The sample may also 
be prepared by transferring aseptically 1.0 milliliter of the penicillin 
in oil and wax to a blending jar containing 100 milliliters of 1 percent 
phosphate buffer at pH 6.0. Using a high-speed blender, blend this 
mixture for 1 minute and then make the proper estimated dilutions in 1 
percent phosphate buffer at pH 6.0. If the label represents the potency 
of the penicillin in oil and wax as 200,000 units per milliliter or 
less, it is satisfactory if it is 85 percent or more of the potency so 
represented; if represented as more than 200,000 units per milliliter, 
it is satisfactory if it is 90 percent or more of the potency so 
represented.
    (b) Sterility. Proceed as directed in Sec. 436.20, using the method 
described in paragraph (e)(2) of that section, except using medium B in 
lieu of medium A.
    (c) Moisture--(1) Reagents--(i) KarlFischer reagent. Preserve the 
reagent in glass-stoppered bottles and use from an all glass automatic 
burette, protecting the solution from the moisture in the air.
    (ii) Water-methanol solution. Use methanol containing approximately 
1 mg. of water per milliliter. Store the solution in a glass bottle 
attached to an automatic burette and protect from moisture in the air at 
all times.

[[Page 461]]

    (2) Standardization of Karl Fischer reagent. Add a known volume of 
the Karl Fischer reagent to a suitable titrating vessel which has been 
previously dried at 105 deg. C. and cooled in a desiccator. Introduce a 
mechanical stirrer and two platinum electrodes which are connected to a 
suitable electrometric apparatus for measurement of the endpoint. Start 
the stirrer and titrate with the water-methanol solution until the 
endpoint is reached. Calculate the milliliters of Karl Fischer reagent 
equivalent to each milliliter of water-methanol. Add an accurately 
weighed quantity of water (approximately 50 milligrams) to a dry 
titrating vessel, add an excess of the Karl Fischer reagent and back 
titrate with the water-methanol solution as above. Calculate the 
milligrams of water equivalent to each milliliter of the Karl Fischer 
reagent. Standardize the Karl Fischer reagent in this manner daily.
[GRAPHIC] [TIFF OMITTED] TR01JA93.103

where:
e=milligrams of water equivalent to 1 ml. Karl Fischer reagent.
w=weight of water in milligrams.
v1=volume of Karl Fischer reagent used.
v2=volume of methanol used.
f=volume ratio of Karl Fischer reagent to water-methanol solution.

    (3) Procedure. Transfer 1.0 milliliter of the penicillin in oil and 
wax to a dry titrating vessel, add 10 milliliters of dry chloroform and 
an excess of the Karl Fischer reagent and back titrate with the water-
methanol solution until the endpoint is reached. Transfer 10 milliliters 
of the dry chloroform used to a dry titrating vessel, add an excess of 
Karl Fischer reagent, and titrate with the water-methanol as above. 
Calculate the milliliters of Karl Fischer reagent equivalent to 10 
milliliters of chloroform.
[GRAPHIC] [TIFF OMITTED] TC01AP94.030

where:
b=milliliters Karl Fischer reagent equivalent to 10 ml. of chloroform.
s=volume of the sample in milliliters.

    (d) Measurement of penicillin particle size. Vigorously shake the 
container to obtain an even suspension of the penicillin particles and 
immediately withdraw therefrom approximately 0.5 milliliter of the drug 
into a clean, dry, tuberculin syringe using a dry 18-gauge needle. 
Discard approximately the first 5 drops of the mixture extruded from the 
needle and then extrude approximately 1 minim of the remaining mixture 
into a test tube containing 3 to 4 milliliters of light mineral oil. 
Thoroughly mix the contents of the tube and by means of a 
bacteriological loop (2 millimeters inside diameter, 22 gauge wire), 
immediately place one loopful of the suspension on each ruled chamber of 
a bright line hemocytometer. (It is not necessary to use a cover slip.) 
Confirm by means of the low power objective of the microscope the even 
distribution of particles over the ruled areas of both chambers and 
repeat with another loopful of the suspension if even dispersion is not 
obtained. Use a magnification of 430 or 440 diameters and a calibrated 
ocular micrometer to measure the penicillin particles. For the purpose 
of measurement and calculation, the predominant type of crystals 
observed shall be considered to represent the type of crystals present 
and the thickness and density of all particles shall be considered 
constant. Center a large penicillin particle in the microscopic field; 
measure the particle and all other particles in the field and repeat 
this operation on other fields until at least 200 particles are 
measured. Particles of less than 5 microns in length are disregarded. 
The grouping of the particles by length, the midpoint, the ratio of the 
midpoints, and the square of the ratio of the midpoints for each group 
are tabulated below:

                                                                        
------------------------------------------------------------------------
                                  Length             Ratio              
             Group                  in       Mid-   of mid-  (Ratio) \2\
                                  microns   point    points             
------------------------------------------------------------------------
1..............................      5-14      9.5     1.00       1.00  
2..............................     15-29     22.0     2.31       5.34  
3..............................     30-49     39.5     4.16      17.31  
4..............................     50-69     59.5     6.26      39.19  
5..............................     70-99     84.5     8.89      79.03  
6..............................   100-149    124.5    13.10     171.61  
7..............................   150-199    174.5    18.36     337.09  
8..............................   200-249    224.5    23.63     558.38  
9..............................   250-300    275.0    28.95     838.10  

[[Page 462]]

                                                                        
------------------------------------------------------------------------


Calculate the percent particles in each group from the total number 
measured. Determine the percent relative weight for each group as 
follows:

    Plate type particles. Relative weight= (ratio) 2 x % of 
total particles in group.
[GRAPHIC] [TIFF OMITTED] TR01JA93.104

    Rod-shaped particles. In the case of rod-shaped particles measure 
the width as well as the length.

Relative weight=ratio  x  average width  x  % of total particles in 
          group
          [GRAPHIC] [TIFF OMITTED] TC01AP94.031
          

When examined by the method described in this section not less than 50 
percent of the total relative weight of the penicillin in the drug 
consists of penicillin having a particle size of not less than 50 
microns in length.



Sec. 436.503  Procaine penicillin and buffered crystalline penicillin for aqueous injection.

    (a) Total potency (except in single-dose container), sterility, 
moisture, pyrogens, toxicity, pH. Proceed as directed in 
Sec. 440.274b(b) of this chapter.
    (b) Buffered crystalline penicillin content--(1) Preparation of the 
solution for assay. Add the indicated amount of distilled water to the 
contents of a vial of the sample, and shake well. Withdraw one dose of 
the suspension with a hypodermic syringe and place in a 10-milliliter 
volumetric flask. Add 20-percent sodium sulfate solution almost to the 
mark, centrifuge sufficiently to see the meniscus, make to volume with 
20-percent sodium sulfate solution, shake well, and centrifuge to obtain 
a clear or reasonably clear solution. Dilute a 5.0-milliliter aliquot of 
this clear solution with 1-percent phosphate buffer, pH 6.0, to give a 
solution for assay of approximately 2,000 units per milliliter.
    (2) Iodometric assay for total penicillin in the solution for assay. 
Determine the quantity of penicillin in the solution for assay by the 
iodometric assay procedure described in Sec. 440.80a(b)(5)(iv)(a) of 
this chapter.
    (3) Colorimetric determination of procaine penicillin in the 
solution for assay. Transfer an aliquot of the solution for assay to a 
50-milliliter volumetric flask. Determine the quantity of procaine 
penicillin in this solution by the following method:
    (i) Reagents--(a) Sodium nitrite solution. Dissolve 0.1 gram of 
sodium nitrite in 100 milliliters of distilled water. Prepare fresh 
solution every week and store under refrigeration.
    (b) Ammonium sulfamate solution. Dissolve 0.5 gram of ammonium 
sulfamate in 100 milliliters of distilled water and store under 
refrigeration.
    (c) N-(1-naphthyl)-ethylenediamine solution. Dissolve 0.1 gram of N-
(1-naphthyl) ethylenediamine dihydrochloride in 100 milliliters of 
distilled water. Prepare fresh solutions every week and store under 
refrigeration.
    (d) Standard procaine solution. Prepare a standard solution 
containing 27.55 milligrams of procaine hydrochloride U.S.P. in a liter 
of distilled water (each milliliter of the standard solution is 
equivalent to 60 units of procaine penicillin).
    (ii) Standards. Transfer, respectively, 1.0, 2.0, 3.0, 4.0, and 5.0 
milliliters of the standard solution and 5.0 milliliters of distilled 
water to each of six 50-milliliter volumetric flasks. Add 4.0, 3.0, 2.0, 
and 1.0 milliliters of water to the first four flasks, respectively, to 
give each a volume to 5.0 milliliters.
    (iii) Procedure. To each flask for the standards and the solution 
for assay add 0.5 milliliter of 4 N HCl, 1.0 milliliter of the sodium 
nitrite solution, 1.0 milliliter of the ammonium sulfamate, and 1.0 
milliliter of the N-(1-naphthyl)-ethylenediamine solution. Mix and wait 
two minutes after each addition. Make each flask to volume of 50 
milliliters with distilled water. Determine the absorbency of the 
colored solutions at 550 M in a suitable photo electric 
colorimeter. The instrument is balanced so that the zero concentration 
reads zero absorbency. Plot the standard curve on coordinate graph 
paper. Obtain the procaine penicillin content of the solution for assay 
directly from the point on the standard curve corresponding to its 
absorbency.

[[Page 463]]

    (4) The content of buffered crystalline pencillin in one dose of the 
product is calculated as follows:

                                A=(B-C)F,

where:
A=buffered crystalline penicillin content of the product.
B=total number of units of penicillin per milliliter as determined in 
paragraph (b)(2) of this section.
C=number of units of procaine penicillin per milliliter as determined in 
paragaph (b)(3) of this section.
F=appropriate dilution factor depending on the dilution made in the 
preparation of the solution for assay.


The content of buffered crystalline penicillin in the batch is 
satisfactory when determined by the method described in this paragraph 
if it is not less than 85 percent of that which it is represented to 
contain.
    (c) Procaine penicillin. The procaine penicillin content of the 
batch is the difference between the total potency determined by the 
method described in paragraph (a) or (d) of this section and the content 
of the buffered crystalline penicillin determined by the method 
described in paragraph (b) of this section. The procaine penicillin 
content of the batch is satisfactory when determined by the method 
described in this paragraph if it is not less than 85 percent of that 
which it is represented to contain.
    (d) Total potency of a one-dose container. Wash out the material 
remaining in the 10-milliliter volumetric flask referred to in paragraph 
(b)(1) of this section with 1-percent phosphate buffer, pH 6.0. Dilute 
to give a concentration of approximately 2,000 units per milliliter, and 
assay by the iodometric method described in Sec. 440.80a (b)(5)(iv)(a) 
of this chapter. Obtain the total potency by adding the number of units 
found in this solution (units per milliliter  x  volume) to the number 
of units found (units per milliliter  x  volume) in the solution assayed 
in accordance with paragraph (b)(2) of this section.



Sec. 436.504  Penicillin-bacitracin ointment.

    (a) Potency--(1) Penicillin content. Proceed as directed in 
Sec. 540.380a(b)(1) of this chapter, except the last sentence of that 
paragraph. Its content of penicillin is satisfactory if it contains not 
less than 85 percent of the number of units it is represented to 
contain.
    (2) Bacitracin content. Proceed as directed in Sec. 448.510a(b)(1) 
of this chapter, except that sufficient penicillinase is added to the 
sample under test to completely inactivate the penicillin present. Its 
content of bacitracin is satisfactory if it contains not less than 85 
percent of the number of units it is represented to contain.
    (b) Moisture. Proceed as directed in Sec. 436.201.
[39 FR 18944, May 30, 1974, as amended at 40 FR 13497, Mar. 27, 1975]



Sec. 436.505  Penicillin-streptomycin-bacitracin ointment; penicillin-dihydrostreptomycin-bacitracin ointment; penicillin-streptomycin-bacitracin methylene 

          disalicylate ointment; penicillin-dihydrostreptomycin-
          bacitracin methylene disalicylate ointment.

    (a) Potency--(1) Content of penicillin, streptomycin, and 
dihydrostreptomycin. Proceed as directed in Sec. 536.501(a) of this 
chapter.
    (2) Bacitracin content. Proceed as directed in Sec. 448.510a(b)(1) 
of this chapter, except that:
    (i) Sufficient penicillinase is added to the sample under test to 
completely inactivate the penicillin present.
    (ii) Use as the test organism the streptomycin dihydrostreptomycin 
resistant strain of either Micrococcus flavus (ATCC 10240A) 1 
or Sarcina subflava (ATCC 7468/d), 1 grown and maintained in 
media containing 500 micrograms of streptomycin or dihydrostreptomycin 
per milliliter of media, or calculate from the quantity of streptomycin 
or dihydrostreptomycin found, using the method prescribed by paragraph 
(a)(1) of this section, the quantity that would be present when the 
sample is diluted to contain one unit of bacitracin (labeled potency) 
per milliliter. Prepare the bacitracin standard curve by adding the 
calculated quantity of streptomycin or dihydrostreptomycin to each 
concentration of bacitracin used for

[[Page 464]]

the curve. Use this standard curve to calculate the bacitracin content 
of the sample.
---------------------------------------------------------------------------

    1 Available from: American Type Culture Collection, 12301 
Parklawn Drive, Rockville, MD 20852.
---------------------------------------------------------------------------

    (3) Bacitracin methylene disalicylate content. Proceed as directed 
in paragraph (a)(2) of this section, except prepare the sample as 
follows: Place a representative portion of the sample (usually 
approximately 1 gram, accurately weighed) or the entire contents of a 
single-dose container in blending jar, add 99 milliliters of a 2.0-
percent aqueous solution of sodium bicarbonate and 1 milliliter of a 10-
percent aqueous solution of polysorbate 80 and blend for 3 minutes in a 
high-speed blender. Allow the foam to subside, remove an aliquot of the 
solution, and dilute to 1 unit per milliliter with 1.0-percent phosphate 
buffer, pH 6.0.
    (b) Moisture. Proceed as directed in Sec. 436.201.
[39 FR 18944, May 30, 1974, as amended at 40 FR 13497, Mar. 27, 1975]



Sec. 436.506  Benzathine penicillin G and buffered crystalline penicillin for aqueous injection.

    (a) Total potency (except in single-dose containers). Proceed as 
directed in Sec. 440.80a(b)(1) of this chapter, except if the bioassay 
method is used prepare the sample by diluting 1.0 milliliter of the drug 
suspension with sufficient dimethyl formamide, formamide, or methyl 
alcohol to dissolve the benzathine penicillin. Make to 100 milliliters 
with buffer. Shake well and dilute to 1.0 unit per milliliter. If the 
iodometric method is used, proceed as directed in Sec. 440.55a(b) of 
this chapter, except in preparing the blank solution dilute 1.0 
milliliter of the drug suspension to 250 milliliters with 1-percent 
phosphate buffer at pH 6.0. In preparing the solution for inactivation 
dissolve 1.0 milliliter of the drug suspension in approximately 20 
milliliters of 0.5 N NaOH. Allow to stand for 15 minutes. Dilute to 250 
milliliters with distilled water. Pipette a 2.0-milliliter aliquot into 
a 125-milliliter glass-stoppered Erlenmeyer flask and add 2.0 
milliliters 1.2 N HCl and 10 milliliters 0.01 N iodine.
    (b) Buffered crystalline penicillin content. Place 1.0 milliliter of 
the drug suspension in a 10-milliliter volumetric flask and add 20 
percent sodium sulfate to make 10 milliliters. Shake well and centrifuge 
to obtain a clear, or reasonably clear, solution. Dilute a 5.0-
milliliter aliquot to 50 milliliters with buffer and proceed as directed 
in Sec. 440.80a(b)(1) of this chapter to determine the number of units 
per milliliter of this solution, and from this value calculate the 
number of units per milliliter of the drug. The content of buffered 
crystalline penicillin is satisfactory if it is not less than 85 percent 
of that which it is represented to contain.
    (c) Benzathine penicillin G content. The benzathine penicillin G 
content of the batch is the difference between the total potency as 
described in paragraph (a) or (d) of this section and the content of 
buffered crystalline penicillin determined by the method prescribed in 
paragraph (b) of this section. The content of benzathine penicillin G is 
satisfactory if it is not less than 85 percent of that which it is 
represented to contain.
    (d) Total potency of a single-dose container. Add sufficient 
distilled water to the material remaining in the 10-milliliter 
volumetric flask referred to in paragraph (b) of this section to bring 
the volume back to 10 milliliters and determine the number of units per 
milliliter of this suspension. If the iodometric method is used, 2.0-
milliliter aliquots are placed in 50-milliliter volumetric flasks (one 
blank and one to be inactivated). Obtain the total potency by adding the 
number of units found in the 10-milliliter volumetric flask to one-half 
the content of buffered crystalline penicillin found in paragraph (b) of 
this section.
    (e) Sterility. Proceed as directed in Sec. 436.20 using the method 
described in paragraph (e)(2) of that section, except use medium C in 
lieu of medium A, and medium F in lieu of medium E. During the period of 
incubation, shake the tubes at least once daily.
    (f) Moisture. Proceed as directed in Sec. 440.74a(b)(5) of this 
chapter.
    (g) Pyrogens. Proceed as directed in Sec. 436.500.
    (h) Toxicity. Proceed as directed in Sec. 440.55a(b)(3) of this 
chapter.
    (i) pH. Proceed as directed in Sec. 440.80a(b)(5)(ii) of this 
chapter, using the suspension resulting when the

[[Page 465]]

product is reconstituted as directed in the labeling.



Sec. 436.507  Benzathine - procaine - buffered crystalline penicillins for aqueous injection.

    (a) Potency--(1) Total potency. Proceed as directed in 
Sec. 440.80a(b)(1) of this chapter, except if the bioassay method is 
used prepare the sample by diluting one dose of the drug suspension with 
sufficient dimethyl formamide or formamide or methyl alcohol to dissolve 
the benzathine penicillin G. Make to 100 milliliters with 1-percent 
phosphate buffer, pH 6.0. Shake well, and dilute to 1.0 unit per 
milliliter with buffer. If the iodometric method of assay is used, add 
the indicated amount of distilled water to the contents of a vial of the 
sample, shake well, and proceed as follows (except for single-dose 
containers):
    (i) Using a standardized hypodermic syringe, withdraw one dose and 
dilute with 1-percent phosphate buffer, pH 6.0, to give a concentration 
of approximately 2,000 units per milliliter. Use 2.0 milliliters of this 
suspension as the blank in the iodometric assay procedure described in 
Sec. 440.80a(b)(5)(iv)(a) of this chapter.
    (ii) Using a standardized hypodermic syringe, withdraw another dose, 
place in a flask, and add 20 milliliters of 0.5 N NaOH for each 300,000 
units of benzathine penicillin, mix well, being sure that all penicillin 
is in solution, and allow to stand for 15 minutes. Add 1 milliliter of 
1.2 N HCl for each 2 milliliters of 0.5 N NaOH, mix, and dilute with 
distilled water to the same volume as was used in paragraph (a)(1)(i) of 
this section. Place 2.0 milliliters in a 125-milliliter glass-stoppered 
Erlenmeyer flask, add 10 milliliters of 0.01 N iodine, allow to stand 
for 15 minutes, and titrate with 0.01 N sodium thiosulfate as directed 
in the iodometric assay procedure in Sec. 440.80a (b)(5)(iv)(a) of this 
chapter. The total potency of the batch is satisfactory if it contains 
not less than 85 percent of that which it is represented to contain.
    (2) Procaine penicillin content (except for single-dose containers). 
Make suitable dilutions of the solution prepared in paragraph (a)(1)(ii) 
of this section to obtain approximately 60 units of procaine penicillin 
per milliliter. Determine the procaine penicillin content by the 
colorimetric procedure described in Sec. 436.503(b)(3). The content of 
procaine penicillin is satisfactory if it contains not less than 85 
percent of the number of units that it is represented to contain.
    (3) Buffered crystalline penicillin content--(i) Preparation of the 
solution for assay. (a) Add the indicated amount of distilled water to 
the contents of a vial of the sample, and shake well. Withdraw one dose 
of the suspension with a hypodermic syringe and place in a 10-milliliter 
volumetric flask. Add 20-percent sodium sulfate solution almost to the 
mark, centrifuge sufficiently to see the meniscus, make to volume with 
20-percent sodium sulfate solution, shake well, and centrifuge to obtain 
a clear or reasonably clear solution; or
    (b) If the original product contains more than 600,000 units, place 
it in a 50-milliliter volumetric flask, add 20-percent sodium sulfate to 
the mark, shake well, place a 10-milliliter portion in a centrifuge 
tube, and centrifuge to obtain a reasonably clear solution.
    (c) Dilute a 5.0-milliliter aliquot of the clear solution obtained 
in paragraph (a) (3)(i) (a) or (b) of this section with 1-percent 
phosphate buffer, pH 6.0, to give a solution for assay of approximately 
2,000 units per milliliter.
    (ii) Iodometric assay for total penicillin in the solution for 
assay. Determine the total quantity of penicillin in the solution for 
assay by the iodometric assay procedure described in Sec. 440.80a 
(b)(5)(iv)(a) of this chapter.
    (iii) Colorimetric determination of procaine penicillin in the 
solution for assay. Proceed as directed in Sec. 436.503 (b)(3). The 
content of procaine penicillin in the batch is satisfactory if it is not 
less than 85 percent of that which it is represented to contain.
    (iv) The buffered crystalline penicillin in one dose of the product 
is calculated as follows:

                                A=(B-C)F,

where:
A=the buffered crystalline penicillin content of the product.
B=the number of units of penicillin per milliliter as determined in 
paragraph (a)(3)(ii) of this section.

[[Page 466]]

C=the number of units of procaine penicillin per milliliter as 
determined in paragraph (a)(3)(iii) of this section.
F=the appropriate dilution factor depending on the dilutions made in the 
preparation of the solution for assay.


The content of buffered crystalline penicillin is satisfactory if the 
batch contains 85 percent of the number of units per milliliter that it 
is represented to contain.
    (4) Benzathine penicillin content. The sum of the procaine 
penicillin content determined under paragraph (a)(2) or (6) of this 
section and the buffered crystalline penicillin content determined under 
paragraph (a)(3) of this section, subtracted from the total potency 
determined in paragraph (a)(1) or (5) of this section, represents the 
benzathine penicillin G content. The benzathine penicillin G content is 
satisfactory if it is not less than 85 percent of the number of units 
that it is represented to contain.
    (5) Total potency of a single-dose container. Wash out the material 
remaining in the volumetric flask referred to in paragraph (a)(3)(i)(a) 
of this section, or combine the contents remaining in the 50-milliliter 
volumetric flask and in the centrifuge tube referred to in paragraph 
(a)(3)(i)(b) of this section. Dissolve the material by adding 10 
milliliters of 1 N NaOH for each 300,000 units of benzathine penicillin 
and allow to stand 15 minutes. Add 1 milliliter of 1.2 N HCl for each 
milliliter of 1 N NaOH and then dilute with distilled water to give a 
concentration of approximately 2,000 units per milliliter. Place 2.0 
milliliters in a 125-milliliter glass-stoppered Erlenmeyer flask, add 10 
milliliters of 0.01 N iodine, allow to stand for 15 minutes, and then 
titrate with 0.01 N sodium thiosulfate as directed in Sec. 440.80a 
(b)(5)(iv)(a) of this chapter. For the blank determination prepare a 
separate sample as directed in paragraph (a)(3)(i) (a) or (b) of this 
section and in the first sentence of this paragraph (a)(5), then dilute 
with 1 percent phosphate buffer, pH 6.0, to give a concentration of 
approximately 2,000 units per milliliter. The total potency of the one-
dose container is equal to the sum of the number of units found in this 
assay (units per milliliter x volume) and the number of units found 
(units per milliliter  x volume) in the solution for assay in paragraph 
(a)(3)(ii) of this section.
    (6) Procaine penicillin content of a single-dose container. Make 
suitable dilutions of the NaOH-inactivated solution prepared in 
paragraph (a)(5) of this section to obtain approximately 60 units of 
procaine penicillin per milliliter. Determine the procaine penicillin 
content (units per milliliter x  volume) of this solution by the 
colorimetric procedure described under Sec. 436.503(b)(3). To this value 
add per procaine penicillin content (unit per milliliter x volume) of 
the solution for assay, as found in paragraph (a)(3)(iii) of this 
section, to obtain the procaine penicillin content of the one-dose 
container. The content of procaine penicillin in the batch is 
satisfactory if it is not less than 85 percent of that which it is 
represented to contain.
    (b) Sterility. Proceed as directed in Sec. 436.20, using the method 
described in paragraph (e)(2) of that section, except use medium C in 
lieu of medium A, and medium F in lieu of medium E. During the period of 
incubation, shake the tubes at least once daily.
    (c) Pyrogens. Proceed as directed in Sec. 440.55a(b)(4) of this 
chapter.
    (d) Toxicity. Proceed as directed in Sec. 440.55a(b)(3) of this 
chapter.
    (e) Moisture. Proceed as directed in Sec. 440.74a(b)(5) of this 
chapter.
    (f) pH. Proceed as directed in Sec. 440.80a(b)(5)(ii) of this 
chapter, using the suspension resulting when the product is 
reconstituted as directed in the labeling.



Sec. 436.508  Penicillin - bacitracin - neomycin ointment; penicillin-bacitracin-neomycin in oil.

    (a) Potency--(1) Penicillin content; bacitracin content. Proceed as 
directed in Sec. 436.504(a).
    (2) Neomycin content. Proceed as directed in Sec. 448.510d(b)(1)(ii) 
of this chapter, except that sufficient penicillinase is added to the 
sample under test to completely inactivate the penicillin present. Its 
content of neomycin is satisfactory if it contains not less than 85 
percent of the number of milligrams per gram that it is represented to 
contain.

[[Page 467]]

    (b) Moisture. Proceed as directed in Sec. 436.201.



Sec. 436.509  Procaine penicillin-streptomycin-polymyxin in oil; procaine penicillin-dihydrostreptomycin-polymyxin in oil; procaine penicillin-streptomycin-

          polymyxin ointment; procaine penicillin - dihydrostreptomycin 
          - polymyxin ointment.

    (a) Potency--(1) Penicillin content. Proceed as directed in 
Sec. 540.380a(b)(1) of this chapter. Its content of penicillin is 
satisfactory if it contains not less than 85 percent of the number of 
units per milliliter or per gram that it is represented to contain.
    (2) Streptomycin content. Proceed as directed in 
Sec. 544.373(b)(1)(i) of this chapter, except inactivate the penicillin 
in the combined extractives with sufficient penicillinase at 37 deg. C. 
for 30 minutes. Its content of streptomycin is satisfactory if it 
contains not less than 85 percent of the number of milligrams per 
milliliter or per gram that it is represented to contain.
    (3) Dihydrostreptomycin content. Proceed as directed in paragraph 
(a)(2) of this section, using the dihydrostreptomycin working standard 
as a standard of comparison. Its content of dihydrostreptomycin is 
satisfactory if it contains not less than 85 percent of the number of 
milligrams per milliliter or per gram that it is represented to contain.
    (4) Polymyxin content. Proceed as directed in Sec. 444.170a(b)(2)(i) 
of this chapter, with the following exceptions:
    (i) In lieu of the directions for the preparation of the sample 
described in Sec. 444.170a(b)(2)(i)(g) of this chapter, prepare the 
sample by one of the following techniques:
    (a) Extraction. Place a convenient-sized representative quantity of 
the sample in a separatory funnel containing approximately 50 
milliliters of peroxide-free ether. Shake the sample and ether until 
homogeneous. Add 25 milliliters of 10-percent potassium phosphate buffer 
(pH 6.0) and shake. Remove the buffer layer and repeat the extraction 
with 25-milliliter portions of buffer at least three times and any 
additional times that may be necessary to insure complete extraction of 
the antibiotic. Combine the extractives. Inactivate the penicillin with 
sufficient penicillinase at 37 deg. C. for 30 minutes. Make the proper 
estimated dilutions in 10-percent potassium phosphate buffer (pH 6.0) to 
give a concentration of 10 units per milliliter (estimated).
    (b) Blending. Place a convenient-sized representative quantity of 
the sample in a blending jar containing 1.0 milliliter of polysorbate 80 
and sufficient 1-percent phosphate buffer (pH 6.0) to give a final 
volume of 200 milliliters. If the sample consists of substantially more 
than 1 gram, use sufficient buffer to give a final volume of 500 
milliliters. If the concentration of polymyxin in the blend is less than 
200 units per milliliter, 10-percent phosphate buffer (pH 6.0) should be 
used in lieu of 1-percent phosphate buffer (pH 6.0). Using a high-speed 
blender, blend the mixture for 2 minutes. Inactivate the penicillin with 
sufficient penicillinase at 37 deg. C. for 30 minutes and make the 
proper estimated dilutions in 10-percent phosphate buffer (pH 6.0) to 
give a concentration of 10 units per milliliter (estimated).
    (ii) The standard curve is prepared in the following concentrations: 
6.4, 8.0, 10.0, 12.5, and 15.6 units per milliliter in 10-percent 
potassium phosphate buffer, pH 6.0. The 10 units per milliliter 
concentration is used as the reference point. Its content of polymyxin 
is satisfactory if it contains not less than 85 percent of the number of 
units per milliliter or per gram that it is represented to contain.
    (b) Moisture. Proceed as directed in Sec. 436.201.
[39 FR 18944, May 30, 1974, as amended at 40 FR 13497, Mar. 27, 1975; 41 
FR 10886, Mar. 15, 1976]



Sec. 436.510  Penicillin-streptomycin-erythromycin ointment; penicillin-dihydro-streptomycin-erythromycin ointment.

    (a) Potency--(1) Penicillin content. Obtain the weight of the 
content of a syringe by weighing before and after ejecting the content 
into a beaker. Stir until homogeneous. Remove a representative sample 
(usually approximately 1.0 gram, accurately weighed) and place in a 
separatory funnel containing 50 milliliters of peroxide-free

[[Page 468]]

ether. Add 20 milliliters of 0.1 M potassium phosphate buffer (pH 8.0) 
and shake. Remove the buffer layer and repeat the extraction with three 
additional 20-milliliter portions of the buffer. Place the buffer 
solution in a second separatory funnel and wash with three 30-milliliter 
portions of ether. Discard the ether washes. Remove an aliquot of the 
buffer solution and proceed as directed in Sec. 440.80a(b) (1) of this 
chapter, except Sec. 440.80a(b)(1)(iv) and (ix) of this chapter. If the 
iodometric chemical assay is used, proceed as directed in 
Sec. 440.80a(b)(5)(iv)(a) of this chapter, except prepare the sample as 
directed in Sec. 536.501(a)(1) of this chapter. Its content of 
penicillin is satisfactory if it contains not less than 85 percent of 
the number of units that it is represented to contain.
    (2) Streptomycin content. Using an aliquot of the buffer solution 
prepared as directed in paragraph (a)(1) of this section, proceed as 
directed in Sec. 444.70a (b)(1) through (9) of this chapter, except add 
sufficient penicillinase to completely inactivate the penicillin 
present. Its content of streptomycin is satisfactory if it contains not 
less than 85 percent of the number of milligrams that it is represented 
to contain.
    (3) Dihydrostreptomycin content. Proceed as directed in paragraph 
(a)(2) of this section, using the dihydrostreptomycin working standard 
as the standard of comparison. Its content of dihydrostreptomycin is 
satisfactory if it contains not less than 85 percent of the number of 
milligrams that it is represented to contain.
    (4) Erythromycin content. Proceed as directed in 
Sec. 444.570b(b)(1)(i)(b) of this chapter, except prepare the sample as 
follows: Place a representative sample (usually approximately 1.0 gram, 
accurately weighed), in a glass blending jar containing 99 milliliters 
of 0.1 M potassium phosphate buffer, pH 8.0, and 1 milliliter of 
polysorbate 80. Using a high-speed blender, blend for 2 to 3 minutes. 
Add 100 milliliters of 0.1 M potassium phosphate buffer, pH 8.0, and 
blend for an additional 2 to 3 minutes. Prepare an intermediate dilution 
by diluting an aliquot of the filtrate with 0.1 M potassium phosphate 
buffer (pH 8.0), and add sufficient penicillinase to inactivate the 
penicillin. Then further dilute with buffer to give an erythromycin 
content of 1.0 microgram per milliliter (estimated). Its content of 
erythromycin is satisfactory if it contains not less than 85 percent of 
the number of milligrams that it is presented to contain.
    (b) Moisture. Proceed as directed in Sec. 436.500(c).
[39 FR 18944, May 30, 1974, as amended at 40 FR 13497, Mar. 27, 1975]



Sec. 436.511  Penicillin-streptomycin-bacitracin methylene disalicylate-neomycin ointment; penicillin-dihydrostreptomycin-bacitracin methylene disalicylate-
          neomycin ointment.

    (a) Potency--(1) Penicillin content. Proceed as directed in 
Sec. 540.380a(b)(1) of this chapter. Its penicillin content is 
satisfactory if it contains not less than 85 percent of the number of 
units that it is represented to contain.
    (2) Streptomycin content. Proceed as directed in Sec. 436.105 of 
this chapter. Its content of streptomycin is satisfactory if it contains 
not less than 85 percent of the number of milligrams that it is 
represented to contain.
    (3) Dihydrostreptomycin content. Proceed as directed in Sec. 436.105 
of this chapter. Its content of dihydrostreptomycin is satisfactory if 
it contains not less than 85 percent of the number of milligrams that it 
is represented to contain.
    (4) Bacitracin methylene disalicylate content. Proceed as directed 
in Sec. 436.505(a)(3). Its potency is satisfactory if it contains not 
less than 85 percent of the equivalent number of units of bacitracin 
that it is represented to contain.
    (5) Neomycin content. Proceed as directed in Sec. 436.105 of this 
chapter. Its content of neomycin is satisfactory if it contains not less 
than 85 percent of the number of milligrams that it is represented to 
contain.
    (b) Moisture. Proceed as directed in Sec. 436.201.
[39 FR 18944, May 30, 1974, as amended at 40 FR 13497, Mar. 27, 1975]



Sec. 436.512  Procaine penicillin G-novobiocin-neomycin-dihydrostreptomycin in oil.

    (a) Potency--(1) Penicillin G content. Proceed as directed in 
Sec. 440.180d (b)(1)(i)(a) of this chapter, using the

[[Page 469]]

novobiocin-resistant strain of Staphylococcus aureus (ATCC 12692),1 
except prepare the sample as follows: Place the equivalent of one dose 
of sample in a blending jar, add 1.0 milliliter of polysorbate 80 and a 
quantity of 1 percent potassium phosphate buffer, pH 6.0, sufficient to 
make a total of 500 milliliters. Blend for 5 minutes with a high-speed 
blender and make appropriate dilutions, using 1 percent potassium 
phosphate buffer, pH 6.0. Its content of penicillin G is satisfactory if 
it contains not less than 85 percent of the number of units that it is 
represented to contain.
---------------------------------------------------------------------------

    1 Available from: American Type Culture Collection, 12301 
Parklawn Drive, Rockville, MD 20852.
---------------------------------------------------------------------------

    (2) Novobiocin content. Proceed as directed in 
Sec. 440.180d(b)(3)(i), with the following exceptions:
    (i) Prepare the sample as follows: Place the equivalent of one dose 
of sample in a blending jar, add 1.0 milliliter of polysorbate 80 and a 
quantity of 0.1M potassium phosphate buffer, pH 8.0, sufficient to make 
a total of 500 milliliters. Blend for 5 minutes with a high-speed 
blender. To an aliquot, add sufficient penicillinase to inactivate the 
penicillin, further dilute with 10 percent potassium phosphate buffer, 
pH 6.0 (solution 6) to give a final concentration of 0.5 microgram 
novobiocin per milliliter (estimated), and allow to stand for \1/2\-hour 
at 37 deg. C. before filling the plates.
    (ii) Aseptically add to the seed agar used for this assay, at the 
time the bacterial suspension is added, a slurry of Dowex 50 WX-4, 
Na+ type 200-400 mesh, sufficient to make a total 
concentration of 2 percent. Prepare the slurry by adding 50 grams of the 
resin to 30 milliliters of distilled water and sterilize for 15 minutes 
at 15 pounds pressure. Mix the slurry thoroughly before adding. Its 
content of novobiocin is satisfactory if it contains not less than 85 
percent of the number of milligrams that it is represented to contain.
    (3) Neomycin content. Proceed as directed in Sec. 436.517(b)(1) of 
this chapter, using the Staphylococcus epidermidis (ATCC 12228) 1 
procedure, except:
    (i) Prepare the sample as follows: Place the equivalent of one dose 
of sample in a blending jar, add 1.0 milliliter of polysorbate 80 and a 
quantity of 0.1M potassium phosphate buffer, pH 8.0, sufficient to make 
a total of 500 milliliters. Blend for 5 minutes with a high-speed 
blender. To an aliquot, add sufficient penicillinase to inactivate the 
penicillin, further dilute with 0.1M potassium phosphate buffer, pH 8.0, 
to give a final concentration of 1.0 microgram neomycin per milliliter 
(estimated), and allow to stand for \1/2\-hour at 37 deg. C. before 
filling the plates.
    (ii) Aseptically add to the seed agar used for this assay, at the 
time the bacterial suspension is added, a slurry of Dowex 1-X8, Cl type 
200-400 mesh, to make a total concentration of 1 percent. Prepare the 
slurry by adding 50 grams of the resin to 30 milliliters of distilled 
water and sterilize for 15 minutes at 15 pounds pressure. Mix the slurry 
thoroughly before adding. Its content of neomycin is satisfactory if it 
contains not less than 85 percent of the number of milligrams that it is 
represented to contain.
    (4) Dihydrostreptomycin content. Proceed as directed in Sec. 436.105 
except prepare the sample by placing the equivalent of one dose in a 
blender, add 1.0 milliliter of polysorbate 80 and a quantity of 0.1M 
potassium phosphate buffer, pH 8.0, sufficient to make a total of 500 
milliliters. Blend for 5 minutes with a high-speed blender. To an 
aliquot, add sufficient penicillinase to inactivate the penicillin, 
further dilute with 0.1M potassium phosphate buffer, pH 8.0, to give a 
final concentration of 1.0 microgram dihydrostreptomycin per milliliter 
(estimated), and allow to stand for \1/2\-hour at 37 deg. C. before 
filling the plates. Its content of dihydrostreptomycin is satisfactory 
if it contains not less than 85 percent of the number of milligrams that 
it is represented to contain.
    (b) Moisture. Proceed as directed in Sec. 436.500(c).
[39 FR 18944, May 30, 1974, as amended at 41 FR 10886, Mar. 15, 1976]



Sec. 436.513  Chlortetracycline troches; tetracycline hydrochloride troches.

    (a) Potency. If it is tetracycline hydrochloride proceed as directed 
in Sec. 446.81a(b)(1) of this chapter and if it is

[[Page 470]]

chlortetracycline hydrochloride troches proceed as directed in 
Sec. 446.10a(b)(1) of this chapter, except Sec. 446.10a(b)(1)(x), and in 
lieu of the directions in Sec. 446.10a(b)(1)(iv) and (viii)(c) of this 
chapter prepare the sample as follows: Place 12 troches in a glass 
blending jar containing 500 milliliters of 0.01N HCl. Using a high-speed 
blender, blend for 3 to 5 minutes and then make the proper estimated 
dilutions in the buffer solution. The average potency of the troches is 
satisfactory if they contain not less than 85 percent of the number of 
milligrams they are represented to contain.
    (b) Moisture. Proceed as directed in Sec. 440.80a(b)(5)(i) of this 
chapter.



Sec. 436.514  Chlortetracycline hydrochloride powder topical; tetracycline hydrochloride powder topical.

    (a) Potency--(1) Dry powder. Using a 3.0-gram sample or the entire 
contents of the immediate container for each determination, prepare the 
sample as follows: Using a high-speed blender, blend a 3.0-gram sample 
in a glass blending jar containing 500 milliliters of 0.01 N HCl (use 
0.1 N HCl if it is tetracycline), or reconstitute in the immediate 
container as directed in the labeling of the drug. Transfer an 
appropriate aliquot of 1.0 milliliter to 5.0 milliliters to a 100-
milliliter volumetric flask and make to mark with 0.01 N HCl (use 0.1 N 
HCl if it is tetracycline). Withdraw an aliquot from the volumetric 
flask, and if it is chlortetracycline hydrochloride dilute to 0.06 
 g. per milliliter, using 0.1 M potassium phosphate buffer, pH 
4.5, and proceed as directed in Sec. 446.10a(b)(1) of this chapter. If 
it is tetracycline hydrochloride, dilute to 0.24  g. per 
milliliter, using 0.1 M potassium phosphate buffer, pH 4.5, and proceed 
as directed in Sec. 446.81a(b)(1) of this chapter. The average potency 
is satisfactory if it contains not less than 85 percent of the number of 
milligrams of chlortetracycline hydrochloride or tetracycline 
hydrocloride per gram or per immediate container that it is represented 
to contain.
    (2) Powder packaged with inert gases. Spray, as directed in the 
labeling, the entire contents of each container to be tested into a 
separate 2-liter Erlenmeyer flask, held in a horizontal position. Add 
500 milliliters of 0.1 N HCl and shake to dissolve the contents. 
Immediately remove aliquots of this solution and, using 0.1 M potassium 
phosphate buffer, pH 4.5, for further dilutions, proceed as directed in 
Sec. 446.10a(b)(1) of this chapter if it is chlortetracycline 
hydrochloride powder or Sec. 446.81a(b)(1) of this chapter if it is 
tetracycline hydrochloride powder. Calculate the average total amount of 
antibiotic expelled from the containers. The total potency is 
satisfactory if it contains not less than 85 percent of the number of 
milligrams of chlortetracycline hydrochloride or tetracycline 
hydrochloride that it is represented to contain.
    (b) Moisture. Proceed as directed in Sec. 440.80a(b)(5)(i) of this 
chapter, except if it is packaged with inert gases proceed as directed 
in Sec. 536.513(c) of this chapter.
[39 FR 18944, May 30, 1974, as amended at 40 FR 13497, Mar. 27, 1975]



Sec. 436.515  Capsules tetracycline and oleandomycin phosphate; capsules tetracycline and troleandomycin; capsules tetracycline hydrochloride and oleandomycin 
          phosphate; capsules tetracycline hydrochloride and 
          troleandomycin.

    (a) Potency--(1) Tetracycline or tetracycline hydrochloride content 
by turbidimetric assay--(i) Test culture and media. Maintain the test 
organism Escherichia coli (ATCC 10536) 1 on the agar 
described in Sec. 440.80a(b)(1) (ii)(a) of this chapter. For use in the 
assay, prepare a suspension of the organism every 2 weeks, as follows: 
Transfer the organism to a fresh agar slant and incubate at 37 deg.C. 
overnight. Wash the growth from the slant with the aid of 2 milliliters 
of sterile distilled water and sterile glass beads into a Roux bottle 
containing 300 milliliters of the maintenance medium. Incubate overnight 
at 37 deg. C. and then harvest the growth with 50 milliliters of sterile 
distilled water and sterile glass beads. Standardize this suspension by 
determining the dilution that will permit 40-percent light

[[Page 471]]

transmission in a photoelectric colorimeter using a 650-millimicron 
filter and an 18-millimeter diameter test tube as an absorption cell. 
Prepare the daily inoculum by adding 10 milliliters of that dilution to 
each liter of nutrient broth, prepared as directed in Sec. 440.80a 
(b)(1)(ii)(c) of this chapter, needed for the test.
---------------------------------------------------------------------------

    1 Available from: American Type Culture Collection, 12301 
Parklawn Drive, Rockville, MD 20852.
---------------------------------------------------------------------------

    (ii) Working standard and solutions. Dissolve an appropriate amount 
of the working standard in sufficient 0.1 N HCl to give a concentration 
of 1,000 micrograms per milliliter. This stock solution may be kept in 
the refrigerator for 1 week. Make daily dilutions of the stock solution 
with 0.1 M potassium phosphate buffer (pH 4.5) to obtain concentrations 
of 0.146, 0.187, 0.240, 0.308, and 0.395 micrograms per milliliter. Add 
1.0 milliliter of each such concentration to each of three 16 
millimeters x 125 millimeters test tubes.
    (iii) Preparation of sample. Dissolve the contents of a 
representative number of capsules in sufficient 0.1 N HCl to give a 
stock solution of convenient concentration. Further dilute the stock 
solution with 0.1 M potassium phosphate buffer (pH 4.5) to obtain a 
final concentration of 0.24 microgram per milliliter (estimated). Add 
1.0-milliliter of this dilution to each of three 16 millimeters x 125 
millimeters test tubes.
    (iv) Procedure. To each of the 16 millimeters x 125 millimeters test 
tubes prepared in paragraph (a)(1)(ii) and (iii) of this section, add 
9.0 milliliters of the inoculated nutrient broth described in paragraph 
(a)(1)(i) of this section and place immediately in a 37 deg. C. water 
bath for 3 to 4 hours. After incubation, add 0.5 milliliter of a 12-
percent formaldehyde solution to each tube and read the absorbance 
values in a suitable photoelectric colorimeter using a wavelength of 530 
millimicrons. Set the instrument at zero absorbance with clear 
uninoculated broth prepared as described in Sec. 440.80a(b)(1)(ii)(c) of 
this chapter.
    (v) Estimation of potency. Plot the average values for each 
concentration of the standard on arithmetic graph paper with absorbance 
values on the ordinate and tetracycline or tetracycline hydrochloride 
concentrations on the abscissa. Construct the best straightline through 
the points, either by inspection or by means of the following equations:

                            L=(3a+2b+c-e)/5,

                            H=(3e+2d+c-a)/5,

where:
L=absorbance value for the lowest concentration of the standard curve.
H=absorbance value for the highest concentration of the standard curve.
a, b, c, d, e=average absorbance values for each concentration of the 
standard curve.


Plot the values obtained for L and H and connect the points with a 
straight line. Average the absorbance values for the sample and read the 
tetracycline or tetracycline hydrochloride concentration from the 
standard curve. Multiply the concentration by appropriate dilution 
factors to obtain the tetracycline or tetracycline hydrochloride content 
of the sample. Its potency is satisfactory if it contains the equivalent 
of not less than 85 percent of the number of milligrams of tetracycline 
hydrochloride that it is represented to contain.
    (2) Oleandomycin content. (i) If oleandomycin phosphate is used, 
proceed as directed in paragraph (c)(1) of this section, except prepare 
the sample as follows: Dissolve the contents of a representative number 
of capsules in sufficient 0.1 M potassium phosphate buffer (pH 8.0) to 
give a stock solution of convenient concentration. Further dilute with 
0.1 M potassium phosphate buffer (pH 8.0) to obtain a final 
concentration of 5.0  g. of oleandomycin activity per 
milliliter (estimated).
    (ii) If troleandomycin is used, proceed as follows: Dissolve the 
contents of a representative number of capsules in chloroform to give a 
stock solution of 1.0 milligram of oleandomycin activity per milliliter. 
Transfer 30 milliliters of the chloroform solution to a glass-stoppered 
test tube (200 millimeters x 22 millimeters) and add 20 milliliters of 1 
N sodium hydroxide. Shake for 1 minute and centrifuge briefly to aid in 
the separation of the layers. With the aid of a syringe and needle, 
remove and discard the aqueous layer. Repeat the washing procedure with 
two more 20-milliter portions of 1 N sodium hydroxide solution. Filter 
the chloroform layer through a pledget of cotton.

[[Page 472]]

Dilute an aliquot of this solution with chloroform to give a solution 
containing approximately 25  g. of oleandomycin per milliliter. 
Transfer a 5.0 milliliter aliquot to a 40 milliliter glass-stoppered 
centrifuge tube, dilute to 20 milliliters, with chloroform, and 
determine the oleandomycin content as directed in paragraph (d)(1)(i) of 
this section.

Its content of oleandomycin is satisfactory if it contains not less than 
85 percent of the number of milligrams that it is represented to 
contain.
    (b) Moisture. Proceed as directed in Sec. 440.80a(b)(5)(1) of this 
chapter.
    (c) Oleandomycin phosphate used in making the capsules--(1) 
Potency--(i) Cylinders (cups). Used cylinders described in 
Sec. 440.80a(b)(1)(i) of this chapter.
    (ii) Culture media. (a) Use the nutrient agar described in 
Sec. 440.80a (b)(1)(ii)(a) of this chapter for the seed layer and base 
layer, except that its pH after sterilization is 7.8 to 8.0.
    (b) Use the nutrient agar described in Sec. 440.80a(b)(1)(ii)(a) of 
this chapter for maintaining the test organism.
    (iii) Working standard. Dissolve a suitable weighed quantity 
(usually 25 milligrams or less) of the working standard (obtained from 
the Food and Drug Administration) in 2 milliliters of ethanol, then add 
sufficient 0.1 M potassium phosphate buffer, pH 8.0, to give a 
concentration of 1,000 micrograms of oleandomycin base per milliliter. 
This stock solution may be kept in the refrigerator for 3 days.
    (iv) Preparation of sample. Dissolve the sample in sufficient 0.1 M 
potassium phosphate buffer (pH 8.0) to give a convenient stock solution. 
Further dilute in 0.1 M potassium phosphate buffer (pH 8.0) to give a 
final concentration of 5.0 micrograms per milliliter (estimated).
    (v) Preparation of test organism. The test organism is 
Staphylococcus epidermidis (ATCC 12228) 1 which is maintained 
on slants or agar described under paragraph (c)(1)(ii)(a) of this 
section. Wash the organism from the agar slant with 3 milliliters of 
sterile physiological saline solution onto a large agar surface such as 
that provided by a Roux bottle containing 300 milliliters of the agar 
described in paragraph (c)(1)(ii)(a) of this section. Spread the 
suspension of organisms over the entire agar surface with the aid of 
sterile glass beads. Incubate for 4 hours at 32 deg. C. and then wash 
the resulting growth from the agar surface with about 30 milliliters of 
sterile physiological saline solution. Standardize the suspension by 
determining the dilution that will give 80-percent light transmission, 
using a suitable photoelectric colorimeter with a 650-millimicron filter 
and an 18-millimeter-diameter test tube as an absorption cell. Run test 
plates to determine the quantity of the diluted suspension (usually 1.5 
milliliters) that should be added to each 100 milliliters of agar to 
give clear, sharp zones of inhibition of appropriate size.
---------------------------------------------------------------------------

    1 Available from: American Type Culture Collection, 12301 
Parklawn Drive, Rockville, MD 20852.
---------------------------------------------------------------------------

    (vi) Preparation of plates. Add 21 milliliters of the agar prepared 
as described in paragraph (c)(1)(ii)(a) of this section to each Petri 
dish (20 millimeters  x  100 millimeters). Distribute the agar evenly in 
the plates and allow it to harden. Use the plates the same day they are 
prepared. Melt a sufficient amount of the agar described in paragraph 
(c)(1)(ii)(a) of this section, cool to 48 deg. C., add the proper amount 
of the test organism as described in paragraph (c)(1)(v) of this section 
and mix thoroughly. Add 4 milliliters of this inoculated agar to each 
Petri dish. Distribute the agar evenly in the plates, cover with 
porcelain covers glazed on the outside, and allow to harden. After the 
agar has hardened, place 6 cylinders on the agar surface so that they 
are at approximately 60 deg. intervals on a 2.8-centimeter radius.
    (vii) Standard curve. Prepare the daily standard curve by further 
diluting the 1,000 micrograms per milliliter stock solution in 0.1 M 
potassium phosphate buffer (pH 8.0) to obtain concentrations of 3.2, 
4.0, 5.0, 6.25 and 7.80 micrograms per milliliter. Use 3 plates for the 
determination of each point on the curve, except the 5.0 micrograms per 
milliliter concentration, a total of 12 plates. On each of 3 plates fill 
3 cylinders with the 5.0 micrograms per milliliter standard, and the 
other 3 cylinders with the concentration under test. Thus, there

[[Page 473]]

will be 36 five-microgram determinations and 9 determinations for each 
of the other points on the curve. After incubation, read the diameters 
of the circles of inhibition in the plates. Average the readings of the 
5.0 micrograms per milliliter concentration and the readings of the 
point tested for each set of 3 plates and average also all 36 readings 
of the 5.0 micrograms per milliliter concentration. The average of the 
36 readings of the 5.0 micrograms per milliliter concentration is the 
correction point for the curve. Correct the average value obtained for 
each point to the figure it would be if the 5.0 micrograms per 
milliliter reading for that set of 3 plates were the same as the 
correction point. Thus, if in correcting the 4.0-microgram 
concentration, the average of the 36 readings of the 5.0-microgram 
concentration were 20.0 millimeters, and the average of the 5.0-
microgram concentration of this set of 3 plates were 19.8 millimeters, 
the correction would be +0.2 millimeter. If the average reading of the 
4.0-microgram concentration of these same 3 plates were 19.0 
millimeters, the corrected value would be 19.2 millimeters. Plot these 
corrected values, including the average of the 5.0 micrograms per 
milliliter concentration, on 2-cycle semilog paper, using the 
concentration in micrograms per milliliter as the ordinate (the 
logarithmic scale) and the diameter of the zone of inhibition as the 
abscissa. Draw the standard curve through these points, either by 
inspection or by means of the following equations:

                            L=(3a+2b+c-e)/5,

                            H=(3e+2d+c-a)/5,

where:
L=corrected zone diameter for the lowest concentration of the standard 
curve,
H=corrected zone diameter for the highest concentration of the standard 
curve,
c=average zone diameter for 36 readings of the 5.0 micrograms per 
milliliter standard.
a, b, d, e=corrected average values for the 3.2, 4.0, 6.25, and 7.81 
micrograms per milliliter standard solutions, respectively.


Plot the values obtained for L and H and connect with a straight line.
    (viii) Assay. Use 3 plates for each sample. Fill 3 cylinders on each 
plate with the standard 5.0 micrograms per milliliter solution and 3 
cylinders with the 5.0 micrograms per milliliter (estimated) sample, 
alternating standard and sample. Incubate all plates, including those 
containing the standard curve, at 32 deg. C.-35 deg. C. overnight, and 
measure the diameter of each circle of inhibition. To estimate the 
potency of the sample, average the zone readings of the standard and the 
zone readings of the sample on the 3 plates used. If the sample gives a 
larger zone size than the average of the standard, add the difference 
between them to the 5.0 micrograms per milliliter zone on the standard 
curve. If the average sample value is lower than the standard value, 
subtract the difference between them from the 5.0 micrograms per 
milliliter value on the curve. From the standard curve, read the 
potencies corresponding to these corrected values of zone sizes.
    (2) Toxicity. Proceed as directed in Sec. 440.80a(b)(4) of this 
chapter, except use physiological salt solution as the diluent, and 
inject 0.5 milliliter of a solution containing 8 milligrams per 
milliliter.
    (3) Moisture. Proceed as directed in Sec. 440.80(b)(5)(i) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 440.80a(b)(5)(ii) of this 
chapter, using a solution containing 100 milligrams per milliliter.
    (5) Crystallinity. Proceed as directed in Sec. 440.80a(b)(5)(iii) of 
this chapter.
    (d) Troleandomycin used in making the capsules--(1) Potency--(i) 
Chemical method--(a) Reagents and equipment. (1) Methyl orange reagent: 
Shake 0.5 M boric acid solution for about 12 hours (to insure 
saturation) with an excess of methyl orange indicators. An alternative 
method is to heat the mixture to about 50 deg. C. and shake for about an 
hour. Then allow to cool. Filter the saturated dye solution and wash 
three times with chloroform. Store the dye solution over chloroform.
    (2) Acid-alcohol solution: Add 2 milliliters of concentrated 
sulfuric acid to 98 milliliters of absolute methyl alcohol.
    (3) Glycerin: Reagent grade.
    (4) Centrifuge tubes: 40 milliliters, glass-stoppered.

[[Page 474]]

    (b) Procedure. Prepare a chloroform solution containing 50.0 
milligrams activity of standard oleandomycin base in 200 milliliters of 
solution. Transfer 10.0 milliliters of the solution to a 100-milliliter 
volumetric flask and dilute to volume with chloroform. Transfer 2.0, 
4.0, 6.0, and 8.0 milliliters of this solution to glass-stoppered 
centrifuge tubes (40-milliliter size) and dilute to a total volume of 
20.0 milliliters each with chloroform. To the 20.0 milliliters of the 
solution present in each (40-milliliter size) centrifuge tube add 0.2 
milliliter of glacial acetic acid, 0.20 milliliter of glycerin, and 0.40 
milliliter of methyl orange reagent. Shake for 5 minutes and centrifuge 
for 3 minutes. Immediately transfer to another tube a 10.0-milliliter 
aliquot from the chloroform (lower) layer. Care must be exercised to see 
that no portion of the dye-glycerin-phase is included with the 
chloroform aliquot. Add 1.0 milliliter of acid-alcohol solution to this 
chloroform aliquot, mix well, and read the absorbancy at 535 m, 
using a 1-centimeter cell and a suitable photometer and using 
chloroform, similarly treated, as a blank. Prepare a standard curve, 
plotting the absorbance values of the standard solutions against the 
concentration expressed in micrograms per aliquot. Accurately weigh the 
sample to be tested to give 50 milligrams (estimated) of oleandomycin 
activity, dissolve in chloroform, and make to 200 milliliters with 
chloroform. Transfer 10.0 milliliters to a 100-milliliter volumetric 
flask and make to volume with chloroform. Transfer 5.0 milliliters to a 
glass-stoppered centrifuge tube and proceed as above. Determine the 
potency of the sample from the standard curve.
    (ii) Microbiological assay. Proceed as directed in paragraph (c)(1) 
of this section, except:
    (a) In lieu of the directions in paragraph (c)(1)(ii)(a) of this 
section, use the nutrient agar described in Sec. 440.80a(b)(1)(ii)(a) of 
this chapter for the seed and base layers, except add 2.0 milliliters of 
polysorbate 80 to each 100 milliliters of agar. Its pH after 
sterilization is 7.8 to 8.0.
    (b) In lieu of the directions in paragraph (c)(1)(iii) of this 
section, dissolve a suitable weighed quantity (usually 25 milligrams or 
less) of the troleandomycin working standard (obtained from the Food and 
Drug Administration) in sufficient 80 percent isopropyl alcohol-water 
solution to give a concentration of 1,000 micrograms per milliliter 
(estimated). Use the solution the day that it is prepared.
    (c) In lieu of the directions in paragraph (c)(1)(iv) of this 
section, dissolve the sample in sufficient 80 percent isopropyl alcohol-
water solution to give a convenient stock solution. Further dilute in 
0.2 M potassium phosphate buffer, pH 10.5 (35 grams of dipotassium 
phosphate plus 2 milliliters of 10 N NaOH, q.s. to 1 liter), to give a 
final concentration of 15 micrograms per milliliter (estimated).
    (d) In lieu of the directions in paragraph (c)(1)(vi) of this 
section, use the agar described in paragraph (d)(1)(ii)(a) of this 
section for both layers. Use the plates as soon after seeding as is 
practical. If they are not to be used shortly after seeding, then they 
should be refrigerated until ready for use.
    (e) In lieu of the directions for preparing the standard curve in 
paragraph (c)(1)(vii) of this section, prepare the standard curve by 
diluting the stock solution in 0.2 M potassium phosphate buffer, pH 
10.5, to give concentrations of 9.6, 12.0, 15.0, 18.8, and 23.4 
micrograms per milliliter. The 15.0 micrograms per milliliter is the 
reference concentration.
    (f) In lieu of the directions in paragraph (c)(1)(viii) of this 
section, incubate the plates at 37 deg. C. overnight. The concentration 
of the sample and standard being tested is 15.0 micrograms per 
milliliter.
    (2) Toxicity. Administer orally, by means of a cannula or other 
suitable device, to each of five mice within the weight range of 18 
grams to 25 grams, 0.5 milliliter of a suspension containing 200 
milligrams per milliliter in normal saline solution. If no animal dies 
within 48 hours, the sample is nontoxic. If one or more animals die 
within 48 hours, repeat the test, using for each test five or more 
previously unused mice weighing 20 grams (plus-minus0.5 gram) 
each; if the total deaths within 48 hours is no greater than 10 percent 
of

[[Page 475]]

the total number of animals tested, including the original test, the 
sample is nontoxic.
    (3) Moisture. Proceed as directed in Sec. 440.80a(b)(5)(i) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 440.80a(b)(5)(ii) of this 
chapter, using a saturated aqueous-ethanol (1:1) solution prepared by 
adding 100 milligrams per milliliter.
    (5) Paper chromatograph method--(i) Apparatus and reagents--(a) 
Chromatographic chamber (cylinder glass-stoppered museum jar 11.5 inches 
 x  3.5 inches).
    (b) Chromatographic paper (8 inches  x  8 inches Whatman No. 1).
    (c) 0.1 N hydrochloric acid.
    (d) Resolving solvent: Butyl acetate, benzene, nitromethane, 
pyridine (5:5:5:1 by volume).
    (e) Spray reagent: 15 grams antimony trichloride per 100 milliliters 
of chloroform.
    (ii) Procedure. Dissolve the sample in chloroform to give a solution 
containing 10 milligrams to 20 milligrams per milliliter. Prepare a 
sheet of chromatographic paper by drawing a line of origin parallel to 
and 1 inch from the edge of the paper. Wet the paper thoroughly with the 
0.1 N hydrochloric acid and blot it firmly between sheets of absorbent 
paper. Starting 2 inches in from the edges and at 1-inch intervals, 
apply 3 microliters to 5 microliters of the sample solutions to the 
starting line. Allow a few minutes for the paper to dry partially. While 
the paper is still damp, form a cylinder by bringing the outer edges 
together, allowing about 1-inch overlap, and secure with a paper clip. 
Stand the paper in the chromatographic chamber, which has been filled to 
a depth of \1/2\-inch with the resolving solvent. After the solvent 
front rises to a height of 4 inches to 5 inches above the origin, remove 
the paper from the tank and hang it up to air dry. Spray the dried paper 
with the antimony trichloride reagent. Hang the paper in a 100 deg. C. 
oven for 3 minutes. A purple spot becomes visible for trioleandomycin at 
an Rf value of about 0.85. The approximate Rf 
values for diacetyloleandomycin, monoacetyloleandomycin, and 
oleandomycin are, respectively, 0.72, 0.27, and 0.13.
    (6) Acetyl determination--(i) Apparatus and reagents. (a) One three-
necked Pyrex flask of approximately 45 milliliters capacity, pear-shaped 
with T-joints, agar inlet tube, glass-stoppered funnel, glass condenser, 
and bubble counter.
    (b) 50-milliliter Pyrex Erlenmeyer flask.
    (c) 10-milliliter burette, calibrated in 0.02 milliliter.
    (d) Anhydrous methanol, reagent grade.
    (e) 2 N sodium hydroxide solution.
    (f) Sulfuric acid solution prepared by adding 100 milliliters of 
concentrated H2SO4 to 200 milliliters of water.
    (g) 1 N barium chloride solution.
    (h) Phenolphthalein solution (1 percent in ethanol).
    (i) Water-pumped nitrogen.
    (j) NaOH solution, 0.015 N.
    (ii) Procedure. Weight accurately (to 0.01 milligram) approximately 
30 milligrams of the sample into the three-necked acetyl flask. Add 2.0 
milliliters of methanol to dissolve the sample, then add slowly with 
gentle swirling, 1.0 milliliter of NaOH solution. Connect the gas inlet 
tube with bubble counter attached, and adjust nitrogen flow to about two 
bubbles a second. Put glass-stoppered funnel in centerneck of acetyl 
flask and put about 5 milliliters of H2O in the funnel. Add a 
boiling chip to the solution and attach condenser in the refluxing 
position with water cooling. Adjust burner flame under acetyl flask to 
reflux solution gently. Reflux for 30 minutes. Cool assembly slightly 
then rinse down condenser (still in reflux position) with a few 
milliliters of H2O. Reassemble condenser to the distillation 
position and add water through the funnel to make a total of 
approximately 5 milliliters of H2O added to acetyl flask. 
Adjust burner flame so that about 5 milliliters of H2O and 
methanol is distilled over in approximately 10 minutes. Discard this 
distillate. Cool acetyl flask slightly. Acidify solution in flask by 
adding 1 milliliter of the sulfuric acid solution through the funnel. 
Adjust burner flame and distill over approximately 20

[[Page 476]]

milliliters of distillate into an Erlenmeyer flask in about 20 minutes, 
adding water through the funnel as necessary. It is important to keep 
the liquid volume in the acetyl flask around 2 milliliters to 3 
milliliters in order to obtain a quantitative recovery of the acetic 
acid. Collect a second fraction of distillate, about 10 milliliters in 
volume. As the second fraction is distilling, process the first 
fraction. Heat the first reaction and boil gently about 20 seconds. Add 
a few drops of BaCL2 solution to check if any sulfate was 
distilled over. If the sulfate is present, discard and repeat the whole 
determination. If the sulfate is absent immediately titrate the solution 
with the 0.015 N NaOH solution to a faint pink endpoint, using one drop 
of phenolphthalein solution as the indicator. Repeat the above procedure 
with the second fraction. If the second fraction requires less than 0.10 
milliliter of the 0.015 N NaOH solution and all the acetic acid has been 
distilled over, the determination is completed. If greater than this, 
collect a third fraction of approximately 10 milliliters and titrate 
this as before. Total volumes of NaOH used and calculate results as 
follows:

(Milliliters of NaOH  x  N NaOH  x  0.043  x  100)/Weight sample in 
          grams=Percent acetyl.

    (7) Crystallinity. Proceed as directed in Sec. 440.80a(b)(5)(iii) of 
this chapter.



Sec. 436.516  Tetracycline-neomycin complex powder topical; tetracycline hydrochloride-neomycin sulfate powder topical.

    (a) Potency--(1) Tetracycline-neomycin complex powder--(i) 
Tetracycline content. Proceed as directed in Sec. 436.514(a)(2), except 
use water in lieu of 0.1 N HCl for dissolving the sample. Its 
tetracycline content is satisfactory if it contains not less than 85 
percent of the equivalent number of milligrams of tetracycline 
hydrochloride that it is represented to contain.
    (ii) Neomycin content. Using 0.1 M potassium phosphate buffer, pH 
8.0, dilute an appropriate aliquot of the aqueous solution, prepared as 
directed in paragraph (a)(1) of this section, to a final concentration 
of 1  g. per milliliter (estimated), and proceed as directed in 
Sec. 436.515(c)(1), except that the neomycin standard stock solution 
described Sec. 436.517(b)(1)(iii) is used to prepare the standard curve, 
by further diluting with pH 8.0 buffer to final concentrations of 0.64, 
0.80, 1.0, 1.25, and 1.56  g. per milliliter. The 1.0  
g per milliliter solution is the reference concentration. In lieu of the 
method described in this subparagraph, the neomycin content may also be 
determined as follows. Using the aqueous solution described, prepare the 
sample and proceed as directed in Sec. 436.517(b)(1), except use 
Staphylococcus aureus (American Type Culture Collection 12715) 1 
as the test organism, which is grown and maintained on agar containing 
100  g. of tetracycline hydrochloride per milliliter of agar. 
Its neomycin content is satisfactory if it contains not less than 85 
percent of the number of milligrams that it is represented to contain.
---------------------------------------------------------------------------

    1 Available from: American Type Culture Collection, 12301 
Parklawn Drive, Rockville, MD 20852.
---------------------------------------------------------------------------

    (2) Tetracycline hydrochloride-neomycin sulfate powder--(i) 
Tetracycline hydrochloride content. Prepare the sample as directed in 
Sec. 436.514(a)(2). Use an appropriate aliquot and proceed as directed 
in Sec. 446.81a(b)(1) of this chapter. Its tetracycline hydrochloride 
content is satisfactory if it contains not less than 85 percent of the 
number of milligrams that it is represented to contain.
    (ii) Neomycin content. Use an appropriate aliquot of the solution 
prepared in paragraph (a)(2)(i) of this section and proceed as directed 
in paragraph (a)(1)(ii) of this section. Its neomycin content is 
satisfactory if it contains not less than 85 percent of the number of 
milligrams that it is represented to contain.
    (b) Sterility. Thoroughly cleanse with a suitable disinfectant the 
value (do not flame) of each container to be tested. Into each of two 
empty, sterile Erlenmeyer flasks stoppered with a cotton plug, spray 
quantitites sufficient to yield a residue of approximately the 
equivalent of 50 milligrams from 10 separate cans by removing the plug 
temporarily and using aseptic technique while spraying; allow propellant 
to evaporate, add 250 milliliters to 500 milliliters of diluting fluid B 
in lieu of diluting fluid A, and swirl the flasks to dissolve the 
contents. Then proceed as

[[Page 477]]

directed in Sec. 436.20 of this chapter using the method described in 
paragraph (e)(1) of that section.
    (c) Moisture. Proceed as directed in Sec. 536.513(c) of this 
chapter.
    (d) Tetracycline-neomycin complex used in making the drug--(1) 
Potency--(i) Tetracycline content. Dissolve the sample to be tested in 
sufficient water to give a convenient stock solution. Using an 
appropriate aliquot, proceed as directed in Sec. 446.81a(b)(1).
    (ii) Neomycin content. Using an aliquot of the stock solution 
prepared as directed in paragraph (d)(1)(i) of this paragraph, proceed 
as directed in paragraph (a)(2) of this section, except the last 
sentence of that subparagraph.
    (2) Toxicity. Proceed as directed in Sec. 440.80a(b)(4) of this 
chapter using 0.5 milliliter of a solution prepared by diluting the 
sample with physiological sodium chloride solution to contain 200 
 g. of neomycin per milliliter (estimated).
    (3) pH. Using a 1-percent aqueous solution, proceed as directed in 
Sec. 440.80a(b)(5)(ii) of this chapter.
[39 FR 18944, May 30, 1974, as amended at 40 FR 13497, Mar. 27, 1975]



Sec. 436.517  Bacitracin-neomycin tablets; zinc bacitracin-neomycin tablets; bacitracin methylene disalicylate-neomycin tablets.

    (a) Tablets--(1) Potency--(i) Bacitracin, zinc bacitracin, or 
bacitracin methylene disalicylate content. Proceed as directed in 
Sec. 448.110a(b)(1). Its content of bacitracin, zinc bacitracin, or 
bacitracin methylene disalicylate is satisfactory if it contains not 
less than 85 percent of the number of units per tablet that it is 
represented to contain.
    (ii) Neomycin content. Place 5 tablets in a blending jar and add 
thereto 200 milliliters of a 500-milliliter quantity of 0.10-percent 
phosphate buffer pH 8.0. After blending for 1 minute with a high-speed 
blender, add the remainder of the buffer. Blend again for 1 minute and 
make the proper estimated dilutions in the buffer and proceed as 
directed in paragraph (b)(1) of this section. Its content of neomycin is 
satisfactory if it contains not less than 85 percent of the number of 
milligrams of activity that it is represented to contain.
    (2) Moisture. Proceed as directed in Sec. 440.80a(b)(5)(i) of this 
chapter.
    (3) Disintegration time. Proceed as directed in Sec. 440.180a(b)(3).
    (b) Neomycin used in making the tablets--(1) Potency--(i) Cylinders 
(cups). Use cylinders described under Sec. 440.80a(b)(1)(i) of this 
chapter.
    (ii) Culture medium. Use the medium described in Sec. 440.80a(b)(1) 
(ii)(a) of this chapter for both the base and seed layers, except its pH 
after sterilization is 7.8 to 8.0.
    (iii) Working standard. Dry the working standard (obtained from the 
U.S.P. Reference Standards Committee, 46 Park Avenue, New York 16, N.Y.) 
for 3 hours at 60 deg. C. and a pressure of 5 millimeters or less and 
weigh out a sufficient quantity to make a convenient stock solution by 
diluting with a 0.1 M potassium phosphate buffer, pH 7.8 to 8.0. The 
stock solution, when stored at a temperature of approximately 15 deg. 
C., or less, may be used for a period not exceeding 1 month.
    (iv) Standard curve. Using the stock solution, prepare a daily 
standard curve as directed in Sec. 444.70a(b)(1)(iv) of this chapter, 
using solutions of the neomycin working standard in 0.1M potassium 
phosphate buffer, pH 8.0, in concentrations of 6.4, 8.0, 10.0, 12.5, and 
15.6 micrograms per milliliter if the test organism Staphylococcus 
aureus (ATCC 6538P), 1 or in concentrations of 0.64, 0.80, 
1.0, 1.25, and 1.56 micrograms per milliliter if the test organism is 
Staphylococcus epidermidis (ATCC 12228).1 The 10.0 micrograms 
per milliliter and the 1.0 microgram per milliliter concentrations are 
used as the reference points.
---------------------------------------------------------------------------

    1 See footnote 1 to Sec. 436.516.
---------------------------------------------------------------------------

    (v) Preparation of test organism. The test organism is 
Staphylococcus aureus (ATCC 6538P), 1 which is maintained on 
agar described in Sec. 440.80a(b)(1)(ii)(a) of this chapter. From a 
stock slant inoculate a Roux bottle containing this same agar and 
incubate for 24 hours at 32 deg. C.-35 deg. C. Wash the resulting growth 
from the agar surface with about 50 milliliters of sterile sodium 
chloride solution. Standardize this suspension by determining the 
dilution that will permit 80 percent light transmission through a filter 
at 6500 Angstrom units

[[Page 478]]

in a photoelectric colorimeter. The suspension may be used for 2 weeks 
if it is stored under refrigeration.Staphylococcus epidermidis (ATCC 
12228), 1 which is maintained on agar as described in 
Sec. 440.80a(b)(1)(ii)(a) of this chapter, may also be used as the test 
organism. From a stock slant, inoculate a Roux bottle containing this 
medium and incubate for 24 hours at 32 deg. C.-35 deg. C. Wash the 
resulting growth from the agar surface, using approximately 30 
milliliters of sterile sodium chloride solution. Standardize the 
suspension by determining the dilution that will permit 80 percent light 
transmission through a filter of 6500 Angstrom units in a photoelectric 
colorimeter. The suspension may be stored for 2 weeks under 
refrigeration.
    (vi) Preparation of plates. Using the agar described in subdivision 
(ii) of this subparagraph and approximately a 0.5 percent inoculum of 
the suspension described in paragraph (b)(1)(v) of this section, prepare 
the plates as directed in Sec. 440.80a(b)(1)(v) of this chapter.
    (vii) Assay. Dissolve volumetrically in 0.1 M potassium phosphate 
buffer, pH 7.8 to 8.0, the sample to be tested to make a convenient 
stock solution. Further dilute volumetrically this solution with 0.1 M 
potassium phosphate buffer, pH 7.8 to 8.0, to a final concentration of 
10.0 micrograms (estimated) per milliliter, if the test organism is 
Staphylococcusaureus or 1.0 microgram per milliliter (estimated) if the 
test organism is Staphylococcus epidermidis.
    (2) Toxicity. Proceed as directed in Sec. 440.80a(b)(4) of this 
chapter, using 0.5 milliliter of a solution prepared by diluting the 
sample to approximately 200 micrograms per milliliter with physiological 
salt solution.
    (3) Moisture. In an atmosphere of about 10 percent relative 
humidity, transfer about 100 milligrams of the finely powdered sample to 
a tared weighing bottle equipped with ground-glass top and stopper. 
Weigh the bottle and place it in a vacuum oven, tilting the stopper on 
its side so that there is no closure during the drying period. Dry at a 
temperature of 60 deg. C. and a pressure of 5 millimeters of mercury or 
less for 3 hours. At the end of the drying period fill the vacuum oven 
with air dried by passing it through a drying agent such as sulfuric 
acid or silica gel. Replace the stopper and place the weighing bottle in 
a desiccator over a desiccating agent such as phosphorous pentoxide or 
silica gel, allow to cool to room temperature, and reweigh. Calculate 
the percent loss.
    (4) pH. Proceed as directed in Sec. 440.80a(b)(5)(ii) of this 
chapter, using a solution containing 33 milligrams per milliliter.



Sec. 436.542  Acid resistance/dissolution test for enteric-coated erythromycin pellets.

    (a) Equipment. Use Apparatus 1 as described in the United States 
Pharmacopeia XX dissolution test.
    (b) Immersion fluids. All immersion fluids may be degassed by 
heating immediately prior to use.
    (1) Acid resistance medium. Use 0.06N hydrochloric acid, pH 1.2.
    (2) Dissolution medium. Dissolve 6.8 grams of monobasic potassium 
phosphate in 250 milliliters of water. Add 109 milliliters of 0.2N 
sodium hydroxide and 400 milliliters of water and adjust the resulting 
solution with 0.2N sodium hydroxide to a pH of 6.80.1. 
Dilute to 1 liter.
    (c) Procedure. Warm the immersion fluids to a temperature of 37 deg. 
5.0 deg. C. Place the contents of one capsule into the 
basket. Lower the basket into 900 milliliters of acid resistance medium 
contained in the beaker. Ensure that all air is displaced from the 
immersed basket and that the pellets remain in the basket. Rotate the 
basket at the speed of 50 revolutions per minute for an accurately timed 
period of 1 hour. Remove the basket from the fluid and immediately lower 
the basket into 900 milliliters of dissolution medium contained in the 
beaker. Again ensure that all air is displaced from the immersed basket 
and that the pellets remain in the basket. Rotate the basket at 50 
revolutions per minute for an accurately timed dissolution period of 45 
minutes. Withdraw a 25-milliliter sample of the dissolution medium from 
a point midway between the stirring shaft and the wall of the vessel and 
approximately midway in depth. Filter the sample through a Whatman 541 
filter paper or equivalent, discarding the first 2 milliliters. Assay 
for erythromycin using

[[Page 479]]

the filtrate as the test solution as directed in Sec. 436.105. Repeat 
the test on five additional capsules.
    (d) Evaluation. Use the interpretation described in the United 
States Pharmacopeia XX dissolution test.
[46 FR 16678, Mar. 13, 1981, as amended at 50 FR 47213, Nov. 15, 1985; 
52 FR 35912, Sept. 24, 1987; 54 FR 41824, Oct. 12, 1989]



Sec. 436.543  Acid resistance test for pellet-filled doxycycline hyclate capsules.

    (a) Equipment. Use Apparatus 1 as described in the United States 
Pharmacopeia XXI dissolution test.
    (b) Acid resistance medium. Use 0.06N hydrochloric acid, pH 1.2. May 
be degassed by heating immediately prior to use.
    (c) Procedure. Warm the acid resistance medium to a temperature of 
372.0  deg. C. Place the contents of one pellet-filled 
capsule into the basket. Lower the basket into a beaker containing 900 
milliliters of acid resistance medium. Ensure that all air is displaced 
from the immersed basket and that the contents of the pellet-filled 
capsule remain in the basket.

Rotate the basket at the speed of 50 revolutions per minute for an 
accurately timed period of 20 minutes. Withdraw a 5-milliliter sample of 
the acid resistance medium from a point midway between the stirring 
shaft and the wall of the vessel and approximately midway in depth (this 
is the sample solution). Assay the sample solution for doxycycline as 
described in paragraph (d) of this section. Repeat the test on five 
additional pellet-filled capsules.
    (d) Doxycycline content--(1) Preparation of working standard 
solution. Dissolve an accurately weighed portion of doxycycline hyclate 
working standard with 0.1M hydrochloric acid to obtain a concentration 
of 0.01 milligram per milliliter.
    (2) Preparation of sample solution. Dilute the 5-milliliter sample 
portion to 25 milliliters with 0.1M hydrochloric acid.
    (3) Procedure. Using a suitable spectrophotometer and 0.1M 
hydrochloric acid as the blank, determine the absorbance of each 
standard and sample solution at the absorbance peak at approximately 345 
nanometers. Determine the exact position of the absorption peak for the 
particular instrument used.
    (4) Calculations. Determine the total amount of doxycycline 
dissolved as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.105

where:
    Au=Absorbance of sample;
    As=Absorbance of standard;
    c=Concentration of working standard in milligrams; and
    d=Dilution factor of sample withdrawn from beaker.

    *If more than 15 milliliters of dissolution medium is removed, 
correct for the volume removed.

    (e) Evaluation. The pellet-filled capsule passes the test if no more 
than 50 percent of the drug is dissolved at 20 minutes. If one pellet-
filled capsule fails to meet this requirement, repeat the test on six 
additional pellet-filled capsules. No more than 2 pellet-filled capsules 
in 12 may exceed 50 percent of the drug dissolved at 20 minutes.
[50 FR 41679, Oct. 15, 1985; 50 FR 45603, Nov. 1, 1985]



Sec. 436.544  Dissolution test for pellet-filled doxycycline hyclate capsules.

    (a) Equipment. Use Apparatus 1 as described in the United States 
Pharmacopeia XXI dissolution test.
    (b) Dissolution medium. Prepare the dissolution medium as follows: 
Dissolve 10.21 grams of potassium biphthalate and 1.4 grams of sodium 
hydroxide in approximately 950 milliliters of distilled water and adjust 
the pH to 5.5 using 1M sodium hydroxide solution. Dilute with distilled 
water to 1,000 milliliters.
    (c) Procedure. Proceed as directed in the United States Pharmacopeia 
XXI dissolution test. Ensure that all air is displaced from the immersed 
basket and that the contents of the pellet-filled capsule remain in the 
basket. Rotate the basket at the speed of 50 revolutions per minute for 
an accurately timed period of 30 minutes. Withdraw a 5-milliliter sample 
of the dissolution medium from a point midway between the stirring shaft 
and the wall of the

[[Page 480]]

vessel and approximately midway in depth (this is the sample solution). 
Assay the sample solution for doxycycline as described in paragraph (d) 
of this section. Repeat the test on five additional pellet-filled 
capsules.
    (d) Doxycycline content--(1) Preparation of working standard 
solution. Dissolve an accurately weighed portion of doxycycline hyclate 
working standard with 0.1M hydrochloric acid to obtain a concentration 
of 0.01 milligram per milliliter.
    (2) Preparation of sample solution. Dilute the 5-milliliter sample 
portion to 25 milliliters with 0.1M hydrochloric acid.
    (3) Procedure. Using a suitable spectrophotometer and 0.1M 
hydrochloric acid as the blank, determine the absorbance of each 
standard and sample solution at the absorbance peak at approximately 345 
nanometers. Determine the exact position of the absorption peak for the 
particular instrument used.
    (4) Calculations. Determine the total amount of doxycycline 
dissolved as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.106

where:
    Au=Absorbance of sample;
    As=Absorbance of standard;
    c=Concentration of working standard in milligrams; and
    d=Dilution factor of sample withdrawn from beaker.

    *If more than 15 milliliters of dissolution medium is removed, 
correct for the volume removed.

    (e) Evaluation. Use the dissolution acceptance table and 
interpretation in the United States Pharmacopeia XXI.
[50 FR 41679, Oct. 15, 1985]



Sec. 436.545  Acid resistance test for erythromycin particles in tablets.

    (a) Equipment. Use Apparatus 2 as described in the United States 
Pharmacopeia XXI dissolution test.
    (b) Acid resistance medium. Use 0.1N hydrochloric acid, 500 
milliliters.
    (c) Procedure. Warm the immersion fluid to a temperature of 
370.5  deg.C. Place one tablet into a vessel containing 500 
milliliters of acid resistance medium. Rotate the paddle at the speed of 
50 revolutions per minute for an accurately timed period of 1 hour. 
Withdraw a 50-milliliter sample of the dissolution medium from a point 
midway between the stirring shaft and the wall of the vessel and 
approximately midway in depth. Filter the sample through a Whatman No. 1 
filter paper or equivalent, discarding the first 5.0 milliliters. Assay 
for dissolved erythromycin as directed in paragraph (d) of this section 
using the filtrate as the sample solution. Repeat the test on five 
additional tablets.
    (d) Arsenomolybdate colorimetric assay for dissolved erythromycin--
(1) Apparatus. Automatic analyzer consisting of (i) a liquid sampler, 
(ii) a proportioning pump, (iii) suitable spectrophotometers equipped 
with matched flow cells and analysis capability at 660 nanometers, (iv) 
a means of recording spectrophotometric readings, and (v) a manifold 
consisting of the components illustrated in the diagram in paragraph 
(d)(4) of this section.
    (2) Reagents--(i) Arsenomolybdate solutions--(a) Stock solution. 
Dissolve 100 grams of ammonium molybdate in approximately 1,700 
milliliters of water contained in a 2-liter volumetric flask. Insert an 
inert plastic coated stirring bar into the flask, and begin mixing. 
While mixing, slowly add 84 milliliters of sulfuric acid (temperature of 
solution should not exceed 50  deg.C). Dissolve 12 grams of sodium 
arsenate in 100 milliliters of water, and add to the solution in the 
flask. Remove the stirring bar, dilute with water to volume, and mix. 
Store in an amber bottle for 24 hours before using. (This solution 
should not be allowed to come into contact with rubber.)
    (b) Working solution. Dilute 1 part of stock solution with 2 parts 
of water, and mix. This solution is freshly prepared on the day of use.
    (ii) Acetate buffer, pH 4.8. Dissolve 133 grams of ACS grade sodium 
acetate crystals in about 3.5 liters of water. Adjust the pH to 
4.80.1 with glacial acetic acid. Dilute with water to 4,000 
milliliters, and mix.
    (iii) 9N Sulfuric acid. Place a 2-liter volumetric flask containing 
an inert plastic coated magnetic stirring bar and about 1,500 
milliliters of water in

[[Page 481]]

an ice bath, and begin mixing. While mixing, cautiously add 300 
milliliters of sulfuric acid. Allow the solution to cool. Remove the 
stirring bar, dilute with water to volume, and mix.
    (3) Preparation of working standard solutions--(i) Working standard 
stock solution. Accurately weigh approximately 400 milligrams of USP 
Erythromycin Reference Standard, previously dried at 60  deg.C for 3 
hours under vacuum (pressure of 5 millimeters of mercury or less), and 
transfer to a 100-milliliter volumetric flask. Dissolve and dilute with 
acetate buffer, pH 4.8 to volume, and mix.
    (ii) Working standard solutions. Pipet 5, 10, 15, and 20 milliliters 
of the standard stock solution into separate 500-milliliter volumetric 
flasks, add acetate buffer, pH 4.8 to volume, and mix. The approximate 
concentrations of these solutions (before adjusting for the standard 
potency) are 40, 80, 120, and 160 micrograms of erythromycin per 
milliliter, respectively.
    (4) Procedure. Use the working standard solutions prepared as 
described in paragraph (d)(3) of this section. The arrangement of the 
apparatus and flow of the samples and reagents are shown in the manifold 
diagram set forth following this paragraph. The sampler rate is usually 
60 per hour, but may be varied. Establish a steady state by pumping 
reagents until the record trace becomes constant. Place cups containing 
the four concentrations of working standard solutions in the sampler 
followed by no more than 12 cups of sample solutions. Then place four 
more cups containing the four concentrations of working standard 
solutions in the sampler. Repeat the sequence above for additional 
samples by bracketing standards around no more than 12 sample solutions 
at a time.

[[Page 482]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.378

    (5) System suitability test. Perform a linear regression analysis of 
absorbance versus concentration in micrograms per milliliter of the 
standards. The system is suitable for calculation if the beginning 
baseline and the ending baseline after assaying a series of standard and 
sample solutions

[[Page 483]]

does not vary by more than 2 percent transmittance, and the correlation 
coefficient for each standard curve is greater than 0.995.
    (6) Calculations. (i) Calculate the concentration of each standard 
curve solution in micrograms of erythromycin per milliliter as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.379

[GRAPHIC] [TIFF OMITTED] TR01JA93.380

[51 FR 37721, Oct. 24, 1986]



PART 440--PENICILLIN ANTIBIOTIC DRUGS--Table of Contents




                          Subpart A--Bulk Drugs

Sec.
440.1a  Sterile azlocillin sodium.
440.2a  Sterile amdinocillin.
440.3  Amoxicillin trihydrate.
440.5  Ampicillin.
440.7  Ampicillin trihydrate.
440.7a  Sterile ampicillin trihydrate.
440.8  Bacampicillin hydrochloride.
440.9a  Sterile ampicillin sodium.
440.10  Benzylpenicilloyl-polylysine concentrate.
440.11  Carbenicillin indanyl sodium.
440.13a  Sterile carbenicillin disodium.
440.15  Cloxacillin sodium monohydrate.
440.17  Cyclacillin.
440.19  Dicloxacillin sodium monohydrate.
440.19a  Sterile dicloxacillin sodium monohydrate.
440.25  Hetacillin.
440.29  Hetacillin potassium.
440.29a  Sterile hetacillin potassium.
440.36a  Sterile methicillin sodium monohydrate.
440.37a  Sterile mezlocillin sodium monohydrate.
440.41  Nafcillin sodium monohydrate.
440.41a  Sterile nafcillin sodium monohydrate.
440.49  Oxacillin sodium monohydrate.
440.49a  Sterile oxacillin sodium monohydrate.
440.55a  Sterile penicillin G benzathine.
440.71  Penicillin V.
440.73  Penicillin V potassium.
440.74a  Sterile penicillin G procaine.
440.80  Penicillin G potassium.
440.80a  Sterile penicillin G potassium.
440.81a  Sterile penicillin G sodium.
440.83a  Sterile piperacillin sodium.
440.90a  Sterile ticarcillin disodium.
440.91  Ticarcillin monosodium monohydrate.

                      Subpart B--Oral Dosage Forms

440.103  Amoxicillin oral dosage forms.
440.103a  Amoxicillin trihydrate capsules.
440.103b  Amoxicillin trihydrate for oral suspension.
440.103c  Amoxicillin trihydrate chewable tablets.
440.103d  Amoxicillin trihydrate and clavulanate potassium tablets.
440.103e  Amoxicillin trihydrate and clavulanate potassium for oral 
          suspension.
440.103f  Amoxicillin trihydrate-clavulanate potassium chewable tablets.
440.105  Ampicillin oral dosage forms.
440.105a  Ampicillin tablets.
440.105b  Ampicillin chewable tablets.
440.105c  Ampicillin capsules.
440.105d  Ampicillin for oral suspension.
440.107  Ampicillin trihydrate oral dosage forms.
440.107a  Ampicillin trihydrate chewable tablets.
440.107b  Ampicillin trihydrate capsules.
440.107c  Ampicillin trihydrate for oral suspension.
440.107d  Ampicillin trihydrate-probenecid for oral suspension.

[[Page 484]]

440.107e  Ampicillin trihydrate-probenecid capsules.
440.108  Bacampicillin hydrochloride dosage forms.
440.108a  Bacampicillin hydrochloride tablets.
440.108b  Bacampicillin hydrochloride for oral suspension.
440.111  Carbenicillin indanyl sodium tablets.
440.115  Cloxacillin sodium monohydrate oral dosage forms.
440.115a  Cloxacillin sodium monohydrate capsules.
440.115b  Cloxacillin sodium monohydrate for oral solution.
440.117  Cyclacillin oral dosage forms.
440.117a  Cyclacillin tablets.
440.117b  Cyclacillin for oral suspension.
440.119  Dicloxacillin sodium monohydrate oral dosage forms.
440.119a  Dicloxacillin sodium monohydrate capsules.
440.119b  Dicloxacillin sodium monohydrate for oral suspension.
440.125  Hetacillin oral dosage forms.
440.125a  Hetacillin chewable tablets.
440.125b  Hetacillin for oral suspension.
440.129  Hetacillin potassium capsules.
440.141  Nafcillin sodium monohydrate oral dosage forms.
440.141a  Nafcillin sodium monohydrate tablets.
440.141b  Nafcillin sodium monohydrate capsules.
440.141c  Nafcillin sodium monohydrate for oral solution.
440.149  Oxacillin sodium monohydrate oral dosage forms.
440.149a  Oxacillin sodium monohydrate capsules.
440.149b  Oxacillin sodium monohydrate for oral solution.
440.155  Penicillin G benzathine oral dosage forms.
440.155a--440.155b  [Reserved]
440.155c  Penicillin G benzathine oral suspension.
440.155d  Penicillin G benzathine tablets.
440.171  Penicillin V oral dosage forms.
440.171a  Penicillin V capsules.
440.171b  Penicillin V for oral suspension.
440.171c  Penicillin V tablets.
440.173  Penicillin V potassium oral dosage forms.
440.173a  Penicillin V potassium capsules.
440.173b  Penicillin V potassium chewable tablets.
440.173c  Penicillin V potassium tablets.
440.173d  Penicillin V potassium for oral solution.
440.180  Penicillin G potassium oral dosage forms.
440.180a  Penicillin G potassium tablets.
440.180c  Penicillin G potassium capsules.
440.180f  Penicillin G potassium for oral solution.
440.180g  Penicillin G potassium tablets for solution.

                   Subpart C--Injectable Dosage Forms

440.201  Sterile azlocillin sodium.
440.202  Sterile amdinocillin.
440.207  Sterile ampicillin trihydrate for suspension.
440.209  Ampicillin sodium injectable dosage forms.
440.209a  Sterile ampicillin sodium.
440.209b  Sterile ampicillin sodium and sulbactam sodium.
440.210  Benzylpenicilloyl-polylysine injection.
440.213  Sterile carbenicillin disodium.
440.219  Dicloxacillin sodium monohydrate injectable dosage forms.
440.219a  Sterile dicloxacillin sodium monohydrate.
440.219b  Dicloxacillin sodium monohydrate for injection.
440.229  Hetacillin potassium injectable dosage forms.
440.229a  Sterile hetacillin potassium.
440.229b  Hetacillin potassium for injection.
440.236  Methicillin sodium monohydrate for injection.
440.237  Sterile mezlocillin sodium monohydrate.
440.241  Nafcillin sodium injectable dosage forms.
440.241a  Nafcillin sodium monohydrate for injection.
440.241b  Nafcillin sodium injection.
440.249  Oxacillin sodium injectable dosage forms.
440.249a  Oxacillin sodium monohydrate for injection.
440.249b  Oxacillin sodium injection.
440.255  Penicillin G benzathine injectable dosage forms.
440.255b  Sterile penicillin G benzathine suspension.
440.255c  Sterile penicillin G benzathine-penicillin G procaine 
          suspension.
440.255d  Sterile penicillin G benzathine for suspension.
440.274  Penicillin G procaine injectable dosage forms.
440.274a  Sterile penicillin G procaine with aluminum stearate 
          suspension.
440.274b  Sterile penicillin G procaine suspension.
440.274c  Sterile penicillin G procaine for suspension.
440.280  Penicillin G potassium injectable dosage forms.
440.280a  Sterile penicillin G potassium.
440.280b  Penicillin G potassium for injection.
440.280c  Penicillin G potassium injection.
440.281  Penicillin G sodium injectable dosage forms.
440.281a  Sterile penicillin G sodium.
440.281b  Penicillin G sodium for injection.
440.283  Sterile piperacillin sodium.

[[Page 485]]

440.290  Ticarcillin disodium injectable dosage forms.
440.290a  Sterile ticarcillin disodium.
440.290b  Sterile ticarcillin disodium and clavulanate potassium.
440.290c  Ticarcillin disodium and clavulanate potassium injection.

                        Subparts D-J  [Reserved]

Subpart K--Bulk Drug Formulations for Repacking or for Manufacturing Use

440.1080a  Sterile penicillin G potassium buffered.
440.1081a  Sterile penicillin G sodium, buffered.

    Authority:  21 U.S.C. 357.



                          Subpart A--Bulk Drugs



Sec. 440.1a  Sterile azlocillin sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile azlocillin sodium is the sodium 
salt of 4-thia-1-azabicyclo[3.2.0]heptane-2-carboxylic acid, 3,3-
dimethyl-7-oxo-6-[[[[(2-oxo-1-imidazolidinyl)carbonyl] amino]phenylac-
etyl]amino]-[2S-[2,5,6(S*)]]-. It is so 
purified and dried that:
    (i) If the azlocillin sodium is not packaged for dispensing, its 
azlocillin content is not less than 859 micrograms and not more than 
1,000 micrograms of azlocillin per milligram on an anhydrous basis. If 
the azlocillin sodium is packaged for dispensing, its azlocillin content 
is not less than 859 micrograms and not more than 1,000 micrograms of 
azlocillin per milligram on an anhydrous basis and also, each container 
contains not less than 90 percent and not more than 115 percent of the 
number of milligrams of azlocillin that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 2.5 percent.
    (v) Its pH in an aqueous solution containing 100 milligrams of 
azlocillin per milliliter is not less than 6.0 and not more than 8.0.
    (vi) Its specific rotation in an aqueous solution containing 10 
milligrams of azlocillin per milliliter is +170 deg. to +200 deg..
    (vii) It gives a positive identity test for azlocillin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, specific rotation, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) If it is packaged for repacking or for use in the manufacture of 
another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams; and 5 packages, each containing 
approximately 1 gram.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If it is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, except:
    (i) Dilute Brij 35 solution. In lieu of the hydroxylamine 
hydrochloride solution described in Sec. 442.40(b)(1)(ii)(b)(1) of this 
chapter, use dilute Brij 35 solution in the reference channel. Prepare 
dilute Brij 35 solution as follows: Place 1 milliliter of Brij 35, 30 
percent solution, into a 1-liter volumetric flask containing 900 
milliliters of distilled water. Swirl gently and dilute to volume slowly 
with distilled water. Mix well.
    (ii) Buffer. In lieu of the buffer described in 
Sec. 442.40(b)(1)(ii)(b)(2) of this chapter, use the buffer prepared as 
follows: Dissolve 200 grams of primary standard tris (hydroxymethyl) 
aminomethane in sufficient distilled water to make 1 liter. Filter 
before use.
    (iii) Preparation of working standard solution. Dissolve and dilute 
an accurately weighed portion of the azlocillin working standard with 
sufficient distilled water to obtain a concentration of 1.0 milligram of 
azlocillin per milliliter.

[[Page 486]]

    (iv) Preparation of sample solutions--(a) Product not packaged for 
dispensing (micrograms of azlocillin per milligram). Dissolve and dilute 
an accurately weighed portion of the sample with sufficient distilled 
water to obtain a stock solution of 1.0 milligram of azlocillin per 
milliliter (estimated).
    (b) Product packaged for dispensing. Determine both micrograms of 
azlocillin per milligram of the sample and milligrams of azlocillin per 
container. Use separate containers for preparation of each sample 
solution as described in paragraphs (b)(1)(iv)(b)(1) and (2) of this 
section.
    (1) Micrograms of azlocillin per milligram. Dissolve and dilute an 
accurately weighed portion of the sample with sufficient distilled water 
to obtain a stock solution of 1.0 milligram of azlocillin per milliliter 
(estimated).
    (2) Milligrams of azlocillin per container. Reconstitute as directed 
in the labeling using distilled water in lieu of the reconstituting 
fluid. Then, using a suitable hypodermic needle and syringe, remove all 
of the withdrawable contents if it is represented as a single-dose 
container; or, if the labeling specifies the amount of potency in a 
given volume of the resultant preparation, remove an accurately measured 
representative portion from each container. Dilute with distilled water 
to obtain a stock solution of convenient concentration. Further dilute 
an aliquot of the stock solution with distilled water to a concentration 
of 1.0 milligram of azlocillin per milliliter (estimated).
    (v) Calculations--(a) Calculate the micrograms of azlocillin per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.032

where:

    Au=Absorbance of sample solution;
    Ps=Potency of working standard solution in micrograms per 
milliliter;
    As=Absorbance of working standard solution;
    Cu=Milligrams of sample per milliliter of sample 
solution; and
    m=Percent moisture in sample.

    (b) Calculate the azlocillin content of the single-dose vial as 
follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.033

where:

    Au=Absorbance of sample solution;
    Ps=Potency of working standard solution in micrograms per 
milliliter;
    As=Absorbance of working standard solution; and
    d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 100 milligrams of azlocillin per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the titration procedure and calculations described in paragraph 
(e)(2) of that section and preparing the sample as follows: Weigh the 
vial. Rapidly transfer a portion of the powder into the titration 
vessel, add the Karl Fischer reagent and restopper the vial immediately. 
Reweigh the vial to obtain the sample weight. A nitrogen purged glove 
bag or glove box should be used for preparing the sample.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams of azlocillin per 
milliliter.
    (6) Specific rotation. Proceed as directed in Sec. 436.210 of this 
chapter, using an aqueous solution containing 10 milligrams of 
azlocillin per milliliter and a 1.0-decimeter polarimeter tube. 
Calculate the specific rotation on an anhydrous basis.
    (7) Identity. Proceed as directed in Sec. 436.336 of this chapter.
[47 FR 53348, Nov. 26, 1982, as amended at 50 FR 1504, Jan. 11, 1985; 55 
FR 11582, Mar. 29, 1990]



Sec. 440.2a  Sterile amdinocillin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile amdinocillin is 4-thia-1-
azabicyclo[3.2.0]heptane-2-carboxylic acid, 6-[[(hexahydro-1H-azepin-1-
yl)-methylene]amino]-3,3-dimethyl-7-oxo-, [2S-
2,5,6)]-. It is so purified and dried that:

[[Page 487]]

    (i) If the amdinocillin is not packaged for dispensing, its 
amdinocillin potency is not less than 950 micrograms and not more than 
1,050 micrograms of amdinocillin per milligram on an anhydrous basis. If 
the amdinocillin is packaged for dispensing, its amdinocillin potency is 
not less than 950 micrograms and not more than 1,050 micrograms of 
amdinocillin per milligram on an anhydrous basis and also, each 
container contains not less than 90 percent and not more than 120 
percent of the number of milligrams of amdinocillin that it is 
represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 0.5 percent.
    (v) Its pH in an aqueous solution containing 100 milligrams of 
amdinocillin per milliliter is not less than 4.0 and not more than 6.2.
    (vi) It is crystalline.
    (vii) It gives a positive identity test for amdinocillin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for amdinocillin 
potency, and if packaged for dispensing, amdinocillin potency and 
container content, sterility, pyrogens, moisture, pH, crystallinity, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) If it is packaged for repacking or for use in the manufacture of 
another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If it is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 25 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Amdinocillin potency and 
container content. Proceed as directed in Sec. 436.353 of this chapter, 
using ambient temperature, an ultraviolet detection system operating at 
a wavelength of 220 nanometers, a column packed with microparticulate (3 
to 10 micrometers in diameter) reversed phase packing material such as 
octadecyl hydrocarbon bonded silicas, e.g., a Whatman ODS-3 column (25-
centimeter column having an inside diameter of 4.6 millimeters and 5 
micrometer particle size or equivalent), a flow rate of 1.0 milliliter 
per minute, and an injection volume of 20 microliters. Reagents, working 
standard and sample solutions, system suitability requirements, and 
calculations are as follows:
    (i) Reagents--(a) Buffer solution 0.01M pH 5.0. Transfer 1.36 grams 
of monobasic potassium phosphate in sufficient water to make 1,000 
milliliters of solution. Adjust the pH to 5.0  0.1 with 18N 
phosphoric acid or 10N sodium hydroxide.
    (b) Mobile phase. Mix acetonitrile (high-pressure liquid 
chromatography grade): 0.01M pH 5.0 phosphate buffer (15:85).
    (ii) Working standard and sample solutions--(a) Preparation of 
working standard solution. Prepare the working standard solution fresh 
before injection by dissolving an accurately weighed portion of the 
amdinocillin working standard with sufficient distilled water to obtain 
a stock solution containing approximately 100 micrograms of amdinocillin 
per milliliter.
    (b) Preparation of sample solutions--(1) Product not packaged for 
dispensing (micrograms of amdinocillin per milligram). Dissolve an 
accurately weighed portion of the sample with sufficient distilled water 
to obtain a solution containing 100 micrograms of amdinocillin per 
milliliter (estimated).
    (2) Product packaged for dispensing. Determine both micrograms of 
amdinocillin per milligram of the sample and milligrams of amdinocillin 
per container. Use separate containers for preparation of each sample 
solution as described in paragraphs (b)(1)(ii)(b)(2) (i) and (ii) of 
this section.
    (i) Micrograms of amdinocillin per milligram. Dissolve an accurately 
weighed

[[Page 488]]

portion of the sample with sufficient distilled water to obtain a 
solution containing 100 micrograms of amdinocillin per milliliter 
(estimated).
    (ii) Milligrams of amdinocillin per container. Reconstitute the 
sample as directed in the labeling. Then, using a suitable hypodermic 
needle and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency in a given volume of the resultant preparation, 
remove an accurately measured representative portion from each 
container. Dilute the solution thus obtained with sufficient distilled 
water to obtain a solution containing 100 micrograms of amdinocillin per 
milliliter (estimated).
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 2.5 at 5 
percent of peak height:
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,500 theoretical plates.
    (c) Resolution factor. The resolution factor (R) between the peak 
for amdinocillin and its nearest eluting impurity is satisfactory if it 
is not less than 2.5.
    (d) Coefficient of variation. The coefficient of variation (SR 
in percent) of five replicate injections is satisfactory if it is not 
more than 2.0 percent.

If the system suitability parameters have been met, then proceed as 
described in Sec. 436.353(b) of this chapter.
    (iv) Calculations. (a) Calculate the micrograms of amdinocillin per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.107

where:
Au=Area of the amdinocillin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the amdinocillin peak in the chromatogram of the 
          amdinocillin working standard;
Ps=Amdinocillin activity in the amdinocillin working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of sample solution; 
          and
m=Percent moisture content of the sample.

    (b) Calculate the amdinocillin content of the container as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.108
    
where:
Au=Area of the amdinocillin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the amdinocillin peak in the chromatogram of the 
          amdinocillin working standard:
Ps=Amdinocillin activity in the amdinocillin working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 40 milligrams of amdinocillin per 
milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams of amdinocillin per 
milliliter.
    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a potassium bromide disc containing 1 milligram of amdinocillin in 
300 milligrams of potassium bromide, prepared as described in paragraph 
(b)(l) of that section.
[50 FR 7765, Feb. 26, 1985; 50 FR 10220, Mar. 14, 1985; 50 FR 18243, 
Apr. 30, 1985; 55 FR 11582, Mar. 29, 1990]



Sec. 440.3  Amoxicillin trihydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amoxicillin trihydrate is the trihydrate 
form of D(-) a-amino-p- hydroxybenzyl penicillin. It is so 
purified and dried that:
    (i) Its potency is not less than 900 micrograms and not more than 
1,050 micrograms of amoxicillin per milligram on an anhydrous basis.
    (ii) [Reserved]

[[Page 489]]

    (iii) Its moisture content is not less than 11.5 percent and not 
more than 14.5 percent.
    (iv) Its pH in an aqueous solution containing 2 milligrams per 
milliliter is not less than 3.5 and not more than 6.0.
    (v) Its amoxicillin content is not less than 90 percent on an 
anhydrous basis.
    (vi) The acid-base titration concordance is such that the difference 
between the percent amoxicillin content when determined by nonaqueous 
acid titration and by nonaqueous base titration is not more than 6. The 
potency-acid titration concordance is such that the difference between 
the potency value divided by 10 and the percent amoxicillin content of 
the sample determined by the nonaqueous acid titration is not more than 
6. The potency-base titration concordance is such that the difference 
between the potency value divided by 10 and the percent amoxicillin 
content of the sample determined by the nonaqueous base titration is not 
more than 6.
    (vii) It is crystalline.
    (viii) It gives a positive identity test for amoxicillin trihydrate.
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, each package shall bear on its outside wrapper or 
container and the immediate container the following statement: ``For use 
in the manufacture of nonparenteral drugs only''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, amoxicillin content, concordance, crystallinity, and identity.
    (ii) Samples required: 12 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the iodometric 
assay shall be conclusive:
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 
sterile distilled water to give a stock solution containing 1.0 
milligram of amoxicillin per milliliter (estimated). Further dilute an 
aliquot of the stock solution with solution 3 to the reference 
concentration of 0.1 microgram of amoxicillin per milliliter 
(estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 2 milligrams per milliliter.
    (5) Amoxicillin content. Proceed as directed in Sec. 436.213 of this 
chapter using both the titration procedures described in paragraphs (e) 
(1) and (2) of that section. Calculate the percent amoxicillin content 
as follows:
    (i) Acid titration.
    [GRAPHIC] [TIFF OMITTED] TR01JA93.109
    
where:
A=Milliliters of lithium methoxide reagent used in titrating the sample.
B=Milliliters of lithium methoxide reagent used in titrating the blank.
m=Percent moisture content of the sample.

    Calculate the difference between the potency and the amoxicillin 
content as follows:

[[Page 490]]

[GRAPHIC] [TIFF OMITTED] TC01AP94.034


    (ii) Base titration.
    [GRAPHIC] [TIFF OMITTED] TR01JA93.110
    
where:
A=Milliliters of perchloric acid reagent used in titrating the sample.
B=Milliliters of perchloric acid reagent used in titrating the blank.
m=Percent moisture content of the sample.

    Calculate the difference between the potency and the amoxicillin 
content as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.035

    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 0.5 percent potassium bromide disc prepared as described in 
paragraph (b)(1) of that section.
[39 FR 34032, Sept. 23, 1974, as amended at 46 FR 16682, Mar. 13, 1981; 
49 FR 3458, Jan. 27, 1984; 50 FR 19918, May 13, 1985]



Sec. 440.5  Ampicillin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ampicillin is 6-[D-a-
aminobenzyl] penicillin. It is a white powder. It is so purified and 
dried that:
    (i) It contains not less than 900 micrograms and not more than 1,050 
micrograms of ampicillin per milligram on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 2.0 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 3.5 and not more than 6.0.
    (v) Its ampicillin content is not less than 90 percent on an 
anhydrous basis.
    (vi) The acid-base titration concordance is such that the difference 
between the percent ampicillin content when determined by nonaqueous 
acid titration and by nonaqueous base titration is not more than six. 
The potency-acid titration concordance is such that the difference 
between the potency value divided by 10 and the percent ampicillin 
content of the sample determined by the nonaqueous acid titration is not 
more than six. The potency-base titration concordance is such that the 
difference between the potency value divided by 10 and the percent 
ampicillin content of the sample determined by the nonaqueous base 
titration is not more than six.
    (vii) It is crystalline.
    (viii) It gives a positive identity test for ampicillin.
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5(b) of this chapter, each package shall bear on its outside 
wrapper or container and the immediate container the following 
statement, ``For use in the manufacture of nonparenteral drugs only''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, ampicillin content, concordance, crystallinity, and 
identity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
any of the following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately

[[Page 491]]

weighed portion of the sample in sufficient sterile distilled water to 
give a stock solution containing 0.1 milligram of ampicillin per 
milliliter (estimated). Further dilute an aliquot of the stock solution 
with 0.1M potassium phosphate buffer, pH 8.0 (solution 3), to the 
reference concentration of 0.1 microgram of ampicillin per milliliter 
(estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Ampicillin content. Proceed as directed in Sec. 436.213 of this 
chapter, using both the titration procedures described in paragraphs (e) 
(1) and (2) of that section. Calculate the percent ampicillin content as 
follows:
    (i) Acid titration.
    [GRAPHIC] [TIFF OMITTED] TR01JA93.111
    
where:
A=Milliliters of lithium methoxide reagent used in titrating the sample;
B=Milliliters of lithium methoxide reagent used in titrating the blank;
m=Percent moisture content of the sample.

    Calculate the difference between the potency and the ampicillin 
content as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.036

    (ii) Base titration.
    [GRAPHIC] [TIFF OMITTED] TC01AP94.037
    
where:
A=Milliliters of perchloric acid reagent used in titrating the samples;
B=Milliliters of perchloric acid reagent used in titrating the blank;
m=Percent moisture content of the sample.

    Calculate the difference between the potency and the ampicillin 
content as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.038

    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 0.5 percent potassium bromide disc, prepared as described in 
paragraph (b)(1) of that section.
[39 FR 18976, May 30, 1974, as amended at 41 FR 42649, Sept. 28, 1976; 
46 FR 16682, Mar. 13, 1981; 49 FR 3458, Jan. 27, 1984; 50 FR 19918, May 
13, 1985]



Sec. 440.7  Ampicillin trihydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ampicillin trihydrate is the trihydrate 
form of D(-)a-amino-benzyl penicillin. It is so purified and 
dried that:
    (i) It contains not less than 900 micrograms and not more than 1,050

[[Page 492]]

micrograms of ampicillin per milligram on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its loss on drying is not less than 12 percent and not more 
than 15 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 3.5 and not more than 6.0.
    (v) Its ampicillin content is not less than 90 percent on an 
anhydrous basis.
    (vi) The acid-base titration concordance is such that the difference 
between the percent ampicillin content when determined by nonaqueous 
acid titration and by nonaqueous base titration is not more than 6. The 
potency-acid titration concordance is such that the difference between 
the potency value divided by 10 and the percent ampicillin content of 
the sample determined by the nonaqueous acid titration is not more than 
6. The potency-base titration concordance is such that the difference 
between the potency value divided by 10 and the percent ampicillin 
content of the sample determined by the nonaqueous base titration is not 
more than 6.
    (vii) It is crystalline.
    (viii) It gives a positive identity test for ampicillin trihydrate.
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5(b) of this chapter, this drug shall be labeled ``ampicillin'' 
and each package shall bear on its outside wrapper or container and the 
immediate container the following statement ``For use in the manufacture 
of nonparenteral drugs only''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, ampicillin content, concordance, crystallinity, and 
identity.
    (ii) Samples required: 10 packages each containing approximately 300 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 
sterile distilled water to give a stock solution containing 0.1 
milligram of ampicillin per milliliter (estimated). Further dilute an 
aliquot of the stock solution with 0.1M potassium phosphate buffer, pH 
8.0 (solution 3), to the reference concentration of 0.1 microgram of 
ampicillin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Ampicillin content. Proceed as directed in Sec. 436.213 of this 
chapter, using both the titration procedures described in paragraphs (e) 
(1) and (2) of that section. Calculate the percent ampicillin content as 
follows:
    (i) Acid titration.

    Percent ampicillin content=[(A-B) (normality of lithium methoxide 
reagent) (349.4) (100) (100)]/[(Weight of sample in milligrams) 
(100-m)],

where:
A=Milliliters of lithium methoxide reagent used in titrating the sample;
B=Milliliters of lithium methoxide reagent used in titrating the blank;
m=Percent moisture content of the sample.

    Calculate the difference between the potency and the ampicillin 
content as follows:

Difference=(Potency in micrograms per milligram/10)-percent ampicillin 
          content.

    (ii) Base titration.

Percent ampicillin content=[(A-B) (normality of perchloric acid reagent) 
          (349.4) (100) (100)]/[(Weight of sample in milligrams) 
          (100-m)],

where:

[[Page 493]]

A=Milliliters of perchloric acid reagent used in titrating the samples;
B=Milliliters of perchloric acid reagent used in titrating the blank;
m=Percent moisture content of the sample.

    Calculate the difference between the potency and the ampicillin 
content as follows:

Difference=(Potency in micrograms per milligram/10)-percent ampicillin 
          content.

    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using an 0.5 percent potassium bromide disc, prepared as described in 
paragraph (b)(1) of that section.
[39 FR 18976, May 30, 1974, as amended at 46 FR 16683, Mar. 13, 1981; 49 
FR 3458, Jan. 27, 1984; 50 FR 19918, May 13, 1985]



Sec. 440.7a  Sterile ampicillin trihydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ampicillin trihydrate is the trihydrate 
form of D(-)-aminobenzyl penicillin. It is so purified and 
dried that:
    (i) It contains not less than 900 micrograms and not more than 1,050 
micrograms of ampicillin per milligram on an anhydrous basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its loss on drying is not less than 12 percent and not more than 
15 percent.
    (vi) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 3.5 and not more than 6.0.
    (vii) Its ampicillin content is not less than 90 percent on an 
anhydrous basis.
    (viii) The acid-base titration concordance is such that the 
difference between the percent ampicillin content when determined by 
nonaqueous acid titration and by nonaqueous base titration is not more 
than 6. The potency-acid titration concordance is such that the 
difference between the potency value divided by 10 and the percent 
ampicillin content of the sample determined by the nonaqueous acid 
titration is not more than 6. The potency-base titration concordance is 
such that the difference between the potency value divided by 10 and the 
percent ampicillin content of the sample determined by the nonaqueous 
base titration is not more than 6.
    (ix) It is crystalline.
    (x) It gives a positive identity test for ampicillin trihydrate.
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5(b) of this chapter, this drug shall be labeled 
``ampicillin.''
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, pH, ampicillin content, concordance, 
crystallinity, and identity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive:
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 
sterile distilled water to give a stock solution containing 0.1 
milligram of ampicillin per milliliter. Further dilute an aliquot of the 
stock solution with 0.1M potassium phosphate buffer, pH 8.0 (solution 3) 
to the reference concentration of 0.1 microgram of ampicillin per 
milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
in lieu of paragraph (e)(1)(i)(a), prepare the sample for test as 
follows: From each of 10 immediate

[[Page 494]]

containers, aseptically transfer approximately 300 milligrams of sample 
into a sterile 500-milliliter Erlenmeyer flask containing approximately 
400 milliliters of diluting fluid D. Add at least 200,000 Levy units 
1 of penicillinase. Repeat the process using 10 additional 
containers. Swirl both of the stoppered flasks to completely solubilize 
the suspension prior to filtration and proceed as directed in paragraph 
(e)(1)(ii) of that section.
---------------------------------------------------------------------------

    1 One Levy unit of penicillinase inactivates 59.3 units 
of penicillin G in 1 hour at 25 deg. C. and at a pH of 7.0 in a 
phosphate buffered solution of a pure alkali salt of penicillin G when 
the substrate is in sufficient concentration to maintain a zero order 
reaction.
---------------------------------------------------------------------------

    (3) Pyrogens. Proceed as directed in Sec. 436.32(f) of this chapter, 
using a solution containing 20 milligrams of ampicillin per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (7) Ampicillin content. Proceed as directed in Sec. 436.213 of this 
chapter, using both the titration procedures described in paragraph 
(e)(1) and (2) of that section. Calculate the ampicillin content as 
follows:
    (i) Acid titration.

Percent ampicillin content=[(A-B) (normality of lithium methoxide 
          reagent) (349.4) (100) (100)]/[(Weight of sample in 
          milligrams) (100-m)].

where:
A=Milliliters of lithium methoxide reagent used in titrating the sample;
B=Milliliters of lithium methoxide reagent used in titrating the blank;
m=Percent moisture content of the sample.

    Calculate the difference between the potency and the ampicillin 
content as follows:

Difference=(Potency in micrograms per milligram/10)-percent ampicillin 
          content.

    (ii) Base titration.

Percent ampicillin content=[(A-B) (normality of perchloric acid reagent) 
          (349.4) (100) (100)]/[(Weight of sample in milligrams) 
          (100-m)].

where:
A=Milliliters of perchloric acid reagent used in titrating the samples;
B=Milliliters of perchloric acid reagent used in titrating the blank;
m=Percent moisture content of the sample.

    Calculate the difference between the potency and the ampicillin 
content as follows:

Difference=(Potency in micrograms per milligram/10)-percent ampicillin 
          content.

    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (9) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 0.5 percent potassium bromide disc, prepared as described in 
paragraph (b)(1) of that section.
[39 FR 18976, May 30, 1974, as amended at 46 FR 16683, Mar. 13, 1981; 49 
FR 3458, Jan. 27, 1984; 50 FR 19918, May 13, 1985]



Sec. 440.8  Bacampicillin hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacampicillin hydrochloride is the 
hydrochloride salt of the 1-ethoxycarbonyloxyethyl ester of ampicillin. 
It is a white powder. It is so purified and dried that:
    (i) Its potency is not less than 623 micrograms and not more than 
727 micrograms of ampicillin per milligram on an ``as is'' basis.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 1.0 percent.
    (iv) Its pH in an aqueous solution containing 20 milligrams per 
milliliter is not less than 3.0 and not more than 4.5.
    (v) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, and identity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the iodometric 
assay shall be conclusive.
    (i) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, except:

[[Page 495]]

    (a) Buffer. In lieu of the buffer described in Sec. 442.40(b)(1)(ii) 
(b)(2) of this chapter, use the buffer prepared as follows: Dissolve 200 
grams of primary standard tris (hydroxymethyl) aminomethane in 
sufficient distilled water to make 1 liter. Filter before use.
    (b) Preparation of working standard solution. Use the ampicillin 
working standard. Dissolve and dilute an accurately weighed portion of 
the ampicillin working standard in sufficient distilled water to obtain 
a concentration of 1.25 milligrams of ampicillin per milliliter.
    (c) Preparation of sample solution. Dissolve and dilute an 
accurately weighed portion of the sample with sufficient distilled water 
to obtain a concentration of 1.25 milligrams of ampicillin per 
milliliter (estimated).
    (d) Calculations. Calculate the ampicillin content in micrograms per 
milligram as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.039

    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except use the ampicillin working standard.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 20 milligrams per milliliter.
    (5) Identity. Proceed as directed in Sec. 436.330 of this chapter.
[46 FR 25603, May 8, 1981, as amended at 50 FR 19918, May 13, 1985]



Sec. 440.9a  Sterile ampicillin sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile ampicillin sodium is the sodium 
salt of D(-)a-aminobenzyl penicillin. It is so purified and 
dried that:
    (i) Its potency is not less than 845 micrograms and not more than 
988 micrograms of ampicillin per milligram on an anhydrous basis. If it 
is packaged for dispensing, it contains not less than 90 percent and not 
more than 115 percent of the number of milligrams of ampicillin that it 
is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 2 percent.
    (vi) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 8.0 and not more than 10.0.
    (vii) Its ampicillin content is not less than 84.5 percent, except 
if the high-performance liquid chromatographic (HPLC) assay method is 
used, then the ampicillin content standard is not applicable.
    (viii) The potency-base titration concordance is such that the 
difference between the potency value divided by 10 and the percent 
ampicillin content of the sample determined by the nonaqueous base 
titration is not more than 6, except if the HPLC assay method is used, 
then the concordance standard is not applicable.
    (ix) It is crystalline.
    (x) It passes the identity test for ampicillin sodium.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, ampicillin content, concordance, crystallinity, 
and identity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in 
manufacturing another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (2) For sterility testing: 20 packages each containing approximately 
300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers or if each vial contains 250 milligrams or less of ampicillin 
a minimum of 24 vials.

[[Page 496]]

    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Dissolve an accurately weighed sample in sufficient sterile distilled 
water to give a stock solution containing 0.1 milligram of ampicillin 
per milliliter (estimated), for the microbiological agar diffusion assay 
and in 1.0 percent potassium phosphate buffer, pH 6.0 (solution 1), for 
the iodometric assay or for the hydroxylamine colorimetric assay to give 
a stock solution of convenient concentration. For the high-performance 
liquid chromatographic assay (HPLC), transfer an accurately weighed 
portion of ampicillin, equivalent to about 100 milligrams of anhydrous 
ampicillin, to a 100-milliliter volumetric flask. Add about 75 
milliliters of diluent (prepared as described in paragraph 
(b)(1)(ii)(d)(1)(ii) of this section), shake and sonicate, if necessary, 
to achieve complete dissolution. Also, if it is packaged for dispensing, 
reconstitute as directed in the labeling. Then using a suitable 
hypodermic needle and syringe, remove all of the withdrawable contents 
if it is represented as a single-dose container, or if the labeling 
specifies the amount of potency in a given volume of the resultant 
preparation, remove an accurately measured representative portion from 
each container. Dilute with either sterile distilled water, solution 1, 
or HPLC diluent to give a stock solution as specified above.
    (ii) Assay procedure. Use any of the following methods; however, the 
results obtained from the microbiological agar diffusion assay shall be 
conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with 0.1M potassium phosphate buffer, pH 8.0 (solution 3), to the 
reference concentration of 0.1 microgram of ampicillin per milliliter 
(estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution.
    (c) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (d) HPLC assay. Proceed as directed in Sec. 436.216 of this chapter, 
using ambient temperature, an ultraviolet detection system operating at 
a wavelength of 254 nanometers, a 4-millimeter X 5-centimeter guard 
column containing 40- to 60-micrometer diameter packing material as 
described for the analytical column, a 4-millimeter X 30-centimeter 
analytical column packed with microparticulate (3 to 10 micrometers in 
diameter) reversed phase packing material such as octadecyl hydrocarbon 
bonded silica, and a flow rate of about 2.0 milliliters per minute. 
Separately inject equal volumes (about 20 microliters) of the working 
standard preparation and the sample solution into the chromatograph, 
record the chromatogram, and measure the responses for the major peaks. 
Reagents, working standard and resolution test solution, system 
suitability requirements, and calculations are as follows:
    (1) Reagents--(i) Mobile phase. Prepare a suitably filtered and 
degassed mixture of water, acetonitrile, 1.0M monobasic potassium 
phosphate, and 1.0N acetic acid (909:80:10:1).
    (ii) Diluent. Mix 10 milliliters of 1.0M monobasic potassium and 1 
milliliter of 1.0N acetic acid, dilute with water to make 1,000 
milliliters, and mix.
    (2) Preparation of working and internal standard solutions--(i) 
Working standard solution. Dissolve a portion of ampicillin working 
standard, accurately weighed, in the diluent to obtain a solution having 
a known concentration of about 1 milligram per milliliter. Shake and 
sonicate, if necessary, to achieve complete dissolution. Use this 
solution promptly after preparation.
    (ii) Resolution test solution. Dissolve caffeine in working standard 
solution to obtain a solution containing about 1 milligram per 
milliliter.
    (3) System suitability requirements--(i) Tailing factor. The tailing 
factor (T) is satisfactory if it is not more than 1.4 at 5 percent of 
peak height.
    (ii) Resolution. The resolution (R) between the caffeine and the 
ampicillin peaks is satisfactory if it is not less

[[Page 497]]

than 2.0. The relative retention times are about 2.0 for caffeine and 
1.0 for ampicillin.
    (iii) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent) of 5 replicate 
injections is satisfactory if it is not more than 2.0 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.216(b) of this chapter. Alternate chromatographic 
conditions are acceptable provided reproducibility and resolution are 
comparable to the system. However, the sample preparation described in 
paragraph (b)(1)(i) of this section should not be changed.
    (4) Calculations. Calculate the micrograms of ampicillin per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.112

where:
Au=Area of the ampicillin peak in the chromatogram of the 
sample (at a retention time equal to that observed for the standard);
As=Area of the ampicillin peak in the chromatogram of the 
ampicillin working standard;
Ps=Ampicillin activity in the ampicillin working standard 
solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of sample solution; 
and
m=Percent moisture content of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 20 milligrams of ampicillin per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams of ampicillin per 
milliliter.
    (7) Ampicillin content. Proceed as directed in Sec. 436.213 of this 
chapter, using the titration procedure described in paragraph (e)(2) of 
that section. Calculate the ampicillin content as follow
[GRAPHIC] [TIFF OMITTED] TR01JA93.113

where:
A=Milliliters of perchloric acid reagent used in titrating the sample;
B=Milliliters of perchloric acid reagent used in titrating the blank;
m=Percent moisture content of the sample.

    Calculate the difference between the potency and the ampicillin 
content as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.114

    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (9) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the method described in paragraph (b)(2) of that section.
[39 FR 18976, May 30, 1974, as amended at 41 FR 10886, Mar. 15, 1976; 41 
FR 42649, Sept. 28, 1976; 42 FR 59857, Nov. 22, 1977; 46 FR 16683, Mar. 
13, 1981; 49 FR 3458, Jan. 27, 1984; 50 FR 19918, May 13, 1985; 52 FR 
42288, Nov. 4, 1987; 52 FR 45281, Nov. 25, 1987; 54 FR 47204, Nov. 13, 
1989]

[[Page 498]]



Sec. 440.10  Benzylpenicilloyl-polylysine concentrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Benzylpenicilloyl-polylysine concentrate 
is a pale yellow to dark yellow aqueous solution of benzylpenicilloyl e 
substituted poly-L-lysine. It contains one or more suitable and harmless 
buffers. It is so purified that:
    (i) It contains not less than 50 percent and not more than 70 
percent benzylpenicilloyl substitution on the polylysine.
    (ii) The benzylpenicilloyl concentration is not less than 1.25 x 10- 
2M and not more than 2.0 x 10- 2M.
    (iii) The penamaldate concentration is not more than 6.0 x 10- 
4M.
    (iv) The penicillenate concentration is not more than 2.0 x 10- 
4M.
    (v) Its pH is not less than 6.5 and not more than 8.5.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for percent 
benzylpenicilloyl substitution, benzylpenicilloyl content, penamaldate 
content, penicillenate content, and pH.
    (ii) Samples required: 2 vials, each containing not less than 5 
milliliters.
    (b) Tests and methods of assay--(1) Percent benzylpenicilloyl 
substitution--(i) Lysine content--(a) Equipment. Amino acid analyzer 
capable of:
    (1) Separating the hydrolysis products of benzylpenicilloyl 
polylysine into discrete components by means of an ion-exchange column.
    (2) Mixing the separated amino acid components with a ninhydrin 
reagent and promoting the reaction in a coil at elevated temperatures.
    (3) Quantitating the ninhydrin positive materials by means of a 
suitable colorimeter and recorder.
    (b) Reagents--(1) Citrate buffer: Dissolve and dilute 19.69 grams of 
sodium citrate dihydrate, 16.5 milliliters of hydrochloric acid, 0.1 
milliliter of pentachlorophenol, 5 milliliters of thiodiglycol in 900 
milliliters of distilled water; adjust to a pH of 2.2 and dilute to 1 
liter with distilled water.
    (2) Calibration mixture: Dissolve and dilute equal molar amounts of 
ammonia, and the L form of lysine in the citrate buffer to result in 
final concentrations of 2.5  x  10- 4M for each.
    (c) Preparation of standard and sample solutions--(1) Standard 
solution (standard lysine solution (2.5 x  10-4 M)). Transfer 
an accurately weighed portion of 54.8 milligrams of lysine 
dihydrochloride to a 100-milliliter volumetric flask. Dissolve and 
dilute to mark with citrate buffer. Make an accurate tenfold dilution of 
this solution with citrate buffer. The resulting standard solution is 
2.5 x 10-4 M with respect to lysine.
    (2) Sample solution. Dilute 1 milliliter of the benzylpenicilloyl-
polylysine concentrate to 10 milliliters with distilled water. Mix 1 
milliliter of the diluted solution with 1.5 milliliters of 6.0N 
hydrochloric acid and seal in an ampule under nitrogen. Hydrolyze the 
solution for 22 hours at 110 deg. C. Transfer the contents of the ampule 
quantitatively into a 50-milliliter round bottom flask and dry by rotary 
evaporation. Wash the contents and evaporate to dryness three times 
using 5-milliliter portions of distilled water. Dissolve the hydrolysate 
in 10 milliliters of citrate buffer.
    (d) Procedure. Standardize the procedure for use of the amino acid 
analyzer with the calibration mixture. Apply 0.5 milliliter of the 
lysine standard solution to the amino acid analyzer and determine the 
area of the lysine peak. Apply 0.5 milliliter of the sample solution to 
the amino acid analyzer and determine the area of the lysine peak.
    (e) Calculations. Calculate the lysine content by the following 
formula:
[GRAPHIC] [TIFF OMITTED] TR01JA93.115

where:
A=The area of the lysine peak of the sample solution.
B=The area of the lysine peak of the standard solution.
C=The percent purity of the lysine dihydrochloride.

    (ii) Benzylpenicilloyl content--(a) Reagents. (1) Mercuric chloride 
solution: Dissolve 35 milligrams of mercuric

[[Page 499]]

chloride in 500 milliliters of distilled water.
    (2) Saline phosphate buffer, pH 7.6: Dissolve 9 grams of sodium 
chloride and 1.38 grams monobasic sodium phosphate in 900 milliliters of 
distilled water, adjust to pH 7.6 and dilute to 1 liter with distilled 
water.
    (b) Preparation of sample solution. Transfer 1 milliliter of the 
benzylpenicilloyl-polylysine concentrate into a 500-milliliter 
volumetric flask and dilute to volume with saline phosphate buffer, pH 
7.6.
    (c) Procedure. Transfer 3 milliliters of the sample solution into a 
spectrophotometric cell. Using a suitable spectrophotometer and the 
saline phosphate buffer, pH 7.6, as a blank, determine the initial 
absorbance at 282 nanometers. Thereafter, react the diluted 
benzylpenicilloyl-polylysine solution with 0.02-milliliter portions of 
the mercuric chloride solution. Determine the absorbance at 282 
nanometers at 1 and 3 minutes after each addition of mercuric chloride 
solution. The increased absorbance at 282 nanometers is used in 
calculating the benzylpenicilloyl content. Calculate the 
benzylpenicilloyl content by means of the following formula:
[GRAPHIC] [TIFF OMITTED] TR01JA93.116

where:
A1=The highest absorbance at 282 nanometers
A2=The initial absorbance at 282 nanometers
22,325=The molar absorptivity of the penamaldate formed by the reaction 
of the benzylpenicilloyl moiety with the mercuric chloride at a pH of 
7.6.

Percent benzylpenicilloyl substitution=(Molar benzylpenicilloyl content 
          x  100)/Molar lysine content

    (2) Penicillenate and penamaldate content. Dilute 1 milliliter of 
the benzylpenicilloyl-polylysine concentrate to 50 milliliters with 
saline phosphate buffer, pH 7.6. Using a suitable spectrophotometer and 
the saline phosphate buffer, pH 7.6, as a blank, determine the 
absorbance at 322 and 282 nanometers. Calculate the penicillenate 
content by the following formula:
[GRAPHIC] [TIFF OMITTED] TR01JA93.117

where:
26,600=Molar absorptivity of the penicillenate moiety at 322 nanometers 
at a pH of 7.6

    Calculate the penamaldate content by the following formula:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.118
    
where:
22,325=Molar absorptivity of the penamaldate moiety at 282 nanometers at 
a pH of 7.6.

    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[39 FR 35346, Oct. 1, 1974; 39 FR 38370, Oct. 31, 1974; 39 FR 39871, 
Nov. 12, 1974; 39 FR 40946, Nov. 22, 1974]



Sec. 440.11  Carbenicillin indanyl sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Carbenicillin indanyl sodium is the 
monosodium salt of N-(2-carboxy-3,3-dimethyl-7-oxo-4-thia-1-azabicyclo 
[3.2.0] hept-6-yl)-2-phenyl-malonamic acid, 1-(5-indanyl) ester. It is 
so purified and dried that:
    (i) Its potency is not less than 659 micrograms and not more than 
769 micrograms of carbenicillin per milligram on an anhydrous basis at 
the time of certification, and not less than 630 micrograms of 
carbenicillin per milligram on an anhydrous basis at any time during the 
expiration period.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 2.0 percent.
    (iv) Its pH in an aqueous solution containing 100 milligrams per 
milliliter is not less than 5.0 nor more than 8.0.
    (v) It gives a positive result to the identity test for 
carbenicillin indanyl sodium.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 500]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, and identity.
    (ii) Samples required: Five packages, each containing approximately 
1.0 gram and one package containing approximately 2.5 grams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.300 of this chapter.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (5) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 0.5-percent potassium bromide disc prepared as described in 
paragraph (b)(1) of that section.
[39 FR 18976, May 30, 1974, as amended at 50 FR 19918, May 13, 1985]



Sec. 440.13a  Sterile carbenicillin disodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Carbenicillin disodium is the disodium 
salt of -carboxy-benzylpenicillin. It is so purified and dried 
that:
    (i) It contains not less than 770 micrograms of carbenicillin per 
milligram on an anhydrous basis. If it is packaged for dispensing, its 
carbenicillin content is not less than 90 percent and not more than 120 
percent of the number of milligrams of carbenicillin that it is 
represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 6 percent.
    (vi) Its pH in an aqueous solution containing 10 milligrams of 
carbenicillin per milliliter (or if packaged for dispensing, after 
reconstitution as directed in the labeling) is not less than 6.0 and not 
more than 8.0.
    (vii) It gives a positive identity test for carbenicillin disodium.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, and identity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams; and 5 packages, each containing 
approximately 1 gram.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1.0 percent 
potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration; and also if it is packaged for 
dispensing, reconstitute as directed in the labeling. Then, using a 
suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is represented as a single-dose container; or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. If it is a single dose container, use a 
separate needle and syringe for each container. Dilute with sufficient 
solution 1 to give a stock solution of convenient concentration. Further 
dilute the stock solution with solution 1 to the reference concentration 
of 20.0 micrograms of carbenicillin per milliliter (estimated).

[[Page 501]]

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 200 milligrams of carbenicillin per 
milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams of carbenicillin per 
milliliter (or if packaged for dispensing, use a solution prepared as 
directed for reconstitution in the labeling).
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 0.5 percent potassium bromide disc prepared as directed in 
paragraph (b)(1) of that section.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59857, Nov. 22, 1977; 45 
FR 22921, Apr. 4, 1980; 50 FR 19918, May 13, 1985; 51 FR 27532, Aug. 1, 
1986]



Sec. 440.15  Cloxacillin sodium monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cloxacillin sodium is the monohydrate 
sodium salt of 5-methyl-3-(o- chlorophenyl)-4-isoxazolyl penicillin. It 
is so purified and dried that:
    (i) Its potency is not less than 825 micrograms of cloxacillin per 
milligram.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 3 percent and not more 
than 5 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 4.5 nor more than 7.5.
    (v) Its cloxacillin content is not less than 82.5 percent.
    (vi) It passes the identity test.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this subchapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this subchapter, each such 
request shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, cloxacillin content, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 1 
percent potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration. Further dilute an aliquot of the 
stock solution with solution 1 to the reference concentration of 5 
micrograms of cloxacillin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
subchapter.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this subchapter.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this 
subchapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using an aqueous solution containing 10 milligrams per milliliter.
    (5) Cloxacillin content. Accurately weigh approximately 100 
milligrams of the sample and dissolve in sufficient 5N sodium hydroxide 
to give a total volume of 25 milliliters. Place in a boiling water bath 
for 30 minutes. Cool, acidify 1 milliliter with 1 milliliter of dilute 
sulfuric acid (1 in 2), add 8 milliliters of water, and extract with two 
25-milliliter portions of ethyl ether. Combine the ether extractives and 
extract with 25-milliliter portions of 0.1N sodium hydroxide. Combine 
the alkaline extractives and dilute to 100 milliliters with carbon 
dioxide-free water. Treat a portion of the cloxacillin working standard 
in the same manner. Using a suitable spectrophotometer, determine the 
absorbance of the solution in a 1-centimeter cell at the absorption 
peaks at 257plus-minus3 nanometers and at 
282plus-minus3 nanometers compared with a reagent blank. 
Determine the percent

[[Page 502]]

cloxacillin in the sample by means of the following calculation:
[GRAPHIC] [TIFF OMITTED] TR01JA93.119

where:
A1=Difference in absorbance for the sample between 257 
nanometers and 282 nanometers:
A2=Difference in absorbance for the cloxacillin working 
standard, similarly treated.

    (6) Identity. Proceed as directed in Sec. 436.211 of this 
subchapter, using the 0.5 percent potassium bromide disc described in 
paragraph (b)(1) of that section.
    (7) Crystallinity. Proceed as directed in Sec. 436.203 of this 
subchapter.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59857, Nov. 22, 1977; 50 
FR 19918, May 13, 1985]



Sec. 440.17  Cyclacillin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cyclacillin is 6-(1-
aminocyclohexanecarboxamido)-3,3-dimethyl-7-oxo-4-thia-1-
azabicyclo[3.2.0] heptane-2-carboxylic acid. It is a white to off-white 
powder. It is so purified and dried that:
    (i) It contains not less than 900 micrograms and not more than 1,050 
micrograms of cyclacillin per milligram.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 1.0 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 4.0 and not more than 6.5.
    (v) Its cyclacillin content is not less than 90 percent on an 
anhydrous basis.
    (vi) The acid-base titration concordance is such that the difference 
between the percent cyclacillin content when determined by nonaqueous 
acid titration and nonaqueous base titration is not more than six. The 
potency-acid titration concordance is such that the difference between 
the potency value divided by 10 and the percent cyclacillin content of 
the sample determined by the nonaqueous acid titration is not more than 
six. The potency base titration concordance is such that the difference 
between the potency value divided by 10 and the percent cyclacillin 
content of the sample determined by the nonaqueous base titration is not 
more than six.
    (vii) It is crystalline.
    (viii) It gives a positive identity test for cyclacillin.
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, each package shall bear on its outside wrapper or 
container and the immediate container the following statement, ``For use 
in the manufacture of nonparenteral drugs only.''
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, cyclacillin content, concordance, crystallinity, and identity.
    (ii) Samples required: 10 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
any of the following methods; however, the results obtained from the 
iodometric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 
sterile distilled water to give a stock solution containing 1 milligram 
of cyclacillin per milliliter (estimated). Further dilute an aliquot of 
the stock

[[Page 503]]

solution with 0.1M potassium phosphate buffer, pH 8.0 (solution 3), to 
the reference concentration of 1.0 microgram of cyclacillin per 
milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Cyclacillin content. Proceed as directed in Sec. 436.213 of this 
chapter, using both the titration procedures described in paragraph 
(e)(1) and (2) of that section. Calculate the percent cyclacillin 
content as follows:
    (i) Acid titration.
    [GRAPHIC] [TIFF OMITTED] TC01AP94.040
    
where:
A=Milliliters of lithium methoxide reagent used in titrating the sample;
B=Milliliters of lithium methoxide reagent used in titrating the blank;
m=Percent moisture content of the sample.

    Calculate the difference between the potency and the cyclacillin 
content as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.041

    (ii) Base titration.
    [GRAPHIC] [TIFF OMITTED] TR01JA93.120
    
where:
A=Milliliters of perchloric acid reagent used in titrating the sample;
B=Milliliters of perchloric acid reagent used in titrating the blank;
m=Percent moisture content of the sample.

    Calculate the difference between the potency and the cyclacillin 
content as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.121

    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 1-percent potassium bromide disc prepared as described in 
paragraph (b)(1) of that section.
[46 FR 2981, Jan. 13, 1981; 46 FR 15880, Mar. 10, 1981, as amended at 50 
FR 19918, May 13, 1985]



Sec. 440.19  Dicloxacillin sodium monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Dicloxacillin sodium monohydrate is the 
monohydrated sodium salt of 5-methyl-3-(2,6-dichlorophenyl)-4-isoxazolyl 
penicillin. It is so purified and dried that:

[[Page 504]]

    (i) Its potency is not less than 850 micrograms of dicloxacillin per 
milligram.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 3 percent nor more than 
5 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 4.5 nor more than 7.5.
    (v) Its organic chlorine content is not less than 13.0 percent nor 
more than 14.2 percent.
    (vi) Its free chloride content is not more than 0.5 percent.
    (vii) It is crystalline.
    (viii) It gives a positive identity test for dicloxacillin sodium 
monohydrate.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, organic chlorine content, free chloride content, crystallinity, and 
identity.
    (ii) Samples required: 10 containers, each containing not less than 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay, as 
follows: Dissolve an accurately weighed portion of the sample in 
sufficient 1 percent potassium phosphate buffer, pH 6.0 (solution 1), to 
give a stock solution of convenient concentration. Further dilute an 
aliquot of the stock solution with solution 1 to the reference 
concentration of 5 micrograms of dicloxacillin per milliliter 
(estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) [Reserved]
    (3) Moisture content. Proceed as directed in Sec. 436.201 of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Organic chlorine content--(i) Reagents. (a) o- Chlorobenzoic 
acid of known purity.
    (b) 0.01N Silver nitrate solution. Store in brown glass reagent 
bottle. Standardize against an accurately weighed sample of 20 to 25 
milligrams of o- chlorobenzoic acid using the procedure described in 
paragraph (b)(5)(ii) of this section.
[GRAPHIC] [TIFF OMITTED] TR01JA93.122

    (c) 0.1N Sodium hydroxide solution.
    (d) 1:1 Nitric acid solution: Mix 1 volume of concentrated nitric 
acid with 1 volume of distilled water.
    (ii) Total chlorine. (Caution--The analyst should wear safety 
glasses and use a suitable shield between himself and the apparatus. The 
glassware must be scrupulously clean.) Accurately weigh 20 to 25 
milligrams of the sample and place it on the center of a piece of 
halide-free filter paper measuring about 4 centimeters square (this is 
specially cut paper with a fuse strip attached to the area that holds 
the sample), and fold the paper to enclose it. Place 10 milliliters of 
0.1N sodium hydroxide into an oxygen combustion flask (Schoniger flask), 
and flush the air from the flask with a stream of rapidly flowing 
oxygen. Place the sample into the platinum sample holder and ignite the 
fuse strip by suitable means. If the strip is ignited outside the flask, 
immediately plunge the stopper into the flask, invert so that the sodium 
hydroxide solution makes a seal around

[[Page 505]]

the stopper, and hold the stopper firmly in place. If the ignition is 
carried out in a closed system, the inversion of the flask may be 
omitted. After combustion is completed, shake the flask vigorously, add 
a small amount of distilled water to the collar to insure an air tight 
seal, and allow to stand for not less than 10 minutes with intermittent 
shaking. Transfer to a suitable titration vessel, heat on a steam bath 
for 20 to 30 minutes, cool to room temperature, add 5 milliliters of 
nitric acid solution, and titrate potentiometrically with 0.01N silver 
nitrate, using one silver electrode and one silver/silver chloride 
electrode.
[GRAPHIC] [TIFF OMITTED] TR01JA93.123

    (iii) Free chloride. Accurately weigh 100 to 150 milligrams of 
sample directly into a titration flask, dissolve in 10 milliliters of 
0.1N sodium hydroxide, and add about 20 milliliters of distilled water, 
heat this solution on the steam bath 20 to 30 minutes. Cool to room 
temperature, add 5 milliliters of 1:1 nitric acid solution and titrate 
potentiometrically with 0.01N silver nitrate using one silver electrode 
and one silver/silver chloride electrode.
[GRAPHIC] [TIFF OMITTED] TR01JA93.124

    (iv) Organic chlorine. Percent organic chlorine=Percent total 
chlorine-percent free chloride.
    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 1 percent potassium bromide disc described in paragraph (b)(1) 
of that section.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59857, Nov. 22, 1977; 44 
FR 10378, Feb. 20, 1979; 50 FR 19918, May 13, 1985]



Sec. 440.19a  Sterile dicloxacillin sodium monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile dicloxacillin sodium monohydrate 
is the monohydrated sodium salt of 5-methyl-3-(2,6-dichloro-phenyl)-4-
isoxazolyl penicillin. It is so purified and dried that:
    (i) Its potency is not less than 850 micrograms of dicloxacillin per 
milligram. If it is packaged for dispensing, its potency is satisfactory 
if it contains not less than 90 percent and not more than 120 percent of 
the number of milligrams of dicloxacillin that it is represented to 
contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not less than 3 percent and not more 
than 5 percent.
    (vi) Its pH in an aqueous solution containing 10 milligrams per 
milliliter or when reconstituted as directed in the labeling, if it is 
packaged for dispensing is not less than 4.5 nor more than 7.5.
    (vii) Its organic chlorine content is not less than 13.0 percent and 
not more than 14.2 percent.
    (viii) Its free chloride content is not more than 0.5 percent.
    (ix) It is crystalline.
    (x) It gives a positive identity test for dicloxacillin sodium 
monohydrate.
    (2) Labeling. If this drug is packaged for dispensing, in addition 
to the labeling requirements of Sec. 432.5 of this chapter, this drug 
shall be labeled ``sterile dicloxacillin sodium''.

[[Page 506]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, organic chlorine content, free chloride content, 
crystallinity, and identity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Sample preparation. Dissolve an accurately weighed sample in 
sufficient 1 percent potassium phosphate buffer, pH 6.0 (solution 1), 
for the microbiological agar diffusion assay and the hydroxylamine 
colorimetric assay or in distilled water for the iodometric assay, to 
give a stock solution of convenient concentration; and also if it is 
packaged for dispensing, reconstitute as directed in the labeling. Then, 
using a suitable hypodermic needle and syringe, remove all of the 
withdrawable contents if it is represented as a single-dose container, 
or if the labeling specifies the amount of potency in a given volume of 
the resultant preparation, remove an accurately measured representative 
portion from each container. Dilute with either solution 1 or distilled 
water, as specified above, to give a stock solution of convenient 
concentration.
    (ii) Assay procedures. Use any of the following methods; however, 
the results obtained from the microbiological agar diffusion assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 5 micrograms of 
dicloxacillin per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
subchapter.
    (c) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this subchapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 20 milligrams of dicloxacillin per 
milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using an aqueous solution containing 10 milligrams per milliliter (or 
using a solution reconstituted as directed in the labeling if it is 
packaged for dispensing).
    (7) Organic chlorine content. Proceed as directed in 
Sec. 440.19(b)(5).
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (9) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 1 percent potassium bromide disc prepared as directed in 
paragraph (b)(1) of that section.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59857, Nov. 22, 1977; 50 
FR 19918, May 13, 1985]



Sec. 440.25  Hetacillin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Hetacillin is 6-(2,2-Dimethyl-5-oxo-4-
phenyl-1-imidazolidinyl)-3,3-dimethyl-7-oxo-4-thia-1-
azabicyclo[3.2.0]heptane-2-carboxylic acid. It occurs as a fine, white 
to off-white powder. It is so purified and dried that:
    (i) Its potency is not less than 810 micrograms of ampicillin per 
milligram.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 1.0 percent.

[[Page 507]]

    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 2.5 nor more than 5.5.
    (v) Its hetacillin content is not less than 90 and not more than 105 
percent.
    (vi) It gives a positive identity test for hetacillin.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, hetacillin content, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed for 
ampicillin in Sec. 436.105 of this chapter, using the ampicillin working 
standard as the standard of comparison and preparing the sample for 
assay as follows: Dissolve an accurately weighed sample in sufficient 
0.1M potassium phosphate buffer, pH 8.0 (solution 3), to give a stock 
solution of convenient concentration. Further dilute the stock solution 
with solution 3 to the reference concentration of 0.1 microgram of 
ampicillin per milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Hetacillin content--(i) Reagents--(a) Hydrochloric acid-acetone 
solution. Dilute 8.5 milliliters of concentrated hydrochloric acid to 1 
liter with acetone and mix well. Use for 1 day only.
    (b) p-Dimethylaminocinnamalde- hyde solution. Dissolve 0.5 gram of 
p- dimethylaminocinnamaldehyde in sufficient hydrochloric acid-acetone 
solution to a final volume of 100 milliliters and shake well, filtering 
if necessary. Prepare immediately before use.
    (ii) Preparation of standard solutions. Transfer about 100 
milligrams of the hetacillin working standard, accurately weighed, to a 
200-milliliter volumetric flask. Add 150 milliliters of refrigerated 
distilled water and 20 milliliters of 1N hydrochloric acid, shake, 
dilute to volume with distilled water, and mix well. Transfer 0.5, 1.0, 
and 2.0 milliliters into three respective 25-milliliter volumetric 
flasks. Add 1.5 and 1.0 milliliters of 0.1N hydrochloric acid 
respectively to the first and second flasks to bring the volume in each 
to 2.0 milliliters.
    (iii) Blank. Use 2.0 milliliters of 0.1N hydrochloric acid in a 25-
milliliter volumetric flask.
    (iv) Preparation of sample solutions. Using a mortar and pestle, 
grind the sample to a fine powder. Transfer an accurately weighed 
portion of about 100 milligrams to a 200-milliliter volumetric flask. 
Add 150 milliliters of refrigerated distilled water and 20 milliliters 
of 1N hydrochloric acid, shake, dilute to volume with distilled water, 
and mix well. Transfer 1.0 milliliter to a 25-milliliter volumetric 
flask, add 1.0 milliliter of 0.1N hydrochloric acid, and mix.
    (v) Procedure. To each of the flasks containing standards, blank, 
and sample, add 15 milliliters of hydrochloric acid-acetone solution and 
mix. Then add 3 milliliters of p- dimeth- ylaminocinnamaldehyde solution 
to each and mix. Add 3 milliliters of 0.1N hydrochloric acid to each, 
dilute to volume with hydrochloric acid-acetone solution, mix well, and 
allow to stand at 25 deg. C. for exactly 30 minutes. (Filter the sample 
solutions, if necessary, to remove any turbidity.) Using a suitable 
spectrophotometer, read the absorbance values of standard and sample 
solutions at a wavelength of 515 nanometers against the blank. Plot the 
absorbance values of the standards versus their concentrations and read 
the sample concentration from this standard response line.
    (vi) Calculations.
    [GRAPHIC] [TIFF OMITTED] TR01JA93.125
    
where:
C=Concentration in milligrams of hetacillin per milliliter of the final 
          solution of the sample obtained from the standard response 
          line.

[[Page 508]]

P=Hetacillin content of the hetacillin working standard in percent.

    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 1 percent potassium bromide disc prepared as directed in 
paragraph (b)(1) of that section.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59857, Nov. 22, 1977; 44 
FR 10379, Feb. 20, 1979; 50 FR 19918, May 13, 1985]



Sec. 440.29  Hetacillin potassium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality and purity. Hetacillin potassium is the potassium salt 
of hetacillin. It occurs as a fine, white to light buff powder. It is so 
purified and dried that:
    (i) Its potency is not less than 735 micrograms of ampicillin per 
milligram.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 1.0 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 7.0 and not more than 9.0.
    (v) Its hetacillin content is not less than 82 percent and not more 
than 95.5 percent.
    (vi) It gives a positive identity test for hetacillin potassium.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, hetacillin content, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed for 
ampicillin in Sec. 436.105 of this chapter, using the ampicillin working 
standard as the standard of comparison and preparing the sample for 
assay as follows: Dissolve an accurately weighed sample in sufficient 
0.1M potassium phosphate buffer pH 8.0 (solution 3), to give a stock 
solution of convenient concentration. Further dilute the stock solution 
with solution 3 to the reference concentration of 0.1 microgram of 
ampicillin per milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Hetacillin content. Proceed as directed in Sec. 440.25(b)(5), 
except use about 110 milligrams of sample and calculate the hetacillin 
content as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.126

where:
C=Concentration in milligrams of hetacillin per milliliter of the final 
          solution of the sample obtained from the standard response 
          line.
P=Hetacillin content of the hetacillin working standard in percent.

    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 1 percent potassium bromide disc prepared as directed in 
paragraph (b)(1) of that section.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59857, Nov. 22, 1977; 44 
FR 10379, Feb. 20, 1979; 50 FR 19918, May 13, 1985]



Sec. 440.29a  Sterile hetacillin potassium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Hetacillin potassium is the potassium 
salt of hetacillin. It occurs as a fine, white to light buff powder. It 
is so purified and dried that:
    (i) Its potency is not less than 735 micrograms of ampicillin per 
milligram. If it is packaged for dispensing, its potency is satisfactory 
if it contains not less than 90 percent and not more than 120 percent of 
the number of milligrams of ampicillin that it is represented to 
contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 1.0 percent.

[[Page 509]]

    (vi) Its pH in an aqueous solution containing 10 milligrams per 
milliliter (or when reconstituted as directed in the labeling, if it is 
packaged for dispensing) is not less than 7.0 and not more than 9.0.
    (vii) Its hetacillin content is not less than 82 percent and not 
more than 95.5 percent.
    (viii) It gives a positive identity test for hetacillin potassium.
    (ix) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, hetacillin content, identity, and crystallinity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers, except if each contains less than 450 milligrams, a minimum 
of 16 immediate containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed for 
ampicillin in Sec. 436.105 of this chapter, using the ampicillin working 
standard as the standard of comparison and preparing the sample for 
assay as follows: Dissolve an accurately weighed sample in sufficient 
0.1M potassium phosphate buffer, pH 8.0 (solution 3), to give a stock 
solution of convenient concentration; and also if it is packaged for 
dispensing, reconstitute as directed in the labeling. Then, using a 
suitable hypodermic needle and syringe, remove the withdrawable contents 
from each container represented as a single-dose container; or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, withdraw an accurately measured representative 
portion from each container. Dilute the sample thus obtained with 
sufficient solution 3 to give a stock solution of convenient 
concentration. Further dilute the stock solution with solution 3 to the 
reference concentration of 0.1 microgram of ampicillin per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter 
using a solution containing the equivalent of 18 milligrams of 
ampicillin per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter (or using a 
solution reconstituted as directed in the labeling, if it is packaged 
for dispensing).
    (7) Hetacillin content. Proceed as directed in Sec. 440.25(b)(5), 
except use about 110 milligrams of sample and calculate the potassium 
hetacillin content as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.127

where:
C=Concentration in milligrams of hetacillin per milliliter of the final 
          solution of the sample obtained from the standard response 
          line.
P=Hetacillin content of the hetacillin working standard in percent.

    (8) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 1 percent potassium bromide disc prepared as directed in 
paragraph (b)(1) of that section.
    (9) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19876, May 30, 1974, as amended at 42 FR 59857, Nov. 22, 1977; 43 
FR 2393, Jan. 17, 1978; 44 FR 10379, Feb. 20, 1979; 50 FR 19918, May 13, 
1985]

[[Page 510]]



Sec. 440.36a  Sterile methicillin sodium monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Methicillin sodium monohydrate is the 
monohydrated sodium salt of (2,6-dimethoxyphenyl) penicillin. It is so 
purified and dried that:
    (i) It contains not less than 815 micrograms of methicillin per 
milligram.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not less than 3 percent and not more 
than 6 percent.
    (vi) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 5.0 and not more than 7.5.
    (vii) Its methicillin content is not less than 81.5 percent.
    (viii) It is crystalline.
    (ix) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this subchapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this subchapter, each such 
request shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, methicillin content, crystallinity, and 
identity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams, plus one package containing approximately 
2 grams.
    (b) For sterility testing: 20 packages, each containing 
approximately 600 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 1 
percent potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration. Further dilute an aliquot of the 
stock solution with solution 1 to the reference concentration of 10 
micrograms of methicillin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
subchapter.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this subchapter.
    (2) Sterility. Proceed as directed in in Sec. 436.20 of this 
subchapter, using the method described in paragraph (e)(1) of that 
section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 60 milligrams of methicillin per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this 
subchapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using an aqueous solution containing 10 milligrams per milliliter.
    (7) Methicillin content. Dissolve an accurately weighed portion of 
the sample in a sufficient accurately measured volume of distilled water 
to obtain a concentration of 0.2 milligram of methicillin per milliliter 
(estimated). Treat a portion of the methicillin working standard in the 
same manner. Using a suitable spectrophotometer equipped with a 1-
centimeter quartz cell and distilled water as the blank, determine the 
absorbance at 280 nanometers. If a recording spectrophotometer is used, 
record the ultraviolet absorption spectrum from 250 nanometers to 300 
nanometers. If a nonrecording spectrophotometer is used, determine the 
absorbance (on a solution containing 10 milligrams per 100 milliliters) 
at the 280-nanometer absorption peak. (The exact position of the peak 
should be determined for the particular instrument used.) Calculate as 
follows:

[[Page 511]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.128


    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
subchapter.
    (9) Identity. Using the sample solution prepared as described in 
paragraph (b)(7) of this section, determine the absorbancies at the 
absorption maximum at 280 nanometers and at the absorption minimum at 
264 nanometers. The ratio of the two

                     A280/A264

should be not less than 1.30 and not more than 1.45.
[39 FR 18976, May 30, 1974, as amended at 40 FR 15089, Apr. 4, 1975; 42 
FR 59858, Nov. 22, 1977; 50 FR 19918, May 13, 1985]



Sec. 440.37a  Sterile mezlocillin sodium monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile mezlocillin sodium monohydrate is 
the monohydrate sodium salt of (2S, 5R, 6R)-3,3-dimethyl-6-[(R)-2-[3-
(methylsulfonyl)-2-oxo-1-imidazolidine-carboxamido]-2-phenylacetamido]-
7-oxo-4-thia-1-azabicyclo[3.2.0] heptane-2-carboxylic acid. It is so 
purified and dried that:
    (i) It contains not less than 838 micrograms and not more than 978 
micrograms of mezlocillin per milligram on an anhydrous basis. If it is 
packaged for dispensing, its mezlocillin content is not less than 90 
percent and not more than 115 percent of the number of milligrams of 
mezlocillin that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 6.0 percent.
    (vi) Its pH in an aqueous solution containing 100 milligrams per 
milliliter is not less than 4.5 and not more than 8.0.
    (vii) The specific rotation in an aqueous solution containing 10 
milligrams of mezlocillin per milliliter at 25 deg. C is 
185 deg.plus-minus10 deg..
    (viii) It gives a positive identity test for mezlocillin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, specific rotation, and identity.
    (ii) Samples required:
    (a) If it is packaged for repacking or for use in the manufacture of 
another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams; and 5 packages, each containing 
approximately 1 gram.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If it is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the hydroxylamine 
colorimetric assay shall be conclusive.
    (i) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, except:
    (a) Buffer. In lieu of the buffer described in 
Sec. 442.40(b)(1)(ii)(b)(2) of this chapter, use the buffer prepared as 
follows: Dissolve 200 grams of primary standard tris (hydroxymethyl) 
aminomethane in sufficient distilled water to make 1 liter. Filter 
before use.
    (b) Preparation of working standard solution. Dissolve and dilute an 
accurately weighed portion of the

[[Page 512]]

mezlocillin working standard with sufficient distilled water to obtain a 
concentration of 2.0 milligrams of mezlocillin per milliliter.
    (c) Preparation of sample solution. Dissolve an accurately weighed 
portion of the sample with sufficient distilled water to obtain a stock 
solution of convenient concentration; also, if packaged for dispensing, 
reconstitute as directed in the labeling using distilled water in lieu 
of the reconstituting fluid. Then using a suitable hypodermic needle and 
syringe, remove all of the withdrawable contents if it is represented as 
a single-dose container; or, if the labeling specifies the amount of 
potency in a given volume of the resultant preparation, remove an 
accurately measured representative portion from each container. Dilute 
with distilled water to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of the stock solution with 
distilled water to a concentration of 2.0 milligrams of mezlocillin per 
milliliter (estimated).
    (d) Calculations--(1) Calculate the mezlocillin content in 
micrograms per milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.129

where:

    Au=Absorbance of sample solution;
    Pa=Potency of working standard solution in micrograms per 
milliliter;
    As=Absorbance of working standard solution;
    Wu=Milligrams of sample per milliliter of sample 
solution.

    (2) Calculate the mezlocillin content of the single-dose vial as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.130

where:

    Au=Absorbance of sample solution;
    Pa=Potency of working standard solution in micrograms per 
milliliter;
    As=Absorbance of working standard solution;
    d=Dilution factor of the sample.

    (3) Calculate the mezlocillin content of the multiple-dose vial as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.131

where:

    Au=Absorbance of sample solution;
    Pa=Potency of working standard solution in micrograms per 
milliliter;
    As=Absorbance of working standard solution;
    d=Dilution factor of the sample;
    n=Volume of sample solution assayed.

    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 100 milligrams of mezlocillin per 
milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams of mezlocillin per 
milliliter.
    (7) Specific rotation. Dilute an accurately weighed sample with 
sufficient distilled water to obtain a concentration of approximately 10 
milligrams of mezlocillin per milliliter. Proceed as directed in 
Sec. 436.210 of this chapter, using a 1-decimeter polarimeter tube.
    (8) Identity. Proceed as directed in Sec. 436.311 of this chapter, 
diluting the sample with distilled water to a concentration of 4 
milligrams of mezlocillin per milliliter, except:
    (i) Use the mezlocillin working standard and dilute with distilled 
water to a concentration of 4 milligrams of mezlocillin per milligram;
    (ii) In lieu of the ninhydrin spray solution, after the plate is 
dried with a current of warm air, expose the plate to iodine vapors for 
about 30 seconds; and
    (iii) Mezlocillin has an Rf value of about 0.67.
[46 FR 58298, Dec. 1, 1981, as amended at 50 FR 19918, 19919, May 13, 
1985]



Sec. 440.41  Nafcillin sodium monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality,

[[Page 513]]

and purity. Nafcillin sodium monohydrate is the monohydrated sodium salt 
of 6-(2-ethoxy-1-naphthamido) penicillanic acid. It is so purified and 
dried that:
    (i) It contains not less than 820 micrograms of nafcillin per 
milligram.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 3.5 percent and not more 
than 5.3 percent.
    (iv) Its pH in an aqueous solution containing 30 milligrams per 
milliliter is not less than 5.0 and not more than 7.0.
    (v) It is crystalline.
    (vi) Its nafcillin content is not less than 82.0 percent.
    (vii) It gives a positive identity test for nafcillin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, crystallinity, nafcillin content, and identity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods: however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 1 
percent potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration. Further dilute an aliquot of the 
stock solution with solution 1 to the reference concentration of 2 
micrograms of nafcillin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 30 milligrams per milliliter.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(b) of this 
chapter.
    (6) Nafcillin content. Dissolve an accurately weighed portion of the 
sample in a sufficient accurately measured volume of distilled water to 
obtain a concentration of 0.05 milligram of nafcillin per milliliter 
(estimated). Treat a portion of the nafcillin working standard in the 
same manner. Using a suitable spectrophotometer equipped with quartz 
cells and distilled water as a blank, scan the absorption spectra of the 
sample and the nafcillin working standard solutions between the 
wavelengths of 245 nanometers and 340 nanometers. Determine the 
absorbance of the sample and working standard solutions at the 
absorption maximum at 280plus-minus3 nanometers. (The exact 
position of the maximum should be determined for the particular 
instrument used.) Calculate as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.132

    (7) Identity. The absorption spectrum of the sample determined as 
directed in paragraph (b)(6) of this section compares qualitatively with 
that of the nafcillin working standard.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59858, Nov. 22, 1977; 46 
FR 16683, Mar. 13, 1981; 50 FR 19918, 19919, May 13, 1985]

[[Page 514]]



Sec. 440.41a  Sterile nafcillin sodium monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile nafcillin sodium monohydrate is 
the monohydrated sodium salt of 6-(2-ethoxy-1-naphthamido) penicillanic 
acid. It is so purified and dried that:
    (i) It contains not less than 820 micrograms of nafcillin per 
milligram.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not less than 3.5 nor more than 5.3 
percent.
    (vi) Its pH in an aqueous solution containing 30 milligrams per 
milliliter is not less than 5.0 and not more than 7.0.
    (vii) It is crystalline.
    (viii) Its nafcillin content is not less than 82.0 percent.
    (ix) It gives a positive identity test for nafcillin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, crystallinity, nafcillin content, and identity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods: however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 1 
percent potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration. Further dilute an aliquot of the 
stock solution with solution 1 to the reference concentration of 2 
micrograms of nafcillin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 80 milligrams of nafcillin per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 30 milligrams per milliliter.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(b) of this 
chapter.
    (8) Nafcillin content. Proceed as directed in Sec. 440.41(b)(6).
    (9) Identity. The absorption spectrum of the sample determined as 
directed in paragraph (b)(8) of this section compares qualitatively with 
that of the nafcillin working standard.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59858, Nov. 22, 1977; 45 
FR 16474, Mar. 14, 1980; 45 FR 22921, Apr. 4, 1980; 50 FR 19918, 19919, 
May 13, 1985]



Sec. 440.49  Oxacillin sodium monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxacillin sodium monohydrate is the 
monohydrated sodium salt of 5-methyl-3-phenyl-4-isoxazolyl penicillin. 
It is so purified and dried that:
    (i) It contains not less than 815 and not more than 950 micrograms 
of oxacillin per milligram.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 3.5 and not more than 
5.0 percent.
    (iv) Its pH in an aqueous solution containing 30 milligrams per 
milliliter is not less than 4.5 and not more than 7.5.
    (v) Its oxacillin content is not less than 81.5 percent and not more 
than 95.0 percent.
    (vi) It is crystalline.

[[Page 515]]

    (vii) It gives a positive identity test for the oxacillin moiety.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, oxacillin content, crystallinity, and identity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
any of the following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 1.0 
percent potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration. Further dilute an aliquot of the 
stock solution with solution 1 to the reference concentration of 5.0 
micrograms of oxacillin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 30 milligrams per milliliter.
    (5) Oxacillin content. Place approximately 60 milligrams of sample, 
accurately weighed, into a 100-milliliter volumetric flask. Dissolve and 
fill to volume with distilled water. Pipette a 5.0-milliliter aliquot of 
the sample solution into a 22- by 200-millimeter test tube, and add 5 
milliliters of 10 N NaOH. Mix the solution, and place the tube in a 
boiling water bath for 60 minutes. Cool the tube, carefully add 10 
milliliters of 6 N HCl, mix, and replace the tube in the boiling water 
bath for 10 minutes. Position the tube in the bath so that the liquid 
level in the tube is the same as the liquid level in the bath. After 
heating, remove the tube from the bath, carefully agitate the contents 
of the tube, and cool to room temperature. Quantitatively transfer the 
contents of the tube to a 250-milliliter volumetric flask. Add 
approximately 200 milliliters of freshly boiled and cooled distilled 
water, then 4.0 milliliters of 7.5 N NH4OH, and dilute to 
volume with freshly boiled and cooled distilled water. Treat a sample of 
the oxacillin working standard in the same manner. Determine the 
absorbance of the sample and working standard solutions on a suitable 
spectrophotometer at 235 nanometers against a reagent blank, and 
calculate as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.133

    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (7) Identity. Use the sample solution prepared as described in 
paragraph (b)(5) of this section and record the ultraviolet spectrum 
between 230 nanometers and 260 nanometers. It should be basically 
identical to that of the standard similarly treated.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59858, Nov. 22, 1977; 49 
FR 5096, Feb. 10, 1984; 50 FR 19918, 19919, May 13, 1985]



Sec. 440.49a  Sterile oxacillin sodium monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality,

[[Page 516]]

and purity. Sterile oxacillin sodium monohydrate is the monohydrated 
sodium salt of 5-methyl-3-phenyl-4-isoxazolyl penicillin. It is so 
purified and dried that:
    (i) It contains not less than 815 and not more than 950 micrograms 
of oxacillin per milligram.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not less than 3.5 and not more than 5.0 
percent.
    (vi) Its pH in an aqueous solution containing 30 milligrams per 
milliliter is not less than 4.5 and not more than 7.5.
    (vii) Its oxacillin content is not less than 81.5 percent and not 
more than 95.0 percent.
    (viii) It is crystalline.
    (ix) It gives a positive identity test for the oxacillin moiety.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, oxacillin content, crystallinity, and identity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams, plus one package containing approximately 
2 grams.
    (b) For sterility testing: 20 packages, each containing 
approximately 600 milligrams.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
any of the following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 1.0 
percent potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration. Further dilute an aliquot of the 
stock solution with solution 1 to the reference concentration of 5.0 
micrograms of oxacillin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 20 milligrams of oxacillin per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 30 milligrams per milliliter.
    (7) Oxacillin content. Proceed as directed in Sec. 440.49(b)(5).
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (9) Identity. Proceed as directed in Sec. 440.49(b)(7).
[39 FR 18976, May 30, 1974, as amended at 42 FR 59858, Nov. 22, 1977; 50 
FR 19918, 19919, May 13, 1985]



Sec. 440.55a  Sterile penicillin G benzathine.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G benzathine is the N,N'- 
dibenzylethylenediamine salt of penicillin G. It is so purified and 
dried that:
    (i) Its potency is not less than 1,090 units and not more than 1,272 
units per milligram.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not less than 5.0 percent and not more 
than 8 percent.
    (vi) Its pH in a 1:1 mixture of absolute ethyl alcohol and water 
containing 0.5 milligram per milliliter is not less than 4.0 and not 
more than 6.5.
    (vii) Its penicillin G content is not less than 57.9 percent and not 
more than 71.6 percent.
    (viii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 517]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, penicillin G content, and crystallinity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 600 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the iodometric 
assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately measured representative portion of the sample in 
sufficient absolute methyl alcohol to give a solution of convenient 
concentration. Immediately, further dilute with 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to the reference concentration of 
1.0 unit of penicillin G per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section, except 
use medium C in lieu of medium A, medium F in lieu of medium E, and 
during the period of incubation shake the tubes at least once daily.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(d) of this chapter, 
using a solution containing 4,000 units of penicillin G per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, except 
prepare the sample as follows: Dissolve 50 milligrams of sample with 50 
milliliters of absolute ethyl alcohol. Add 50 milliliters of distilled 
water and mix well.
    (7) Penicillin G content. Accurately weigh approximately 50 
milligrams of the sample, dissolve in absolute methyl alcohol, and 
dilute to 100 milliliters with absolute methyl alcohol. Treat a portion 
of the working standard in the same manner. Using a suitable 
spectrophotometer equipped with a quartz cell and absolute methyl 
alcohol as the blank, determine the absorbance at 263 nanometers. 
Calculate the percent penicillin G as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.134

    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[42 FR 59858, Nov. 22, 1977, as amended at 45 FR 16472, Mar. 14, 1980; 
49 FR 6092, Feb. 17, 1984; 50 FR 19918, 19919, May 13, 1985]



Sec. 440.71  Penicillin V.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin V is 3,3-dimethyl - 7-oxo-6-
(2-phenoxyacetamido)-4-thia-1-azabicyclo[3.2.0]heptane-2-carboxylic 
acid. It is so purified and dried that:
    (i) Its potency is not less than 1,525 units nor more than 1,780 
units per milligram.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 2.0 percent.
    (iv) Its pH in a saturated aqueous solution is not less than 2.5 and 
not more than 4.0.
    (v) Its penicillin V content is not less than 90 percent and not 
more than 105 percent.
    (vi) It is crystalline.
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, each package shall bear on its outside wrapper or 
container and the immediate container the statement ``For

[[Page 518]]

use in the manufacture of nonparenteral drugs only.''
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, penicillin V content, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
any of the following methods; however, the results obtained from the 
bioassay method shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample (approximately 30 milligrams) in 
2.0 milliliters of absolute methyl alcohol. Further dilute an aliquot of 
this solution with sufficient 1 percent potassium phosphate buffer, pH 
6.0 (solution 1), to the reference concentration of 1.0 unit of 
penicillin V per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
saturated aqueous solution prepared by adding approximately 30 
milligrams per milliliter.
    (5) Penicillin V content. Accurately weigh approximately 20 
milligrams of the sample, dissolve in absolute methanol, and make to 100 
milliliters with absolute methyl alcohol. Treat a portion of the working 
standard in the same manner. Using a suitable spectrophotometer equipped 
with a quartz cell and absolute methyl alcohol as the blank, determine 
the absorbance of the peak at 276 nanometers. Calculate the percent 
penicillin V as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.135

    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[42 FR 59859, Nov. 22, 1977, as amended at 50 FR 19918, 19919, May 13, 
1985]



Sec. 440.73  Penicillin V potassium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin V potassium is the potassium 
salt of 3,3-dimethyl-7-oxo-6-(2-phenoxyacetamido) - 4 -thia - 1 - 
azabicyclo[3.2.0]heptane - 2 - carboxylic acid. It is so purified and 
dried that:
    (i) Its potency is not less than 1,380 units nor more than 1,610 
units per milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 1.5 percent.
    (iv) Its pH in an aqueous solution containing 30 milligrams per 
milliliter is not less than 4.0 and not more than 7.5.
    (v) Its penicillin V content is not less than 81.2 percent and not 
more than 94.7 percent.
    (vi) It is crystalline.
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, each package shall bear on its outside wrapper or 
container and the immediate container the statement ``For use in the 
manufacture of nonparental drugs only.''
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, penicillin V content, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.

[[Page 519]]

    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
any of the following methods; however, the results obtained from the 
iodometric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to obtain a stock solution of 
convenient concentration. Further dilute an aliquot of the stock 
solution with solution 1 to the reference concentration of 1.0 unit of 
penicillin V per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 30 milligrams per milliliter.
    (5) Penicillin V content. Dissolve and dilute approximately 20 
milligrams of the sample, accurately weighed to 100 milliliters with 
0.1N sodium hydroxide solution. Treat a portion of the penicillin V 
working standard in the same manner. Using a suitable spectrophotometer 
equipped with a quartz cell and 0.1N sodium hydroxide solution as the 
blank, determine the absorbance of the peak at 275 nanometers. Calculate 
the percent penicillin V as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.136

    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[42 FR 59859, Nov. 22, 1977, as amended at 50 FR 19918, 19919, May 13, 
1985]



Sec. 440.74a  Sterile penicillin G procaine.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G procaine is 3,3 - dimethyl - 
7 - oxo - 6 - (2 - phenylacetamido) - 4 - thia - 1 - azabicyclo 
[3.2.0]heptane-2-carboxylic acid 2-(diethylamino) ethyl p-aminobenzoate 
compound (1:1). It is so purified and dried that:
    (i) Its potency is not less than 900 units and not more than 1,050 
units per milligram.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not less than 2.8 percent and not more 
than 4.2 percent.
    (vi) Its pH in a saturated aqueous solution (about 300 milligrams 
per milliliter) is not less than 5.0 and not more than 7.5.
    (vii) Its penicillin G content is not less than 51.0 percent and not 
more than 59.6 percent.
    (viii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, penicillin G content, and crystallinity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 600 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the iodometric 
assay shall be conclusive.

[[Page 520]]

    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to give a stock solution of 
convenient concentration. Further dilute an aliquot of the stock 
solution with solution 1 to the reference concentration of 1.0 unit of 
penicillin G per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed 
Sec. 436.205 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
add sufficient penicillinase to diluting fluid A and swirl the flask to 
completely solubilize the sample before filtration. If the product 
contains lecithin, use diluting fluid D in lieu of A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(h) of this chapter, 
using a solution containing 2,000 units of penicillin G per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
saturated solution prepared by suspending 300 milligrams of sample per 
milliliter.
    (7) Penicillin G content. Proceed as directed in Sec. 436.316 of 
this chapter.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[42 FR 59860, Nov. 22, 1977, as amended at 45 FR 16472, Mar. 14, 1980; 
45 FR 22921, Apr. 4, 1980; 50 FR 19918, 19919, May 13, 1985]



Sec. 440.80  Penicillin G potassium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality and purity. Penicillin G potassium is potassium 3,3-
dimethyl-7-oxo-6-(2-phenylacetamido)-4-thia-1-azabicyclo[3.2.0]heptane-
2-carboxylate. It is so purified and dried that:
    (i) Its potency is not less than 1,440 units and not more than 1,680 
units per milligram.
    (ii) Its loss on drying is not more than 1.5 percent.
    (iii) The pH of an aqueous solution containing 60 milligrams per 
milliliter is not less than 5.0 and not more than 7.5.
    (iv) Its penicillin G content is not less than 80.8 percent and not 
more than 94.3 percent.
    (v) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of test and assays on the batch for potency, loss on 
drying, pH, penicillin G content, and crystrallinity.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research: 10 packages, each containing approximately 300 milligrams.
    (b) Test and methods of assay--(1) Potency. Proceed as directed in 
Sec. 440.80a(b)(1).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 60 milligrams per milliliter.
    (4) Penicillin G content. Proceed as directed in Sec. 436.316 of 
this chapter.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[55 FR 38674, Sept. 20, 1990]



Sec. 440.80a  Sterile penicillin G potassium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G potassium is potassium 3,3-
dimethyl-7-oxo-6-(2-phenylacetamido)-4-thia-1-azabicyclo [3.2.0] 
heptane-2-carboxylate. It is so purified and dried that:
    (i) Its potency is not less than 1,440 units and not more than 1,680 
units per milligram. If it is packaged for dispensing, its potency is 
satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of units of penicillin G that it is represented to 
contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]

[[Page 521]]

    (v) Its loss on drying is not more than 1.5 percent.
    (vi) Its pH in an aqueous solution containing 60 milligrams per 
milliliter is not less than 5.0 and not more than 7.5.
    (vii) Its penicillin G content is not less than 80.8 percent and not 
more than 94.3 percent.
    (viii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, pH, penicillin G content, and crystallinity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 600 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Dissolve an accurately weighed portion of the sample in sufficient 1 
percent potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration; also, if it is packaged for 
dispensing, reconstitute as directed in the labeling. Then using a 
suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is represented as a single-dose container; or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute with solution 1 to give a stock 
solution of convenient concentration.
    (ii) Assay procedures. Use any of the following methods; however, 
the results obtained from the iodometric assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 unit of penicillin 
G per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (c) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 20,000 units of penicillin G per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 60 milligrams per milliliter.
    (7) Penicillin G content. Proceed as directed in Sec. 436.316 of 
this chapter.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[42 FR 59860, Nov. 22, 1977, as amended at 45 FR 16472, Mar. 14, 1980; 
45 FR 22922, Apr. 4, 1980; 50 FR 19918, 19919, May 13, 1985]



Sec. 440.81a  Sterile penicillin G sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G sodium is sodium 3,3-
dimethyl-7-oxo-6-(2 - phenylacetamido) - 4 - thia - 1 - azabicyclo 
[3.2.0] heptane-2-carboxylate. It is so purified and dried that:
    (i) Its potency is not less than 1,500 units and not more than 1,750 
units per milligram. If it is packaged for dispensing, its content is 
satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of units of penicillin G that it is represented to 
contain.
    (ii) It is sterile.

[[Page 522]]

    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its loss on drying is not more than 1.5 percent.
    (vi) Its pH in an aqueous solution containing 60 milligrams per 
milliliter is not less than 5.0 and not more than 7.5.
    (vii) Its penicillin G content is not less than 84.5 percent and not 
more than 98.5 percent.
    (viii) It is crystalline.
    (ix) It passes the test for heat stability if it does not show a 
loss of more than 10 percent of its original potency.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, pH, penicillin G content, crystallinity, and 
heat stability.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 600 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Dissolve an accurately weighed portion of the sample in sufficient 1 
percent potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration; also, if it is packaged for 
dispensing, reconstitute as directed in the labeling. Then using a 
suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is represented as a single-dose container; or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
aliquot from each container. Dilute with solution 1 to give a stock 
solution of convenient concentration.
    (ii) Assay procedures. Use any of the following methods; however, 
the results obtained from the iodometric assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 unit of penicillin 
G per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (c) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 20,000 units of penicillin G per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 60 milligrams per milliliter.
    (7) Penicillin G content. Proceed as directed in Sec. 436.316 of 
this chapter.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (9) Heat stability. Proceed as directed in Sec. 436.214 of this 
chapter.
[42 FR 59861, Nov. 22, 1977, as amended at 45 FR 22922, Apr. 4, 1980; 50 
FR 19918, 19919, May 13, 1985]



Sec. 440.83a  Sterile piperacillin sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile piperacillin sodium is the sodium 
salt of (2S,5R, 6R)-6-[(R)-2-(4-ethyl-2,3-dioxo-1-piperazine-
carboxamido)-2-phenylacetamido]-3,3-dimethyl-7-oxo-4-thia-1-azabicyclo-
[3.2.0]heptane-2-carboxylate. It is so purified and dried that:
    (i) Its potency is not less than 863 micrograms and not more than 
1,007

[[Page 523]]

micrograms of piperacillin per milligram on an anhydrous basis. If it is 
packaged for dispensing, it contains not less than 90.0 percent and not 
more than 120.0 percent of the number of grams of piperacillin that it 
is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 1.0 percent.
    (vi) Its pH in an aqueous solution containing 400 milligrams per 
milliliter is not less than 5.5 and not more than 7.5.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, and pH.
    (ii) Samples required:
    (a) If it is packaged for repacking or for use in the manufacture of 
another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams; and 5 packages, each containing 
approximately 1 gram.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If it is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.334 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 150 milligrams of piperacillin per 
milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the sample preparation method described in paragraph (d)(4) of 
that section.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 400 milligrams per milliliter
[47 FR 15769, Apr. 13, 1982, as amended at 50 FR 19918, 19919, May 13, 
1985]



Sec. 440.90a  Sterile ticarcillin disodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile ticarcillin disodium is 6-
[(carboxy-3-thienylacetyl)] amino - 3, 3-dimethyl - 7 - oxo - 4 - thia - 
1 - azabicyclo[3.2.0]heptane-2-carboxylic acid disodium salt. It is so 
purified and dried that:
    (i) It contains not less than 800 micrograms of ticarcillin per 
milligram on an anhydrous basis. If it is packaged for dispensing, its 
ticarcillin content is not less than 90 percent and not more than 115 
percent of the number of milligrams of ticarcillin that it is 
represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 6.0 percent.
    (vi) Its pH in an aqueous solution containing 10 milligrams of 
ticarcillin per milliliter (or if packaged for dispensing after 
reconstitution as directed in the labeling) is not less than 6.0 and not 
more than 8.0.
    (vii) It gives a positive identity test for ticarcillin.
    (viii) Its ticarcillin content is not less than 80 percent and not 
more than 94 percent on an anhydrous basis.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, identity, and ticarcillin content.
    (ii) Samples required:
    (a) If it is packaged for repacking or for use in the manufacture of 
another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams; and 5 packages, each containing 
approximately 1 gram.

[[Page 524]]

    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If it is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1.0 percent 
potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration; and also, if it is packaged for 
dispensing, reconstitute as directed in the labeling. Then using a 
suitable hypodermic needle and syringe, remove all the withdrawable 
contents if it is represented as a single-dose container; or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. If it is a single-dose container, use a 
separate needle and syringe for each container. Dilute with sufficient 
solution 1 to give a stock solution of convenient concentration. Further 
dilute an aliquot of the stock solution with solution 1 to the reference 
concentration of 5.0 micrograms of ticarcillin per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 100 milligrams of ticarcillin per 
milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams of ticarcillin per 
milliliter (or if packaged for dispensing, use a solution prepared as 
directed for reconstitution in the labeling).
    (7) Identity and ticarcillin content. Transfer an accurately weighed 
portion of approximately 40 milligrams of the sample to a 100-milliliter 
volumetric flask. Dissolve and dilute to volume with distilled water. 
Transfer 5.0 milliliters of this solution to another 100-milliliter 
volumetric flask and dilute to volume with 0.1N methanolic hydrochloric 
acid (prepared by diluting 0.8 milliliter of 12N hydrochloric acid to 
100 milliliters with methyl alcohol). Treat a portion of the ticarcillin 
standard in the same manner. Using a suitable spectrophotometer equipped 
with a 1.0-centimeter quartz cell and 0.1N methanolic acid as a blank, 
scan the absorption spectrum of the methanolic solution of the sample 
and the standard between the wavelengths of 300 and 200 nanometers. 
Determine the absorbance of each solution at the maxima, at 
approximately 230 nanometers. The spectrum of the samples should compare 
qualitatively with that of the ticarcillin working standard. Determine 
the percent ticarcillin as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.137

where: m=Percent moisture in the sample.
[42 FR 14093, Mar. 15, 1977, as amended at 50 FR 19918, 19919, May 13, 
1985]



Sec. 440.91  Ticarcillin monosodium monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ticarcillin monosodium monohydrate is 6-
[(carboxy-3-thienylacetyl)] amino-3,3-dimethyl-7-oxo-4-thia-1-azabicyclo 
[3.2.0] heptane-2-carboxylic acid monosodium salt monohydrate. It is so 
purified and dried that:

[[Page 525]]

    (i) Its ticarcillin potency is not less than 890 micrograms of 
ticarcillin per milligram calculated on an anhydrous basis.
    (ii) Its moisture content is not less than 4.0 and not more than 6.0 
percent.
    (iii) The pH of an aqueous solution containing 10 milligrams of 
ticarcillin per milliliter is not less than 2.5 and not more than 4.0.
    (iv) It gives a positive identity test for ticarcillin.
    (v) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, identity, and crystallinity.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research: 10 packages, each containing approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Ticarcillin potency. Determine 
the micrograms of ticarcillin activity per milligram of sample. Proceed 
as directed in Sec. 436.355 of this chapter using the equipment, 
conditions, reagents, and system suitability requirements as described 
in Sec. 440.290b(b), except use the resolution test solution to 
determine resolution in lieu of the working standard solution. Prepare 
the working standard solution, sample solution, and resolution test 
solution and calculate the micrograms of ticarcillin per milligrams as 
follows:
    (i) Preparation of working standard, sample, and resolution test 
solutions--(A) Working standard solution. Accurately weigh a quantity of 
the ticarcillin working standard containing the equivalent of 
approximately 90 milligrams of ticarcillin activity and transfer to a 
100-milliliter volumetric flask. Dissolve and dilute to volume with 
diluent pH 6.4 phosphate buffer prepared as described in 
Sec. 440.290b(b)(1)(i)(c).
    (B) Sample solution. Dissolve an accurately weighed portion of the 
sample with diluent pH 6.4 buffer as prepared in 
Sec. 440.290b(b)(1)(i)(c) to obtain a solution containing 0.9 milligram 
of ticarcillin activity per milliliter (estimated).
    (C) Resolution test solution. Accurately weigh a quantity of the 
ticarcillin working standard containing the equivalent of approximately 
90 milligrams of ticarcillin activity and transfer to a 100-milliliter 
volumetric flask. Prepare a solution of the clavulanic acid working 
standard containing the equivalent of 30 milligrams of clavulanic acid 
activity in a 100-milliliter volumetric flask. Dissolve and dilute to 
volume with diluent. Transfer 10 milliliters of this solution into the 
flask containing the ticarcillin standard. Dilute the combined standard 
solution to volume with diluent and mix. Use within 8 hours of 
preparation.
    (ii) Calculations. Calculate the micrograms of ticarcillin per 
milligram as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.042

where:
Au=Area of the ticarcillin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the ticarcillin peak in the chromatogram of the 
          ticarcillin working standard;
Ps=Ticarcillin activity in the ticarcillin working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of ticarcillin sample per milliliter of sample 
          solution; and
m=Percent moisture content of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams of ticarcillin per 
milliliter.
    (4) Identity. Proceed as directed in Sec. 440.90a(b)(7).
    (5) Crystallinity. Proceed as directed in Sec. 436.203 of this 
chapter.
[55 FR 5839, Feb. 20, 1990]

[[Page 526]]



                      Subpart B--Oral Dosage Forms



Sec. 440.103  Amoxicillin oral dosage forms.



Sec. 440.103a  Amoxicillin trihydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amoxicillin trihydrate capsules are 
composed of amoxicillin trihydrate with or without one or more suitable 
and harmless lubricants, diluents, and drying agents, enclosed in a 
gelatin capsule. Each capsule contains amoxicillin trihydrate equivalent 
to 250 milligrams or 500 milligrams of amoxicillin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of amoxicillin that it is 
represented to contain. Its moisture content is not more than 14.5 
percent. It passes the identity test. The amoxicillin trihydrate used 
conforms to the standards prescribed by Sec. 440.3(a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``amoxicillin 
capsules''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The amoxicillin trihydrate used in making the batch for potency, 
moisture, pH, amoxicillin content, concordance, crystallinity, and 
identity.
    (b) The batch for potency, moisture, and identity.
    (ii) Samples required:
    (a) The amoxicillin trihydrate used in making the batch: 12 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
either of the following methods; however, the results obtained from the 
iodometric assay shall be conclusive:
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 0.1M potassium phosphate buffer, pH 
8.0 (solution 3), to give a stock solution of convenient concentration. 
Blend for 3 to 5 minutes. Remove an aliquot and further dilute with 
solution 3 to the reference concentration of 0.1 microgram of 
amoxicillin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution. Prepare the sample as 
follows: Place the contents of a representative number of capsules into 
a high-speed glass blender jar and add sufficient distilled water to 
give a convenient concentration. Blend for 3 to 5 minutes. Further 
dilute an aliquot with distilled water to the prescribed concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Identity. Proceed as directed in Sec. 436.311 of this chapter, 
preparing the sample solution as follows: Dissolve an accurately weighed 
portion of the amoxicillin capsule contents in 0.1N hydrochloric acid to 
give a solution containing 4 milligrams of amoxicillin per milliliter.
[39 FR 34033, Sept. 23, 1974, as amended at 49 FR 3458, Jan. 27, 1984; 
50 FR 19919, May 13, 1985]



Sec. 440.103b  Amoxicillin trihydrate for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amoxicillin trihydrate for oral 
suspension is a mixture of amoxicillin trihydrate with one or more 
suitable and harmless colorings, flavorings, buffers, sweetening 
ingredients, preservatives, stabilizers, and suspending agents. When 
reconstituted as directed in the labeling, it contains amoxicillin 
trihydrate equivalent to either 25 or 50 milligrams of amoxicillin per 
milliliter. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
amoxicillin that it is represented to contain. Its moisture content is 
not more than 3.0 percent. Its pH, when reconstituted as directed

[[Page 527]]

in the labeling, is not less than 5.0 and not more than 7.5. It passes 
the identity test. The amoxicillin trihydrate used conforms to the 
standards prescribed by Sec. 440.3(a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``amoxicillin for 
oral suspension''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The amoxicillin trihydrate used in making the batch for potency, 
moisture, pH, amoxicillin content, concordance, crystallinity, and 
identity.
    (b) The batch for potency, moisture, pH, and identity.
    (ii) Samples required:
    (a) The amoxicillin trihydrate used in making the batch: 12 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
either of the following methods; however, the results obtained from the 
iodometric assay shall be conclusive:
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute the drug as directed in the labeling. Place an accurately 
measured representative portion of the sample into a suitable volumetric 
flask and dilute to volume with 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to give a stock solution of convenient concentration. Mix 
well. Further dilute an aliquot with solution 3 to the reference 
concentration of 0.1 microgram of amoxicillin per milliliter 
(estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution. Prepare the sample as 
follows: Reconstitute the drug as directed in the labeling. Place an 
accurately measured aliquot (usually a single dose) into an 
appropriately sized volumetric flask and dilute to volume with distilled 
water. Mix well. Further dilute with distilled water to the prescribed 
concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the suspension reconstituted as directed in the labeling.
    (4) Identity. Proceed as directed in Sec. 436.311 of this chapter, 
preparing the sample solution as follows: From an aliquot of suspension 
prepared in accordance with the label, make either a 6.25:1 dilution for 
the 25-milligrams-per-milliliter dosage; or a 12.5:1 dilution for the 
50-milligrams-per-milliliter dosage, with 0.1N hydrochloric acid. The 
slight dilution of the acid does not have a significant effect on the 
test.
[39 FR 34033, Sept. 23, 1974, as amended at 49 FR 3458, Jan. 27, 1984; 
50 FR 19919, May 13, 1985; 54 FR 47351, Nov. 14, 1989]



Sec. 440.103c  Amoxicillin trihydrate chewable tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amoxicillin trihydrate chewable tablets 
are composed of amoxicillin trihydrate with or without one or more 
suitable lubricants, diluents, preservatives, drying agents, flavorings, 
and colorings. Each tablet contains amoxicillin trihydrate equivalent to 
either 125 or 250 milligrams of amoxicillin. Its potency is satisfactory 
if it contains not less than 90 percent and not more than 120 percent of 
the number of milligrams of amoxicillin that it is represented to 
contain. Its moisture content is not more than 6.0 percent. It passes 
the identity test. The amoxicillin trihydrate used conforms to the 
standards prescribed by Sec. 440.3(a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by

Sec. 432.5 of this chapter, this drug shall be labeled ``amoxicillin 
tablets.''
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assay on:

[[Page 528]]

    (a) The amoxicillin trihydrate used in making the batch for potency, 
moisture, pH, amoxicillin content, concordance, crystallinity, and 
identity.
    (b) The batch for potency, moisture, and identity.
    (ii) Samples required:
    (a) The amoxicillin trihydrate used in making the batch: 12 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 tablets.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
either of the following methods; however, the results obtained from the 
iodometric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec.  436.105 of this chapter, preparing the sample for assay as 
follows: Place a representative number of tablets into a high-speed 
glass blender jar with sufficient 0.1M potassium phosphate buffer, pH 
8.0 (solution 3), to obtain a stock solution of convenient 
concentration. Blend for 3 to 5 minutes. Remove an aliquot and dilute 
with solution 3 to the reference concentration of 0.1 microgram of 
amoxicillin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec.  436.204 of this 
chapter, preparing the sample as follows: Place a representative number 
of tablets into a high-speed glass blender jar containing sufficient 1 
percent potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration. Blend for 5 minutes. Further 
dilute with solution 1 to the prescribed concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Identity. Proceed as directed in Sec. 436.311 of this chapter, 
preparing the sample as follows: Using a mortar and pestle, grind a 
representative number of tablets into a fine powder. Dissolve an 
accurately weighed amount of this powder in 0.1N hydrochloric acid to 
give a solution containing 4 milligrams of amoxicillin per milliliter.
[45 FR 64569, Sept. 30, 1980, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.103d  Amoxicillin trihydrate and clavulanate potassium tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amoxicillin trihydrate and clavulanate 
potassium tablets are composed of amoxicillin trihydrate and clavulanate 
potassium with or without one or more suitable lubricants, diluents, and 
binders. Each tablet contains amoxicillin trihydrate equivalent to 
either 250 or 500 milligrams of amoxicillin and clavulanate potassium 
equivalent to 125 milligrams of clavulanic acid. Its amoxicillin 
trihydrate content is satisfactory if it contains not less than 90 
percent and not more than 120 percent of the number of milligrams of 
amoxicillin that it is represented to contain. Its clavulanate potassium 
content is satisfactory if it contains not less than 90 percent and not 
more than 120 percent of the number of milligrams of clavulanic acid 
that it is represented to contain. Its moisture content is not more than 
7 percent if it contains 250 milligrams of amoxicillin and not more than 
10 percent if it contains 500 milligrams of amoxicillin. It passes the 
dissolution test if the quantity Q, at 30 minutes, is 85 percent or 
greater if it contains 250 milligrams of amoxicillin and 75 percent or 
greater if it contains 500 milligrams of amoxicillin. The amoxicillin 
trihydrate conforms to the standards prescribed by Sec. 440.3(a)(1). The 
clavulanate potassium conforms to the standards prescribed by 
Sec. 455.15(a)(1) of this chapter.
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``amoxicillin and 
clavulanate potassium tablets''.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The amoxicillin trihydrate used in making the batch for potency, 
moisture, pH, amoxicillin content, concordance, crystallinity, and 
identity.
    (b) The clavulanate potassium used in making the batch for 
clavulanic acid content, moisture, pH, identity, and clavam-2-
carboxylate content.

[[Page 529]]

    (c) The batch for amoxicillin content, clavulanic acid content, 
moisture, and dissolution rate.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The amoxicillin trihydrate used in making the batch: 12 
packages, each containing approximately 300 milligrams.
    (b) The clavulanate potassium used in making the batch: 12 packages, 
each containing approximately 300 milligrams.
    (c) The batch: A minimum of 100 tablets.
    (b) Tests and methods of assay--(1) Amoxicillin and clavulanic acid 
contents. Proceed as directed in Sec. 436.351 of this chapter, using 
ambient temperature, an ultraviolet detection system operating at a 
wavelength between 220 and 230 nanometers, and a column packed with 
microparticulate (3 to 10 micrometers in diameter) reversed phase 
packing material such as octadecyl silane bonded silica. Reagents, 
working standard and sample solutions, system suitability requirements, 
and calculations for amoxicillin or clavulanic acid content are as 
follows:
    (i) Reagents--(a) 0.5M Sodium phosphate buffer solution, pH 4.4. 
Transfer 7.8 grams of monobasic sodium phosphate to a 1-liter volumetric 
flask and dissolve in 900 milliliters of distilled water. Adjust the pH 
to 4.4  0.1 with 18N phosphoric acid or 10N sodium 
hydroxide. Dilute to volume with distilled water. Mix well.
    (b) Mobile phase. Mix methanol: 0.05M sodium phosphate buffer 
solution, pH 4.4 (5:95 v/v) and ultrasonicate for no less than 2 
minutes. Degas by passing through a 0.5-micron filter with vacuum. The 
mobile phase may be sparged with the helium through a 2-micrometer metal 
filter for the duration of the analysis. Adjust the ratio of methanol to 
aqueous buffer as necessary to obtain satisfactory retention of the 
peaks.
    (ii) Working standard and sample solutions--(a) Preparation of 
working standard solution. Accurately weigh and transfer into a 200-
milliliter volumetric flask approximately 100 milligrams of amoxicillin 
working standard and approximately 50 milligrams of the clavulanic acid 
working standard. Dissolve and dilute to volume with distilled water. 
Use within 8 hours after preparation.
    (b) Preparation of sample solution. To obtain a concentration of 0.5 
milligram of amoxicillin per milliliter, dissolve a representative 
number of tablets in water with the aid of a magnetic stirrer or 
ultrasonication. Filter a small aliquot through Whatman 42 filter paper 
or equivalent, discarding the first 10 milliliters of filtrate. 
Alternatively, a suitable membrane filter may be used. Prepare samples 
not more than 1 hour before the chromatographic injection.
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.5.
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 550 theoretical plates.
    (c) Resolution factor. The resolution factor (R) between the 
clavulanic acid and amoxicillin peaks is satisfactory if it is not less 
than 3.5.
    (d) Coefficient of variation. The coefficient of variation (SR 
in percent) is satisfactory if it is not more than 2.0 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.351(b) of this chapter.
    (iv) Calculations. Calculate the milligrams of amoxicillin or 
clavulanic acid content per tablet as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.138

where:
Au=Response of the amoxicillin or clavulanic acid peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
As=Response of the amoxicillin or clavulanic acid peak in the 
          chromatogram of the amoxicillin or clavulanic acid working 
          standard;
Cs=Concentration of standards in milligrams of amoxicillin or 
          clavulanic acid per milliliter of the standard solution;
V=Volume of sample solution (milliliters); and
N=Number of tablets taken for assay.


[[Page 530]]


    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter. Dissolution rate is determined by dissolution of the 
amoxicillin component using the high-performance liquid chromatographic 
assay described in this section.
[49 FR 39672, Oct. 10, 1984, as amended at 50 FR 19919, May 13, 1985; 55 
FR 11582, Mar. 29, 1990]



Sec. 440.103e  Amoxicillin trihydrate and clavulanate potassium for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amoxicillin trihydrate and clavulanate 
potassium for oral suspension is a dry mixture of amoxicillin trihydrate 
and clavulanate potassium with one or more suitable and harmless 
colorings, flavorings, buffers, sweetening ingredients, preservatives, 
stabilizers, and suspending agents. When reconstituted as directed in 
the labeling, each milliliter contains either amoxicillin trihydrate 
equivalent to 25 milligrams of amoxicillin with clavulanate potassium 
equivalent to 6.25 clavulanic acid or amoxicillin trihydrate equivalent 
to 50 milligrams of amoxicillin with clavulanate potassium equivalent to 
12.5 milligrams of clavulanic acid. Its amoxicillin trihydrate content 
is satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of amoxicillin that it is 
represented to contain. Its clavulanate potassium content is 
satisfactory if it is not less than 90 percent and not more than 125 
percent of the number of milligrams of clavulanic acid that it is 
represented to contain. The moisture content of the dry powder is not 
more than 7.5 percent when the reconstituted solution is to contain 25 
milligrams of amoxicillin per milliliter and not more than 8.5 percent 
when the reconstituted solution is to contain 50 milligrams of 
amoxicillin per milliliter. When reconstituted as directed in the 
labeling, its pH is not less than 4.8 and not more than 6.6. The 
amoxicillin trihydrate used conforms to the standards prescribed by 
Sec. 440.3(a)(1). The clavulanate potassium conforms to the standards 
prescribed by Sec. 455.15(a)(1) of this chapter.
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``amoxicillin and 
clavulanate potassium for oral suspension''.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The amoxicillin trihydrate used in making the batch for potency, 
moisture, pH, amoxicillin content, concordance, crystallinity, and 
identity.
    (b) The clavulanate potassium used in making the batch for 
clavulanic acid content, moisture, pH, identity, and clavam-2-
carboxylate content.
    (c) The batch for amoxicillin content, clavulanic acid content, 
moisture, and pH.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The amoxicillin trihydrate used in making the batch: 12 
packages, each containing approximately 300 milligrams.
    (b) The clavulanate potassium used in making the batch: 12 packages, 
each containing approximately 300 milligrams.
    (c) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Amoxicillin content or 
clavulanic acid content. Proceed as directed in Sec. 436.351 of this 
chapter, using ambient temperature, an ultraviolet detection system 
operating at a wavelength between 220 and 230 nanometers, and a column 
packed with microparticulate (3 to 10 micrometers in diameter) reversed 
phase packing material such as octadecyl silane bonded silica. Reagents, 
working standard and sample solutions, system suitability requirments, 
and calculations for amoxicillin and clavulanic acid content are as 
follows:
    (i) Reagents--(a) 0.05M Sodium phosphate buffer solution, pH 4.4. 
Transfer 7.8 grams of monobasic sodium phosphate to a 1-liter volumetric 
flask and dissolve in 900 milliliters of distilled water. Adjust to pH 
4.4  0.1 with 18N

[[Page 531]]

phosphoric acid or 10N sodium hydroxide. Dilute to volume with distilled 
water. Mix well.
    (b) Mobile phase. Mix methanol:0.05M sodium phosphate buffer 
solution, pH 4.4 (5:95 v/v) and mix or ultrasonicate for no less than 2 
minutes. Degas by passing through a 0.5-micron filter with vacuum. The 
mobile phase may be sparged with helium through a 2-micrometer metal 
filter for the duration of the analysis. Adjust the ratio of methanol to 
aqueous buffer as necessary to obtain satisfactory retention of the 
peaks.
    (ii) Working standard and sample solutions--(a) Preparation of 
working standard solution. Accurately weigh and transfer into a 200-
milliliter volumetric flask approximately 100 milligrams of amoxicillin 
working standard and approximately 50 milligrams of the clavulanate 
working standard. Dissolve and dilute to volume with distilled water. 
Use within 8 hours after preparation.
    (b) Preparation of sample solution. Reconstitute the suspension as 
directed in the labeling. Immediately transfer an appropriate aliquot to 
a suitable volumetric flask to obtain an approximate amoxicillin 
concentration of 0.5 milligram per milliliter and dilute to volume with 
distilled water. Mix well for 10 minutes using a magnetic stirrer. 
Filter an aliquot through Whatman 42 or equivalent filter paper. 
Alternatively, a suitable membrane filter may be used. Samples should be 
prepared just prior to chromatographic injection. Inject the sample 
solution within 1 hour after the addition of water.
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.5.
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 550 theoretical plates.
    (c) Resolution factor. The resolution factor (R) between the 
clavulanic acid and amoxicillin peaks is satisfactory if it is not less 
than 3.5.
    (d) Coefficient of variation. The coefficient of variation (SR 
in percent) is satisfactory if it is not more than 2.0 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.351(b) of this chapter.
    (iv) Calculations. Calculate the quantity of amoxicillin or 
clavulanic acid content in milligrams per milliliter of the oral 
suspension as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.139

where:
Au=Response of the amoxicillin or clavulanic acid peaks in 
          the sample chromatogram;
As=Response of the amoxicillin or clavulanic acid peaks in 
          the standard chromatogram;
C=Concentration of the standard (milligrams per milliliter of 
          amoxicillin X potency of amoxicillin standard or milligrams 
          per milliliter of clavulanate X potency of clavulanate 
          standard); and
V=Dilution volume in milliliters.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the suspension reconstituted as directed in the labeling.
[49 FR 39673, Oct. 10, 1984, as amended at 50 FR 19919, May 13, 1985; 55 
FR 11582, Mar. 29, 1990]



Sec. 440.103f  Amoxicillin trihydrate-clavulanate potassium chewable tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amoxicillin trihydrate-clavulanate 
potassium chewable tablets are composed of amoxicillin trihydrate and 
clavulanate potassium with or without one or more suitable lubricants, 
diluents, flavorings, and binders. Each tablet contains amoxicillin 
trihydrate equivalent to either 125 or 250 milligrams of amoxicillin and 
clavulanate potassium equivalent to 31.25 or 62.5 milligrams of 
clavulanic acid. Its amoxicillin trihydrate content is satisfactory if 
it contains not less than 90 percent and not more than 120 percent of 
the number of milligrams of amoxicillin that it is represented to 
contain. Its clavulanate potassium content is satisfactory if it 
contains not less than 90 percent and not more than 120 percent

[[Page 532]]

of the number of milligrams of clavulanic acid that it is represented to 
contain. Its moisture content is not more than 6 percent. It passes the 
dissolution test if the quantity Q, of amoxicillin at 30 minutes, is 85 
percent or greater. The amoxicillin trihydrate conforms to the standards 
prescribed by Sec. 440.3(a)(1). The clavulanate potassium conforms to 
the standards prescribed by Sec. 455.15(a)(1) of this chapter.
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``amoxicillin-
clavulanate potassium chewable tablets''.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The amoxicillin trihydrate used in making the batch for potency, 
safety, moisture, pH, amoxicillin content, concordance, crystallinity, 
and identity.
    (b) The clavulanate potassium used in making the batch for 
clavulanic acid content, moisture, pH, identity, and clavam-2-
carboxylate content.
    (c) The batch for amoxicillin content, clavulanic acid content, 
moisture, and dissolution rate.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The amoxicillin trihydrate used in making the batch: 12 
packages, each containing approximately 300 milligrams.
    (b) The clavulanate potassium used in making the batch: 12 packages, 
each containing approximately 300 milligrams.
    (c) The batch: A minimum of 100 tablets.
    (b) Tests and methods of assay--(1) Amoxicillin and clavulanic acid 
contents. Proceed as directed in Sec. 436.351 of this chapter, using 
ambient temperature, an ultraviolet detection system operating at a 
wavelength between 220 and 230 nanometers, and a column packed with 
microparticulate (3 to 10 micrometers in diameter) reversed phase 
packing material such as octadecyl hydrocarbon bonded silicas. Reagents, 
working standard and sample solutions, system suitability requirements, 
and calculations for amoxicillin or clavulanic acid content are as 
follows:
    (i) Reagents--(a) 0.05M Sodium phosphate buffer solution, pH 4.4. 
Transfer 7.8 grams of sodium monobasic phosphate to a 1-liter volumetric 
flask and dissolve in 900 milliliters of distilled water. Adjust the pH 
to 4.40.1 with 18N phosphoric acid or 10N sodium hydroxide. 
Dilute to volume with distilled water. Mix well.
    (b) Mobile phase. Mix methanol: 0.05M sodium phosphate buffer 
solution, pH 4.4 (5:95 v/v) and ultrasonicate for no less than 2 
minutes. Degas by passing through a 0.5-micron filter with vacuum. The 
mobile phase may be sparged with the helium through a 2-micrometer metal 
filter for the duration of the analysis. Adjust the ratio of methanol to 
aqueous buffer as necessary to obtain satisfactory retention of the 
peaks.
    (ii) Working standard and sample solutions--(a) Preparation of 
working standard solution. Dissolve and dilute accurately weighed 
portions each of the amoxicillin trihydrate working standard and the 
clavulanate lithium working standard with water to obtain a solution 
containing 0.5 milligram of amoxicillin and 0.25 milligram of clavulanic 
acid per milliliter. Use within 1 hour after preparation or within 4 
hours if stored under refrigeration.
    (b) Preparation of sample solution. To obtain a concentration of 0.5 
milligram of amoxicillin per milliliter, dissolve a representative 
number of tablets in water with the aid of a magnetic stirrer or 
ultrasonication. Filter an aliquot through Whatman 42 filter paper or 
equivalent, discard the first 10 milliliters of filtrate, and use the 
remaining portion as the sample solution. Alternatively, a suitable 
membrane filter may be used. Prepare samples not more than 1 hour before 
the chromatographic injection.
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.5.
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,000 theoretical plates in a 30-
centimeter column for each active component.

[[Page 533]]

    (c) Resolution. The resolution (R) between the clavulanic acid and 
amoxicillin peaks is satisfactory if it is not less than 3.5.
    (d) Coefficient of variation. The coefficient of variation (SR 
in percent) of five replicate injections is satisfactory if it is not 
more than 2.0 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.351(b) of this chapter.
    (iv) Calculations. Calculate the milligrams of amoxicillin or 
clavulanic acid content per tablet as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.140

where
Au=Response of the amoxicillin or clavulanic acid peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
As=Response of the amoxicillin or clavulanic acid peak in the 
          chromatogram of the amoxicillin or clavulanic acid working 
          standard;
Cs=Concentration of standards in milligrams of amoxicillin or 
          clavulanic acid per milliliter of the standard solution;
V=Volume of sample solution (milliliters); and
N=Number of tablets taken for assay.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter. Dissolution rate is determined by dissolution of the 
amoxicillin component using the high-performance liquid chromatographic 
assay described in this section.
[50 FR 42933, Oct. 25, 1985; 50 FR 47367, Nov. 17, 1985, as amended at 
55 FR 11582, Mar. 29, 1990]



Sec. 440.105  Ampicillin oral dosage forms.



Sec. 440.105a  Ampicillin tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ampicillin tablets are composed of 
ampicillin with one or more suitable and harmless diluents and 
lubricants. Each tablet contains 250 or 500 milligrams of ampicillin. 
Its potency is satisfactory if it is not less than 90 percent and not 
more than 120 percent of the number of milligrams of ampicillin that it 
is represented to contain. Its loss on drying is not more than 4 
percent. The tablets disintegrate within 15 minutes. The ampicillin used 
conforms to the standards prescribed by Sec. 440.5(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The ampicillin used in making the batch for potency, loss on 
drying, pH, ampicillin content, concordance, crystallinity, and 
identity.
    (b) The batch for potency, loss on drying, and disintegration time.
    (ii) Samples required:
    (a) The ampicillin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar with sufficient 0.1M potassium phosphate buffer, pH 8.0 (solution 
3), to give a stock solution of convenient concentration. Blend for 3 to 
5 minutes. Remove an aliquot and further dilute with solution 3 to the 
reference concentration of 0.1 microgram of ampicillin per milliliter 
(estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution. Prepare the sample as 
follows: Place a representative number of tablets in a high-speed glass 
blender jar and add sufficient distilled water to give a convenient 
concentration. Blend for 3 to 5 minutes. Further dilute an aliquot with 
distilled water to the prescribed concentration.
    (2) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.

[[Page 534]]

    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure described in paragraph (e)(1) of that 
section.
[39 FR 18976, May 30, 1974, as amended at 49 FR 3458, Jan. 27, 1984; 50 
FR 19919, May 13, 1985]



Sec. 440.105b  Ampicillin chewable tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Each ampicillin chewable tablet contains 
125 milligrams or 250 milligrams of ampicillin with suitable binders, 
lubricants, flavorings, and colorings. Its potency is satisfactory if it 
is not less than 90 percent and not more than 120 percent of the number 
of milligrams of ampicillin that it is represented to contain. Its loss 
on drying is not more than 3 percent. The ampicillin used conforms to 
the standards prescribed by Sec. 440.5(a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``ampicillin 
tablets''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) The results of tests and assays on:
    (a) The ampicillin used in making the batch for potency, loss on 
drying, pH, ampicillin content, concordance, crystallinity, and 
identity.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:
    (a) The ampicillin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 tablets.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar with sufficient 0.1M potassium phosphate buffer, pH 8.0 (solution 
3), to give a stock solution of convenient concentration. Blend for 3 to 
5 minutes. Remove an aliquot and further dilute with solution 3 to the 
reference concentration of 0.1 microgram of ampicillin per milliliter 
(estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution. Prepare the sample as 
follows: Blend a representative number of tablets in a high-speed 
blender with sufficient distilled water to give a stock solution of 
convenient concentration. Blend for 3 to 5 minutes. Further dilute an 
aliquot of the stock solution with distilled water to the prescribed 
concentration.
    (2) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
[39 FR 18976, May 30, 1974, as amended at 43 FR 9800, Mar. 10, 1978; 49 
FR 3458, Jan. 27, 1984; 50 FR 19919, May 13, 1985]



Sec. 440.105c  Ampicillin capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ampicillin capsules are composed of 
ampicillin with or without one or more buffer substances, diluents, 
binders, lubricants, vegetable oils, colorings, and flavorings, enclosed 
in a gelatin capsule. Each capsule contains 125 milligrams, 250 
milligrams, or 500 milligrams of ampicillin. Its potency is satisfactory 
if it is not less than 90 percent and not more than 120 percent of the 
number of milligrams of ampicillin that it is represented to contain. 
The loss on drying is not more than 4.0 percent. The ampicillin used 
conforms to the standards prescribed by Sec. 440.5(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The ampicillin used in making the batch for potency, loss on 
drying, pH, ampicillin content, concordance, crystallinity, and 
identity.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:

[[Page 535]]

    (a) The ampicillin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
either of the following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar with sufficient 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to give a convenient concentration. Blend for 3 to 5 
minutes. Remove an aliquot and further dilute with solution 3 to the 
reference concentration of 0.1 microgram of ampicillin per milliliter 
(estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution. Prepare the sample as 
follows: Place the contents of a representative number of capsules into 
a high-speed glass blender jar and add sufficient distilled water to 
give a convenient concentration. Blend for 3 to 5 minutes. Filter 
through Whatman No. 2 filter paper. Further dilute an aliquot of the 
filtrate with distilled water to the prescribed concentration.
    (2) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
[39 FR 18976, May 30, 1974, as amended at 49 FR 3458, Jan. 27, 1984; 50 
FR 19919, May 13, 1985]



Sec. 440.105d  Ampicillin for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ampicillin for oral suspension is a 
mixture of ampicillin with one or more suitable and harmless colorings, 
flavorings, buffer substances, sweetening ingredients, and 
preservatives. When reconstituted as directed in the labeling, it 
contains either 25 milligrams, 50 milligrams, or 100 milligrams of 
ampicillin per milliliter. Its potency is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the number of 
milligrams of ampicillin that it is represented to contain. Its moisture 
content is not more than 2.5 percent. When reconstituted as directed in 
the labeling, its pH is not less than 5.0 and not more than 7.5. The 
ampicillin used conforms to the standards prescribed by 
Sec. 440.5(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The ampicillin used in making the batch for potency, loss on 
drying, pH, ampicillin content, concordance, crystallinity, and 
identity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The ampicillin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
either of the following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute the drug as directed in the labeling. Place an accurately 
measured representative portion of the sample into a suitable volumetric 
flask and dilute to volume with 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to give a convenient concentration. Mix well. Further 
dilute an aliquot with solution 3 to the reference concentration of 0.1 
microgram of ampicillin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution. Prepare the sample as

[[Page 536]]

follows: Reconstitute the drug as directed in the labeling. Place an 
accurately measured aliquot (usually a single dose) into an 
appropriately sized volumetric flask and dilute to volume with distilled 
water. Mix well. Further dilute with distilled water to the prescribed 
concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
[39 FR 18976, May 30, 1974, as amended at 49 FR 3458, Jan. 27, 1984; 50 
FR 19919, May 13, 1985]



Sec. 440.107  Ampicillin trihydrate oral dosage forms.



Sec. 440.107a  Ampicillin trihydrate chewable tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ampicillin trihydrate chewable tablets 
are composed of ampicillin trihydrate with or without one or more 
suitable diluents, lubricants, preservatives, and flavorings. Each 
tablet contains ampicillin trihydrate equivalent to 125 or 250 
milligrams of ampicillin. Its potency is satisfactory if it contains not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of ampicillin that it is represented to contain. Its moisture 
content is not more than 5.0 percent. The ampicillin trihydrate used 
conforms to the standards prescribed by Sec. 440.7(a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``ampicillin 
tablets.''
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The ampicillin trihydrate used in making the batch for potency, 
loss on drying, pH, ampicillin content, concordance, crystallinity, and 
identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The ampicillin trihydrate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 tablets.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar with sufficient 0.1M potassium phosphate buffer, pH 8.0 (solution 
3), to give a stock solution of convenient concentration. Blend for 3 to 
5 minutes. Remove an aliquot and further dilute with solution 3 to the 
reference concentration of 0.1 microgram of ampicillin per milliliter 
(estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution. Prepare the sample as 
follows: Place a representative number of tablets into a high-speed 
glass blender jar containing sufficient distilled water to give a 
convenient concentration. Blend for 5 minutes. Further dilute with 
distilled water to the prescribed concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 18976, May 30, 1974, as amended at 49 FR 3459, Jan. 27, 1984; 50 
FR 19919, May 13, 1985]



Sec. 440.107b  Ampicillin trihydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ampicillin trihydrate capsules are 
composed of ampicillin trihydrate with or without one or more buffer 
substances, diluents, binders, lubricants, vegetable oils, colorings, 
and flavorings enclosed in a gelatin capsule. Each capsule contains 
ampicillin trihydrate equivalent to 250 milligrams or 500 milligrams of 
ampicillin. Its potency is satisfactory if it contains not less than 90 
percent and not more than

[[Page 537]]

120 percent of the number of milligrams of ampicillin that it is 
represented to contain. Its loss on drying is not less than 10 percent 
and not more than 15 percent. The ampicillin trihydrate used conforms to 
the standards prescribed by Sec. 440.7(a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``ampicillin 
capsules''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The ampicillin trihydrate used in making the batch for potency, 
loss on drying, pH, ampicillin content, concordance, crystallinity, and 
identity.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:
    (a) The ampicillin trihydrate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
either of the following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar with sufficient 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to give a convenient concentration. Blend for 3 to 5 
minutes. Remove an aliquot and further dilute with solution 3 to the 
reference concentration of 0.1 microgram of ampicillin per milliliter 
(estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution. Prepare the sample as 
follows: Place the contents of a representative number of capsules into 
a high-speed glass blender jar and add sufficient distilled water to 
give a convenient concentration. Blend for 3 to 5 minutes. Filter 
through Whatman No. 2 filter paper. Further dilute an aliquot of the 
filtrate with distilled water to the prescribed concentration.
    (2) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
[39 FR 18976, May 30, 1974, as amended at 49 FR 3459, Jan. 27, 1984; 50 
FR 19919, May 13, 1985]



Sec. 440.107c  Ampicillin trihydrate for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ampicillin trihydrate for oral suspension 
is a mixture of ampicillin trihydrate with one or more suitable and 
harmless colorings, flavorings, buffers, sweetening ingredients, and 
preservatives. When reconstituted as directed in the labeling, it 
contains ampicillin trihydrate equivalent to either 25, 50, or 100 
milligrams of ampicillin per milliliter. Its potency is satisfactory if 
it is not less than 90 percent and not more than 120 percent of the 
number of milligrams of ampicillin that it is represented to contain. 
Its moisture content is:
    (i) Not more than 2.5 percent if it contains sugar and is intended 
to contain the equivalent of 25 or 50 milligrams of ampicillin per 
milliliter when reconstituted as directed in the labeling; or
    (ii) Not more than 5 percent if it contains sugar and is intended to 
contain the equivalent of 100 milligrams of ampicillin per milliliter 
when reconstituted as directed in the labeling; or


Its pH, when reconstituted as directed in the labeling, is not less than 
5.0 and is not more than 7.5. The ampicillin trihydrate used conforms to 
the standards prescribed by Sec. 440.7(a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``ampicillin for 
oral suspension.''
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The ampicillin trihydrate used in making the batch for potency, 
loss on

[[Page 538]]

drying, pH, ampicillin content, concordance, crystallinity, and 
identity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The ampicillin trihydrate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
either of the following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute the drug as directed in the labeling. Place an accurately 
measured representative portion of the sample into a suitable volumetric 
flask and dilute to volume with 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to give a convenient concentration. Mix well. Further 
dilute an aliquot with solution 3 to the reference concentration of 0.1 
microgram of ampicillin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution. Prepare the sample as 
follows: Reconstitute the drug as directed in the labeling. Place an 
accurately measured aliquot (usually a single dose) into an 
appropriately sized volumetric flask and dilute to volume with distilled 
water. Mix well. Further dilute with distilled water to the prescribed 
concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
[39 FR 18976, May 30, 1974, as amended at 49 FR 3459, Jan. 27, 1984; 49 
FR 5096, Feb. 10, 1984; 50 FR 19919, May 13, 1985]



Sec. 440.107d  Ampicillin trihydrate-probenecid for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ampicillin trihydrate and probenecid for 
oral suspension is a dry mixture of ampicillin trihydrate and probenecid 
with suitable flavorings, lubricants, colorings, and suspending agents 
packaged in a single-dose container. When reconstituted as directed in 
the labeling, each single dose will contain ampicillin trihydrate 
equivalent to 3.5 grams of ampicillin and 1.0 gram of probenecid. Its 
ampicillin content is satisfactory if it is not less than 90 percent and 
not more than 120 percent of the number of grams of ampicillin that it 
is represented to contain. Its probenecid content is satisfactory if it 
is not less than 90 percent and not more than 110 percent of the number 
of grams of probenecid that it is represented to contain. Its moisture 
content is not more than 5.0 percent. When reconstituted as directed in 
the labeling, its pH is not less than 5.0 and not more than 7.5. The 
ampicillin trihydrate used conforms to the standards prescribed by 
Sec. 440.7(a)(1). The probenecid used conforms to the standards 
prescribed by the U.S.P.
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``ampicillin-
probenecid for oral suspension''.
    (3) Requests for certification, samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The ampicillin trihydrate used in making the batch for potency, 
loss on drying, pH, ampicillin content, concordance, crystallinity, and 
identity.
    (b) The probenecid used in making the batch for all U.S.P. 
specifications.
    (c) The batch for ampicillin content, probenecid content, moisture, 
and pH.
    (ii) Samples required:
    (a) The ampicillin trihydrate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch: A minimum of 10 immediate containers.
    (b) Tests and methods of assay--(1) Ampicillin content--(i) Sample 
preparation. Reconstitute as directed in the labeling and mix well. 
Drain the suspension from the bottle for 30 seconds into a high-speed 
glass blender jar containing

[[Page 539]]

sufficient sterile distilled water to obtain a total volume of 500 
milliliters. Blend for 10 minutes.
    (ii) Assay procedures. Use any of the following methods; however, 
the results obtained from the microbiological agar diffusion assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the aqueous 
solution with 0.1M potassium phosphate buffer, pH 8.0 (solution 3), to 
the reference concentration of 0.1 microgram of ampicillin per 
milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution. Dilute an aliquot of the 
aqueous solution to the prescribed concentration.
    (2) Probenecid content--(i) Preparation of standard solution. 
Transfer approximately 25 milligrams of U.S.P. probenecid reference 
standard, accurately weighed, to a 25-milliliter volumetric flask. 
Dissolve and dilute to volume with 1 percent aqueous sodium carbonate 
solution.
    (ii) Preparation of sample solution. Reconstitute the sample as 
directed in the labeling and mix well. Drain the suspension from the 
bottle for 30 seconds into a 1,000-milliliter volumetric flask. Dilute 
to volume with 1 percent aqueous sodium carbonate solution, shake well, 
and filter through Whatman No. 6 filter paper. Discard the first 10-
milliliter portion.
    (iii) Procedure. Transfer 2.0 milliliters of the clear filtrate to a 
125-milliliter separatory funnel and add 8.0 milliliters of 1.0N 
hydrochloric acid. Extract the solution with four 20-milliliter portions 
of chloroform, filtering each extract through a glass wool pledget into 
a 100-milliliter volumetric flask. Wash the pledget with chloroform, 
dilute to volume with chloroform and mix. Treat 2.0 milliliters of the 
standard solution in the same manner. Using a suitable spectrophotometer 
equipped with a 1-centimeter cell and chloroform washed with 1 percent 
aqueous sodium carbonate solution as a blank, determine the absorbance 
of the sample and standard solutions at the peak near 257 nanometers.
    (iv) Calculations. Calculate the probenecid content as follows:

Grams probenecid per container=(Absorbance of sample x weight of 
          standard in milligrams x percent purity of standard)/
          (Absorbance of standard x 25 x 100)
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
[39 FR 18976, May 30, 1974, as amended at 40 FR 49083, Oct. 21, 1975; 49 
FR 3459, Jan. 27, 1984; 50 FR 19919, May 13, 1985]



Sec. 440.107e  Ampicillin trihydrate-probenecid capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ampicillin trihydrate-probenecid capsules 
are composed of ampicillin trihydrate and probenecid with or without one 
or more buffer substances, diluents, binders, lubricants, vegetable 
oils, colorings, and flavorings enclosed in a gelatin capsule. Each 
capsule contains ampicillin trihydrate equivalent to 389 milligrams of 
ampicillin and 111 milligrams of probenecid. Its ampicillin content is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of ampicillin that it is represented 
to contain. Its probenecid content is satisfactory if it is not less 
than 90 percent and not more than 110 percent of the number of 
milligrams of probenecid that it is represented to contain. Its loss on 
drying is not less than 8.5 percent and not more than 13.0 percent. The 
ampicillin trihydrate used conforms to the standards prescribed by 
Sec. 440.7(a)(1). The probenecid used conforms to the standards 
prescribed by the U.S.P.
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``ampicillin-
probenecid capsules''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:

[[Page 540]]

    (a) The ampicillin trihydrate used in making the batch for potency, 
loss on drying, pH, ampicillin content, concordance, crystallinity, and 
identity.
    (b) The probenecid used in making the batch for all U.S.P. 
specifications.
    (c) The batch for ampicillin content, probenecid content, and loss 
on drying.
    (ii) Samples required:
    (a) The ampicillin trihydrate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Ampicillin content. Use any of 
the following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar with sufficient 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to give a stock solution of convenient concentration. 
Blend for 8 to 10 minutes. Remove an aliquot and further dilute with 
solution 3 to the reference concentration of 0.1 microgram of ampicillin 
per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution. Prepare the sample as 
follows: Place the contents of a representative number of capsules into 
a high-speed glass blender jar with sufficient distilled water to give a 
convenient concentration. Blend for 8 to 10 minutes. Filter through 
Whatman No. 2 filter paper. Further dilute an aliquot of the filtrate 
with distilled water to the prescribed concentration.
    (2) Probenecid content--(i) Preparation of standard solution. 
Transfer approximately 25 milligrams of probenecid reference standard 
U.S.P., accurately weighed, to a 25-milliliter volumetric flask. 
Dissolve and dilute to volume with 1 percent aqueous sodium carbonate 
solution.
    (ii) Preparation of sample solution. Place the contents of a 
representative number of capsules into a high-speed glass blender jar 
with 100 milliliters of 1 percent aqueous sodium carbonate solution for 
each capsule. Blend for 8 to 10 minutes. Filter a portion through 
Whatman No. 2 filter paper, discarding the first 10-milliliter portion 
of the filtrate.
    (iii) Procedure. Transfer 2.0 milliliters of the clear filtrate to a 
125-milliliter separatory funnel and add 8.0 milliliters of 1.0N 
hydrochloric acid. Extract the solution with four 20-milliliter portions 
of chloroform, filtering each extract into a 100-milliliter volumetric 
flask through a glass wool pledget and 6 grams of chloroform-washed 
anhydrous sodium sulfate. Wash the pledget and sodium sulfate with 
chloroform, dilute to volume with chloroform and mix. Treat 2.0 
milliliters of the standard solution in the same manner. Using a 
suitable spectrophotometer equipped with a 1-centimeter cell and 
chloroform washed with 1 percent aqueous sodium carbonate solution as a 
blank, determine the absorbance of the sample and standard solutions at 
the peak near 257 nanometers.
    (iv) Calculations. Calculate the probenecid content as follows:

Milligrams probenecid per capsule = (Absorbance of sample  x  weight of 
          standard in milligrams  x  percent purity of standard)/
          (Absorbance of standard  x  25)

    (3) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
[40 FR 58288, Dec. 16, 1975, as amended at 45 FR 16474, Mar. 14, 1980; 
49 FR 3459, Jan. 27, 1984; 50 FR 19919, May 13, 1985]



Sec. 440.108  Bacampicillin hydrochloride dosage forms.



Sec. 440.108a  Bacampicillin hydrochloride tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacampicillin hydrochloride tablets are 
composed of bacampicillin hydrochloride with one or more suitable and 
harmless diluents and lubricants. Each tablet contains bacampicillin 
hydrochloride equivalent to either 280 or 560 milligrams of ampicillin. 
Its potency is satisfactory if it is not less than 90 percent and not 
more

[[Page 541]]

than 125 percent of the number of milligrams of ampicillin that it is 
represented to contain. Its moisture content is not more than 2.5 
percent. It passes the dissolution test. The bacampicillin hydrochloride 
used conforms to the standards prescribed by Sec. 440.8(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacampicillin hydrochloride used in making the batch for 
potency, moisture, pH, and identity.
    (b) The batch for potency, moisture, and dissolution.
    (ii) Samples required:
    (a) The bacampicillin hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 100 tablets.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the iodometric 
assay shall be conclusive.
    (i) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 440.8(b)(1)(i) of this chapter, except prepare the sample solution 
and calculate the potency of the sample as follows:
    (a) Preparation of sample solution. Place one tablet into a high-
speed glass blender jar with sufficient distilled water to obtain a 
concentration of 1.25 milligrams of ampicillin per milliliter 
(estimated). Blend for 3 to 5 minutes. Filter before using.
    (b) Calculations. Calculate the ampicillin content in milligrams per 
tablet as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.043

where:
Au=Absorbance of sample solution;
Pa=Potency of working standard in micrograms per milliliter;
As=Absorbance of working standard solution;
d=Dilution factor of the sample.

    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except use the ampicillin working standard. Prepare the sample 
as follows: Dissolve and dilute a representative number of tablets with 
distilled water to the prescribed concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter, except in lieu of paragraph (d) of that section use the 
interpretation described in the United States Pharmacopeia XX 
dissolution test. The quantity, Q (the amount of ampicillin dissolved) 
is 85 percent at 30 minutes.
[46 FR 25604, May 8, 1981. Redesignated at 47 FR 23711, June 1, 1982, 
and amended at 48 FR 51293, 51294, Nov. 8, 1983; 50 FR 19919, May 13, 
1985]



Sec. 440.108b  Bacampicillin hydrochloride for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacampicillin hydrochloride for oral 
suspension is a mixture of bacampicillin hydrochloride with one or more 
suitable and harmless buffers, diluents, sweetening ingredients, 
suspending agents, flavorings, and colorings. When reconstituted as 
directed in the labeling, it contains bacampicillin hydrochloride 
equivalent to 17.5 milligrams of ampicillin per milliliter. Its potency 
is satisfactory if it is not less than 90 percent and not more than 125 
percent of the number of milligrams of ampicillin that it is represented 
to contain. Its loss on drying is not more than 2.0 percent. When 
reconstituted as directed in the labeling, its pH is not less than 6.5 
and not more than 8.0. It gives a positive identity test for 
bacampicillin hydrochloride. The bacampicillin hydrochloride conforms to 
the standards prescribed by Sec. 440.8(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:

[[Page 542]]

    (a) The bacampicillin used in making the batch for potency, 
moisture, pH, and identity.
    (b) The batch for potency, loss on drying, pH, and identity.
    (ii) Samples required:
    (a) The bacampicillin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.204 of this chapter, except:
    (i) Use the ampicillin working standard as the standard of 
comparison;
    (ii) Use 4.0 milliliters of sample solution in lieu of the 2.0 
milliliters specified in paragraph (c)(1) of that section; and
    (iii) Calculate the potency of the sample as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.141
    

Prepare the sample as follows: Reconstitute the drug as directed in the 
labeling. Place an accurately measured portion equivalent to one dose 
into a 250-milliliter volumetric flask. Add 200 milliliters of a solvent 
mixture of 95 percent ethanol and 0.1M phosphoric acid (8:2). Shake for 
30 minutes on a wrist action shaker and dilute to volume with the 
solvent mixture. Centrifuge a portion of the sample solution for 10 
minutes at 6,000 rpm. Use the clear supernatant without further 
dilution.
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
    (4) Identity. Proceed as directed in Sec. 436.330 of this chapter, 
except prepare the sample as follows: Reconstitute as directed in the 
labeling. Place 8.0 milliliters of the sample into a 100-milliliter 
volumetric flask, add 70 milliliters of 95 percent ethyl alcohol and 
shake for 30 minutes. Dilute to volume with 95 percent ethyl alcohol.
[47 FR 23711, June 1, 1982, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.111  Carbenicillin indanyl sodium tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Carbenicillin indanyl sodium tablets are 
composed of carbenicillin indanyl sodium and one or more suitable and 
harmless diluents, binders, lubricants, colorings, and coating 
substances. Each tablet contains carbenicillin indanyl sodium equivalent 
to 382 milligrams of carbenicillin. Its potency is satisfactory if it 
contains not less than 90 percent and not more than 120 percent of the 
number of milligrams of carbenicillin that it is represented to contain. 
Its moisture content is not more than 2.0 percent. It gives a positive 
identity test for carbenicillin indanyl sodium. The tablets shall 
disintegrate within 1 hour. The carbenicillin indanyl sodium used 
conforms to the standards prescribed by Sec. 440.11(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The carbenicillin indanyl sodium used in making the batch for 
potency, moisture, pH, and identity.
    (b) The batch for potency, moisture, identity, and disintegration 
time.
    (ii) Samples required:
    (a) The carbenicillin indanyl sodium used in making the batch: Five 
packages, each containing approximately 1 gram and one package 
containing approximately 2.5 grams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.300 of this chapter, except:
    (i) Preparation of the sample. Accurately weigh 20 tablets and 
determine the average tablet weight. Using a mortar and pestle, grind 
the tablets to a fine powder. Accurately weigh a portion of the powder 
approximately equivalent to the weight of one tablet and transfer it 
into a 100-milliliter volumetric flask. Add approximately 70 milliliters 
of distilled water and shake

[[Page 543]]

the flask for 5 minutes. Dilute to volume and mix well. Transfer a 5-
milliliter aliquot of the stock solution to a 50-milliliter glass-
stoppered centrifuge tube. (The solution will be slightly turbid.) Add 
15 milliliters of phosphate-citrate buffer and 20 milliliters of 4-
methyl-2-pentanone to the tube. Stopper the tube and shake it for 10 
seconds. Centrifuge at 2,000 revolutions per minute to separate the 
phases. Remove about 15 milliliters of the upper phase and proceed as 
directed in Sec. 436.300(e) of this chapter.
    (ii) Calculations. Calculate the carbenicillin content (potency) of 
the tablets as follows:

Milligrams of carbenicillin per tablet=(Degrees of rotation of sample 
          solution  x  weight of working standard  x  average tablet 
          weight  x  100  x  micrograms of carbenicillin in each 
          milligram of the working standard)/(Degrees of rotation of 
          working standard x weight of sample  x  25  x  1,000)

where:

100 and 25=The volume of the sample and working standard solutions, 
respectively;
1,000=Factor to correct micrograms to milligrams.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Identity. Proceed as directed in Sec. 436.301 of this chapter, 
preparing the sample as follows: Using a mortar and pestle, grind a 
representative number of tablets into a fine powder. Dissolve a weighed 
amount of this powder in sufficient extraction solvent (described in 
Sec. 436.301(b)(1) of this chapter) to give 10 milligrams of 
carbenicillin per milliliter. Shake the mixture for 5 minutes and 
promptly dilute an aliquot in extraction solvent to obtain a final 
concentration of 1 milligram carbenicillin per milliliter.
    (4) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure described in paragraph (e)(2) of that 
section.
[39 FR 18976, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.115  Cloxacillin sodium monohydrate oral dosage forms.



Sec. 440.115a  Cloxacillin sodium monohydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cloxacillin sodium monohydrate capsules 
are composed of cloxacillin sodium and one or more suitable and harmless 
diluents and lubricants. Each capsule contains cloxacillin sodium 
monohydrate equivalent to 125 milligrams, 250 milligrams, or 500 
milligrams of cloxacillin. Its potency is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the number of 
milligrams of cloxacillin that it is represented to contain. Its 
moisture content is not more than 5 percent. The cloxacillin sodium 
monohydrate used conforms to the standards prescribed by 
Sec. 440.15(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``cloxacillin sodium 
capsules''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this subchapter, each such 
request shall contain:
    (i) Results of tests and assays on:
    (a) The cloxacillin sodium monohydrate used in making the batch for 
potency, moisture, pH, cloxacillin content, identity, and crystallinity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The cloxacillin sodium monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 1 percent potassium phosphate buffer, 
pH 6.0 (solution 1), to give a stock solution of convenient 
concentration. Blend for 3 to 5 minutes.

[[Page 544]]

    (ii) Assay procedure. Use either of the following methods; however, 
the results obtained from the microbiological agar diffusion assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 5 micrograms of 
cloxacillin per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
subchapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this 
subchapter.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59861, Nov. 22, 1977; 50 
FR 19919, May 13, 1985]



Sec. 440.115b  Cloxacillin sodium monohydrate for oral solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cloxacillin sodium monohydrate for oral 
solution is a mixture of sodium cloxacillin with one or more suitable 
and harmless colorings, flavorings, buffer substances, and 
preservatives. When reconstituted as directed in the labeling, each 
milliliter contains the equivalent of 25 milligrams or 50 milligrams of 
cloxacillin. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
cloxacillin that it is represented to contain. Its moisture content is 
not more than 1 percent. When reconstituted as directed in its labeling, 
its pH is not less than 5.0 nor more than 7.5. The cloxacillin sodium 
monohydrate used conforms to the standards prescribed by 
Sec. 440.15(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``cloxacillin sodium for 
oral solution''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this subchapter, each such 
request shall contain:
    (i) Results of tests and assays on:
    (a) The cloxacillin sodium monohydrate used in making the batch for 
potency, moisture, pH, cloxacillin content, identity, and crystallinity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The cloxacillin sodium monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Reconstitute the sample as directed in the labeling. Dilute an 
accurately measured representative aliquot of the sample with sufficient 
1 percent potassium phosphate buffer, pH 6.0 (solution 1), to give a 
stock solution of convenient concentration.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the microbiological agar diffusion assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 5 micrograms of 
cloxacillin per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this 
subchapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using the drug reconstituted as directed in its labeling.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59861, Nov. 22, 1977; 43 
FR 9800, Mar. 10, 1978; 50 FR 19919, May 13, 1985]



Sec. 440.117  Cyclacillin oral dosage forms.



Sec. 440.117a  Cyclacillin tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cyclacillin tablets are composed of 
cyclacillin with one or more suitable and harmless diluents, lubricants, 
colorings, and disintegrants. Each tablet contains 250 or 500 milligrams 
of cyclacillin. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of

[[Page 545]]

the number of milligrams of cyclacillin that it is represented to 
contain. Its moisture content is not more than 5 percent. The tablets 
disintegrate within 15 minutes. It gives a positive identity test for 
cyclacillin. The cyclacillin used conforms to the standards prescribed 
by Sec. 440.17(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cyclacillin used in making the batch for potency, moisture, 
pH, cyclacillin content, concordance, crystallinity, and identity.
    (b) The batch for potency, moisture, disintegration time, and 
identity.
    (ii) Samples required:
    (a) The cyclacillin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the iodometric 
assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar with sufficient 0.1M potassium phosphate buffer, pH 8.0 (solution 
3), to give a stock solution of convenient concentration. Blend for 3 to 
5 minutes. Remove an aliquot and further dilute with solution 3 to the 
reference concentration of 1.0 microgram of cyclacillin per milliliter 
(estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, preparing the sample solution as follows: Place a 
representative number of tablets in a high-speed glass blender jar and 
add sufficient distilled water to give a convenient concentration. Blend 
for 3 to 5 minutes. Further dilute an aliquot with distilled water to 
the prescribed concentration.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, except prepare the working 
standard and sample solutions and calculate the potency of the sample as 
follows:
    (a) Preparation of working standard solution. Dissolve and dilute an 
accurately weighed portion of the cyclacillin working standard in 
sufficient distilled water to obtain a concentration of 1.25 milligrams 
of cyclacillin per milliliter.
    (b) Preparation of sample solution. Place one tablet into a high-
speed glass blender jar and add sufficient distilled water to obtain a 
concentration of 1.25 milligrams of cyclacillin per milliliter. Blend 
for 3 to 5 minutes. Filter, if necessary.
    (c) Calculations. Calculate the cyclacillin content in milligrams 
per tablet as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.044

where:
Au=Absorbance of sample solution;
Pa=Potency of working standard in micrograms per milliliter;
As=Absorbance of working standard solution;
d=Dilution factor of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure in paragraph (e)(1) of that section, except 
do not use discs.
    (4) Identity. Proceed as directed in Sec. 436.327 of this chapter, 
preparing the sample as follows: Dissolve a representative portion of 
finely powdered tablets with sufficient 0.1N sodium hydroxide to obtain 
a solution containing 1 milligram of cyclacillin per milliliter. Allow 
the sample solution to stand for 15 minutes before using.
[46 FR 2985, Jan. 13, 1981; 46 FR 15880, Mar. 10, 1981, as amended at 50 
FR 19919, May 13, 1985]



Sec. 440.117b  Cyclacillin for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cyclacillin for oral suspension is a 
mixture of cyclacillin with one or more suitable and harmless colorings, 
flavorings, buffer substances, sweetening ingredients, preservatives,

[[Page 546]]

and suspending agents. When reconstituted as directed in the labeling, 
it contains either 25 milligrams, 50 milligrams, or 100 milligrams of 
cyclacillin per milliliter. Its potency is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of cyclacillin that it is represented to contain. Its 
moisture content is not more than 1.5 percent. When reconstituted as 
directed in the labeling, its pH is not less than 4.5 and not more than 
6.5. It gives a positive identity test for cyclacillin. The cyclacillin 
used conforms to the standards prescribed by Sec. 440.17(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cyclacillin used in making the batch for potency, moisture, 
pH, cyclacillin content, concordance, crystallinity, and identity.
    (b) The batch for potency, moisture, pH, and identity.
    (ii) Samples required:
    (a) The cyclacillin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch: A minimum of seven immediate containers.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
any of the following methods; however, the results obtained from the 
iodometric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute the drug as directed in the labeling. Place an accurately 
measured representative portion of the sample into a suitable volumetric 
flask and dilute to volume with 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to give a convenient concentration. Mix well. Further 
dilute an aliquot with solution 3 to the reference concentration of 1.0 
microgram of cyclacillin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, preparing the sample as follows: Reconstitute the drug as 
directed in the labeling. Place an accurately measured representative 
portion of the sample into an appropriate-sized volumetric flask and 
dilute to volume with 1 percent potassium phosphate buffer, pH 6.0 
(solution 1). Mix well. Further dilute with solution 1 to the prescribed 
concentration.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, except prepare the working 
standard and sample solutions and calculate the potency of the sample as 
follows:
    (a) Preparation of working standard solution. Dissolve and dilute an 
accurately weighed portion of the cyclacillin working standard in 
sufficient distilled water to obtain a concentration of 1.25 milligrams 
of cyclacillin per milliliter.
    (b) Preparation of sample solution. Reconstitute the sample as 
directed in the labeling. Place an accurately measured aliquot of the 
sample into an appropriate-sized volumetric flask and dilute to volume 
with distilled water to yield a concentration of 1.25 milligrams of 
cyclacillin per milliliter. Mix well. Filter, if necessary.
    (c) Calculations. Calculate the cyclacillin content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.142
    
where:
Au=Absorbance of sample solution;
Pa=Potency of working standard in micrograms per milliliter;
As=Absorbance of working standard solution;
d=Dilution factor of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
    (4) Identity. Proceed as directed in Sec. 436.327 of this chapter, 
preparing the sample as follows: Dilute an accurately measured 
representative portion of the reconstituted suspension with 0.1N sodium 
hydroxide to obtain a solution containing 1 milligram of cyclacillin

[[Page 547]]

per milliliter. Allow the sample solution to stand 45 minutes before 
using.
[46 FR 2985, Jan. 13, 1981; 46 FR 15880, Mar. 10, 1981, as amended at 50 
FR 19919, May 13, 1985]



Sec. 440.119  Dicloxacillin sodium monohydrate oral dosage forms.



Sec. 440.119a  Dicloxacillin sodium monohydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Dicloxacillin sodium monohydrate capsules 
are composed of dicloxacillin sodium monohydrate and one or more 
suitable diluents and lubricants. Each capsule contains dicloxacillin 
sodium monohydrate equivalent to 62.5, 125, 250, or 500 milligrams of 
dicloxacillin. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
dicloxacillin that it is represented to contain. The moisture content is 
not more than 5 percent. The dicloxacillin sodium monohydrate conforms 
to the requirements of Sec. 440.19(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``dicloxacillin sodium 
capsules''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The dicloxacillin sodium monohydrate used in making the batch 
for potency, moisture, pH, organic chlorine content, free chloride 
content, crystallinity, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The dicloxacillin sodium monohydrate used in making the batch: 
10 containers, each containing not less than 500 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 1 percent potassium phosphate buffer, 
pH 6.0 (solution 1), to give a stock solution of convenient 
concentration. Blend for 3 to 5 minutes. Remove an aliquot and further 
dilute with solution 1 to the reference concentration of 5.0 micrograms 
of dicloxacillin per milliliter (estimated) for the microbiological agar 
diffusion assay and to the prescribed concentration for the iodometric 
assay.
    (ii) Assay procedure. Assay for potency by either of the following 
methods; however, the results obtained from the microbiological agar 
diffusion assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter.
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59861, Nov. 22, 1977; 43 
FR 2393, Jan. 17, 1978; 44 FR 10379, Feb. 20, 1979; 50 FR 19919, May 13, 
1985]



Sec. 440.119b  Dicloxacillin sodium monohydrate for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Dicloxacillin sodium monohydrate for oral 
suspension is a mixture of dicloxacillin sodium monohydrate with one or 
more suitable colorings, flavorings, buffer substances, and 
preservatives. When reconstituted as directed in the labeling, it 
contains the equivalent of 12.5 or 25 milligrams of dicloxacillin per 
milliliter. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
dicloxacillin that it is represented to contain. Its moisture content is 
not more than 2 percent. The pH of the suspension, when reconstituted as 
directed in the labeling, is not less than 4.5 nor more than 7.5. The 
dicloxacillin sodium monohydrate used conforms to the requirements of 
Sec. 440.19(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``dicloxacillin sodium for 
oral suspension''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assay on:

[[Page 548]]

    (a) The dicloxacillin sodium monohydrate used in making the batch 
for potency, moisture, pH, organic chlorine content, free chloride 
content, crystallinity, and identity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The dicloxacillin sodium monohydrate used in making the batch: 
10 containers, each containing not less than 500 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Reconstitute the sample as directed in the labeling. Place an accurately 
measured aliquot of the sample containing an estimated 125 milligrams of 
dicloxacillin into a 100-milliliter volumetric flask. Add 20 milliliters 
of dimethylformamide and shake mechanically for 30 minutes. Dilute to 
volume with 1 percent potassium phosphate buffer, pH 6.0 (solution 1). 
The addition of dimethylformamide may be omitted if complete solution 
can be obtained with solution 1. Further dilute an aliquot with 
sufficient solution 1 to the reference concentration of 5.0 micrograms 
of dicloxacillin per milliliter (estimated) for the microbiological agar 
diffusion assay and to the prescribed concentration for the iodometric 
assay.
    (ii) Assay procedure. Assay for potency by either of the following 
methods; however, the results obtained from the microbiological agar 
diffusion assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter.
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59861, Nov. 22, 1977; 50 
FR 19919, May 13, 1985]



Sec. 440.125  Hetacillin oral dosage forms.



Sec. 440.125a  Hetacillin chewable tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Each hetacillin chewable tablet contains 
an amount of hetacillin equivalent to 112.5 milligrams of ampicillin 
with suitable buffers, preservatives, binders, flavorings, colorings, 
and sweetening ingredients. Its potency is satisfactory if it contains 
not less than 90 percent and not more than 120 percent of the number of 
milligrams of ampicillin that it is represented to contain. The moisture 
content is not more than 2.0 percent. The hetacillin used conforms to 
the requirements of Sec. 440.25(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The hetacillin used in making the batch for potency, moisture, 
pH, hetacillin content, identity, and crystallinity.
    (b) The batch for potency and moisture.
    (ii) Samples required.
    (a) The hetacillin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed for 
ampicillin in Sec. 436.105 of this chapter, using the ampicillin working 
standard as the standard of comparison and preparing the sample for 
assay as follows: Place a representative number of tablets in a high-
speed glass blender with sufficient 0.1M potassium phosphate buffer, pH 
8.0 (solution 3), to give a stock solution of convenient concentration. 
Blend for 3 to 5 minutes. Further dilute an aliquot of the stock 
solution with solution 3 to the reference concentration of 0.1 microgram 
of ampicillin per milliliter (estimated).

[[Page 549]]

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 18976, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.125b  Hetacillin for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Hetacillin for oral suspension is a 
mixture of hetacillin with one or more suitable preservatives, 
suspending agents, sweetening ingredients, flavorings, and colorings. 
When reconstituted as directed in the labeling, it contains the 
equivalent of 22.5, 45, or 112.5 milligrams of ampicillin per 
milliliter. Its potency is satisfactory if it contains not less than 90 
percent and not more than 120 percent of the number of milligrams of 
ampicillin that it is represented to contain. Its moisture content is 
not more than 2.0 percent. The pH of the suspension, when reconstituted 
as directed in its labeling, is not less than 2.0 and not more than 5.0. 
The hetacillin used conforms to the requirements of Sec. 440.25(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The hetacillin used in making the batch for potency, moisture, 
pH, hetacillin content, identity, and crystallinity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The hetacillin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed for 
ampicillin in Sec. 436.105 of this chapter, preparing the sample for 
assay as follows: Reconstitute the sample as directed in the labeling. 
Remove an accurately measured representative portion with a suitable 
syringe and hypodermic needle and place into a suitable volumetric 
flask. Dilute to volume with 0.1M potassium phosphate buffer, pH 8.0 
(solution 3). Further dilute an aliquot with solution 3 to the reference 
concentration of 0.1 microgram of ampicillin per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the sample after reconstituting as directed in the labeling.
[39 FR 18976, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.129  Hetacillin potassium capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Hetacillin potassium capsules are 
composed of potassium hetacillin with or without one or more suitable 
diluents, lubricants, and drying agents. Each capsule contains an amount 
of potassium hetacillin equivalent to 112.5, 225, or 450 milligrams of 
ampicillin. Its potency is satisfactory if it contains not less than 90 
percent and not more than 120 percent of the number of milligrams of 
ampicillin that it is represented to contain. The moisture content is 
not more than 3 percent. The potassium hetacillin used conforms to the 
requirements of Sec. 440.29(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The hetacillin potassium used in making the batch for potency, 
moisture, pH, hetacillin content, identity, and crystallinity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The hetacillin potassium used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed for 
ampicillin in Sec. 436.105 of this chapter, using the ampicillin working 
standard as the standard of comparison and preparing

[[Page 550]]

the sample for assay as follows: Place a representative number of 
capsules in a high-speed glass blender with sufficient 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), to give a stock solution of 
convenient concentration. Blend for 3 to 5 minutes. Further dilute an 
aliquot of the stock solution with solution 3 to the reference 
concentration of 0.1 microgram of ampicillin per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59861, Nov. 22, 1977; 50 
FR 19919, May 13, 1985]



Sec. 440.141  Nafcillin sodium monohydrate oral dosage forms.



Sec. 440.141a  Nafcillin sodium monohydrate tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nafcillin sodium monohydrate tablets are 
composed of nafcillin sodium monohydrate with one or more suitable 
buffers, binders, disintegrants, diluents, and lubricants. Each tablet 
contains nafcillin sodium monohydrate equivalent to 500 milligrams of 
nafcillin. Its potency is satisfactory if it is not less than 90 percent 
and not more than 120 percent of the number of milligrams of nafcillin 
that it is represented to contain. Its moisture content is not more than 
5 percent. It shall disintegrate within 20 minutes. The nafcillin sodium 
monohydrate used conforms to the standards prescribed by 
Sec. 440.41(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``nafcillin sodium 
tablets''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The nafcillin sodium monohydrate used in making the batch for 
potency, moisture, pH, crystallinity, nafcillin content, and identity.
    (b) The batch for potency, moisture, and disintegration time.
    (ii) Samples required:
    (a) The nafcillin sodium monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to give a stock solution of convenient concentration. 
Blend for 3 to 5 minutes.
    (ii) Assay procedures. Assay for potency by any of the following 
methods; however, the results obtained from the microbiological agar 
diffusion assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 2.0 micrograms of 
nafcillin per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (c) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the method described in paragraph (e)(1) of that section, 
except use distilled water in lieu of simulated gastric fluid as the 
immersion fluid.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59862, Nov. 22, 1977; 50 
FR 19919, May 13, 1985]



Sec. 440.141b  Nafcillin sodium monohydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nafcillin sodium monohydrate capsules are 
composed of nafcillin sodium monohydrate and one or more suitable and 
harmless buffer substances and lubricants. Each capsule contains 
nafcillin sodium monohydrate equivalent to 250 milligrams of nafcillin. 
The potency is satisfactory if it is not less than 90 percent and not 
more than 120 percent of the number of milligrams of nafcillin

[[Page 551]]

that it is represented to contain. The moisture content is not more than 
5.0 percent. The nafcillin sodium monohydrate conforms to the standards 
prescribed by Sec. 440.41(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``nafcillin sodium 
capsules''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The nafcillin sodium monohydrate used in making the batch for 
potency, moisture, pH, crystallinity, nafcillin content, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The nafcillin sodium monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 1 percent potassium phosphate buffer, 
pH 6.0 (solution 1), to give a stock solution of convenient 
concentration. Blend for 3 to 5 minutes. Remove an aliquot and further 
dilute with solution 1 to the reference concentration of 2.0 micrograms 
of nafcillin per milliliter (estimated) for the microbiological agar 
diffusion assay and to the prescribed concentration for the iodometric 
assay.
    (ii) Assay procedures. Assay for potency by either of the following 
methods; however, the results obtained from the microbiological agar 
diffusion assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter.
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59862, Nov. 22, 1977; 50 
FR 19919, May 13, 1985]



Sec. 440.141c  Nafcillin sodium monohydrate for oral solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nafcillin sodium monohydrate for oral 
solution is a packaged combination of one immediate container of 
nafcillin sodium monohydrate and one immediate container of an aqueous 
diluent containing one or more suitable and harmless colorings, 
flavoring, buffers, dispersants, diluents, and preservatives. When 
reconstituted as directed in the labeling, each milliliter contains the 
equivalent of 50 milligrams of nafcillin. Its potency is satisfactory if 
it is not less than 90 percent and not more than 120 percent of the 
number of milligrams of nafcillin that it is represented to contain. Its 
moisture content is not more than 5 percent. When reconstituted as 
directed in the labeling, its pH is not less than 5.5 and not more than 
7.5. The nafcillin sodium monohydrate used conforms to the standards 
prescribed by Sec. 440.41(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``nafcillin sodium for oral 
solution''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The nafcillin sodium monohydrate used in making the batch for 
potency, moisture, pH, crystallinity, nafcillin content, and identity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The nafcillin sodium monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Reconstitute as directed in the labeling. Place an accurately measured 
representative aliquot of the sample into a 250-milliliter volumetric 
flask and dilute to volume with 1 percent potassium phosphate buffer, pH 
6.0 (solution 1). Mix well. Further dilute an aliquot with solution 1 to 
the reference concentration

[[Page 552]]

of 2.0 micrograms of nafcillin per milliliter (estimated) for the 
microbiological agar diffusion assay and to the prescribed concentration 
for the iodometric assay.
    (ii) Assay procedures. Assay for potency by either of the following 
methods; however, the results obtained from the microbiological agar 
diffusion assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter.
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59862, Nov. 22, 1977; 50 
FR 19919, May 13, 1985]



Sec. 440.149  Oxacillin sodium monohydrate oral dosage forms.



Sec. 440.149a  Oxacillin sodium monohydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxacillin sodium monohydrate capsules are 
composed of oxacillin sodium monohydrate with or without one or more 
diluents and lubricants, enclosed in a gelatin capsule. Each capsule 
contains oxacillin sodium monohydrate equivalent to 125, 250, or 500 
milligrams of oxacillin. Its potency is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the number of 
milligrams of oxacillin that it is represented to contain. Its moisture 
content is not more than 6.0 percent. The oxacillin sodium monohydrate 
used conforms to the standards prescribed by Sec. 440.49(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``oxacillin sodium 
capsules''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oxacillin sodium monohydrate used in making the batch for 
potency, moisture, pH, oxacillin content, crystallinity, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The oxacillin sodium monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 1 percent potassium phosphate buffer, 
pH 6.0 (solution 1), to give a stock solution of convenient 
concentration. Blend for 3 to 5 minutes.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the microbiological agar diffusion assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 5 micrograms of 
oxacillin per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59862, Nov. 22, 1977; 50 
FR 19919, May 13, 1985]



Sec. 440.149b  Oxacillin sodium monohydrate for oral solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxacillin sodium monohydrate for oral 
solution is a mixture of oxacillin sodium monohydrate with one or more 
suitable colorings, flavorings, buffer substances, stabilizers, and 
preservatives. When reconstituted as directed in the labeling, each 
milliliter contains the equivalent of either 25 or 50 milligrams of 
oxacillin. Its potency is satisfactory if it is not less than 90 percent 
and not more than 120 percent of the number of milligrams of

[[Page 553]]

oxacillin that it is represented to contain. Its moisture content is not 
more than 1.0 percent. When reconstituted as directed in its labeling, 
the pH of the solution is not less than 5.0 and not more than 7.5. The 
oxacillin sodium monohydrate used conforms to the standards prescribed 
by Sec. 440.49(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``oxacillin sodium for oral 
solution''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oxacillin sodium monohydrate used in making the batch for 
potency, moisture, pH, oxacillin content, crystallinity, and identity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The oxacillin sodium monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Reconstitute as directed in the labeling. Place an accurately measured 
representative aliquot of the sample into an appropriate-sized 
volumetric flask with sufficient 1 percent potassium phosphate buffer, 
pH 6.0 (solution 1), to give a stock solution of convenient 
concentration.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the microbiological agar diffusion assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 5 micrograms of 
oxacillin per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter using 
the drug reconstituted as directed in the labeling.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59862, Nov. 22, 1977; 50 
FR 19919, May 13, 1985]



Sec. 440.155  Penicillin G benzathine oral dosage forms.



Secs. 440.155a--440.155b  [Reserved]



Sec. 440.155c  Penicillin G benzathine oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G benzathine oral suspension 
contains penicillin G benzathine with one or more suitable dispersing 
agents, buffer substances, preservatives, colorings, and flavorings. 
Each milliliter contains penicillin G benzathine equivalent to 30,000 
units or 60,000 units of penicillin G. Its potency is satisfactory if it 
is not less than 90 percent and not more than 120 percent of the number 
of units of penicillin G that it is represented to contain. Its pH is 
not less than 6.0 and not more than 7.0. The penicillin G benzathine 
used conforms to the standards prescribed by Sec. 440.55a(a)(1), except 
sterility and pyrogens.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G benzathine used in making the batch for 
potency, moisture, pH, penicillin G content, and crystallinity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The penicillin G benzathine used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the iodometric 
assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this

[[Page 554]]

chapter, preparing the sample for assay as follows: Dissolve an 
accurately measured representative volume of the sample in sufficient 
absolute methyl alcohol to give a solution of convenient concentration. 
Immediately further dilute with 1 percent potassium phosphate buffer, pH 
6.0 (solution 1), to the reference concentration of 1.0 unit of 
penicillin G per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, using a representative aliquot of the drug prepared for assay 
as described in paragraph (b)(2) of that section.
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[42 FR 59862, Nov. 22, 1977, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.155d  Penicillin G benzathine tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G benzathine tablets contain 
penicillin G benzathine with one or more suitable and harmless diluents, 
binders, lubricants, colorings, and flavorings. Each tablet contains 
penicillin G benzathine equivalent to 200,000 units of penicillin G. Its 
potency is satisfactory if it is not less than 90 percent and not more 
than 120 percent of the number of units of penicillin G that it is 
represented to contain. Its moisture content is not more than 8.0 
percent. The tablets shall disintegrate within 1 hour. The penicillin G 
benzathine used conforms to the standards prescribed by 
Sec. 440.55a(a)(1), except sterility and pyrogens.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G benzathine used in making the batch for 
potency, moisture, pH, penicillin G content, and crystallinity.
    (b) The batch for potency, moisture, and disintegration time.
    (ii) Samples required:
    (a) The penicillin G benzathine used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Using the penicillin G 
working standard as the standard of comparison, assay for potency by 
either of the following methods; however, the results obtained from the 
iodometric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar containing 200 milliliters of absolute methyl alcohol. Blend for 1 
minute. Add an additional 300 milliliters of absolute methyl alcohol and 
blend again for 2 to 3 minutes. Immediately further dilute an aliquot 
with 1 percent potassium phosphate buffer, pH 6.0 (solution 1), to the 
reference concentration of 1.0 unit of penicillin G per milliliter 
(estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, preparing the sample as follows: Weigh and finely powder six 
tablets. Transfer two accurately weighed portions of the tablets, each 
equivalent to 200,000 units of penicillin G, to two separate 100-
milliliter volumetric flasks. Dilute one flask, which is to be used as 
the blank, to volume with 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), and proceed as directed in Sec. 436.204(d) of this 
chapter. In lieu of directions in Sec. 436.204(e) (1), (2), and (3), to 
the other flask add 10 milliliters of 1.0N NaOH and mix well. Allow to 
stand for 15 minutes, then add 10 milliliters of 1.2N HC1, and dilute to 
volume with distilled water. Pipette a 2.0-milliliter aliquot into a 
125-milliliter glass-stoppered Erlenmeyer flask and proceed as directed 
in Sec. 436.204(c)(4) of this chapter.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter.
[42 FR 59862, Nov. 22, 1977, as amended at 50 FR 19919, May 13, 1985]

[[Page 555]]



Sec. 440.171  Penicillin V oral dosage forms.



Sec. 440.171a  Penicillin V capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin V capsules are composed of 
penicillin V with one or more suitable and harmless lubricants. Each 
capsule contains either 125 milligrams (200,000 units) or 250 milligrams 
(400,000 units) of penicillin V. Its potency is satisfactory if it is 
not less than 90 percent and not more than 120 percent of the number of 
milligrams or units of penicillin V that it is represented to contain. 
Its moisture content is not more than 2 percent. The penicillin V used 
conforms to the standards prescribed by Sec. 440.71(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin V used in making the batch for potency, moisture, 
pH, penicillin V content, and crystallinity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The penicillin V used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient absolute methyl alcohol to give a 
solution of convenient concentration. Blend for 3 to 5 minutes.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the iodometric assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter. Immediately dilute an aliquot of the 
methyl alcohol solution with 1 percent potassium phosphate buffer, pH 
6.0 (solution 1), to the reference concentration of 1.0 unit of 
penicillin V per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the methyl alcohol with solution 1 to 
the prescribed concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[42 FR 59863, Nov. 22, 1977, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.171b  Penicillin V for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin V for oral suspension is 
composed of penicillin V with or without one or more suitable and 
harmless suspending agents, colorings, flavorings, and buffer 
substances. When reconstituted as directed in the labeling, each 
millilter contains 25 milligrams (40,000 units), 50 milligrams (80,000 
units) or 208.3 milligrams (333,333 units) of penicillin V. Its potency 
is satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams or units of penicillin V that it is 
represented to contain. Its moisture content is not more than 1 percent. 
When reconstituted as directed in the labeling, its pH is not less than 
2.0 and not more than 4.0. The penicillin V used conforms to the 
standards prescribed by Sec. 440.71(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin V used in making the batch for potency, moisture, 
pH, penicillin V content, and crystallinity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The penicillin V used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the iodometric 
assay shall be conclusive.

[[Page 556]]

    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dissolve an accurately 
measured representative volume of the sample in sufficient absolute 
methyl alcohol to give a solution of convenient concentration. 
Immediately further dilute an aliquot with 1 percent potassium phosphate 
buffer, pH 6.0 (solution 1), to the reference concentration of 1.0 unit 
of penicillin V (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, preparing the sample as follows: Reconstitute as directed in 
the labeling. Dissolve an accurately measured representative portion of 
the sample in absolute methyl alcohol and dilute with 1.0 percent 
potassium phosphate buffer, pH 6.0 (solution 1) to the prescribed 
concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the sample reconstituted as directed in the labeling.
[42 FR 59863, Nov. 22, 1977, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.171c  Penicillin V tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin V tablets are composed of 
penicillin V with or without one or more suitable and harmless diluents, 
binders, lubricants, and colorings. Each tablet contains 125 milligrams 
(200,000 units), 300 milligrams (500,000 units), or 500 milligrams 
(800,000 units) of penicillin V. Its potency is satisfactory if it 
contains not less than 90 percent and not more than 120 percent of the 
number of milligrams or units of penicillin V that it is represented to 
contain. Its moisture content is not more than 3 percent. It shall 
disintegrate within 1 hour. The penicillin V used conforms to the 
standards prescribed by Sec. 440.71(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin V used in making the batch for potency, moisture, 
pH, penicillin V content, and crystallinity.
    (b) The batch for potency, moisture, and disintegration time.
    (ii) Samples required:
    (a) The penicillin V used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient absolute methyl alcohol to give a stock 
solution of convenient concentration. Blend for 2 to 5 minutes.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the iodometric assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter. Immediately dilute an aliquot of the 
methyl alcohol solution with 1 percent potassium phosphate buffer, pH 
6.0 (solution 1), to the reference concentration of 1.0 unit of 
penicillin V per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the methyl alcohol solution with 
solution 1 to the prescribed concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter.
[42 FR 59864, Nov. 22, 1977, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.173  Penicillin V potassium oral dosage forms.



Sec. 440.173a  Penicillin V potassium capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin V potassium capsules are 
composed of penicillin V potassium and one or more suitable lubricants 
and fillers. Each capsule contains penicillin V potassium equivalent to 
250 milligrams (400,000 units) or 500 milligrams (800,000 units) of 
penicillin V. The potency is satisfactory if it is not less than 90 
percent and more than 115

[[Page 557]]

percent of the number of milligrams or units of penicillin V that it is 
represented to contain. Its loss on drying is not more than 2.0 percent. 
The penicillin V potassium used conforms to the standards prescribed by 
Sec. 440.73(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin V potassium used in making the batch for potency, 
loss on drying, pH, crystallinity, penicillin V content.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:
    (a) The penicillin V potassium used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 1 percent potassium phosphate buffer, 
pH 6.0 (solution 1), to give a stock solution of convenient 
concentration. Blend for 3 to 5 minutes.
    (ii) Assay procedures. Using the penicillin V working standard as 
the standard of comparison, assay by either of the following methods; 
however, the results obtained from the iodometric assay shall be 
conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 unit of penicillin 
V per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[42 FR 59864, Nov. 22, 1977, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.173b  Penicillin V potassium chewable tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin V potassium chewable tablets 
are composed of penicillin V potassium with suitable diluents, binders, 
buffers, colorings, and flavorings. Each tablet contains penicillin V 
potassium equivalent to 125 milligrams (200,000 units) or 250 milligrams 
(400,000 units) of penicillin V. Its potency is satisfactory if it 
contains not less than 90 percent and not more than 125 percent of the 
number of milligrams or units of penicillin V that it is represented to 
contain. The loss on drying is not more than 1.5 percent. The penicillin 
V potassium used conforms to the standards prescribed by Sec. 440.73(a) 
(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``penicillin V 
potassium tablets''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin V potassium used in making the batch for potency, 
loss on drying, pH, penicillin V content, and crystallinity.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:
    (a) The penicillin V potassium used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 tablets.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to give a stock solution of convenient concentration. 
Blend for 3 to 5 minutes.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the iodometric assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the

[[Page 558]]

reference concentration of 1.0 unit of penicillin V per milliliter 
(estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[42 FR 59864, Nov. 22, 1977, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.173c  Penicillin V potassium tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin V potassium tablets are 
composed of penicillin V potassium with or without one or more suitable 
and harmless buffer substances, diluents, binders, lubricants, 
colorings, and flavorings. Each tablet contains penicillin V potassium 
equivalent to 125 milligrams (200,000 units), 250 milligrams (400,000 
units), or 500 milligrams (800,000 units) of penicillin V. Its potency 
is satisfactory if it contains not less than 90 percent and not more 
than 120 percent of the number of milligrams or units of penicillin V 
that it is represented to contain. Its loss on drying is not more than 
1.5 percent. It shall disintegrate within 1 hour. The penicillin V 
potassium used conforms to the standards prescribed by 
Sec. 440.73(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin V potassium used in making the batch for potency, 
loss on drying, pH, penicillin V content and crystallinity.
    (b) The batch for potency, loss on drying, and disintegration time.
    (ii) Samples required:
    (a) The penicillin V potassium used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to give a stock solution of convenient concentration. 
Blend for 3 to 5 minutes.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the iodometric assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 unit of penicillin 
V per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescibed concentration.
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter.
[42 FR 59865, Nov. 22, 1977, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.173d  Penicillin V potassium for oral solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin V potassium for oral solution 
is composed of penicillin V potassium with or without one or more 
suitable and harmless suspending agents, colorings, flavorings, buffer 
substances, and preservatives. When reconstituted as directed in the 
labeling, each milliliter contains penicillin V potassium equivalent to 
either 25 milligrams (40,000 units) or 50 milligrams (80,000 units) of 
penicillin V. Its potency is satisfactory if it contains not less than 
90 percent and not more than 135 percent of the number of milligrams or 
units of penicillin V that it is represented to contain. Its moisture 
content is not more than 1 percent. When reconstituted as directed in 
the labeling, its pH is not less than 5.0 and not more than 7.5. The 
penicillin V potassium used conforms to the standards prescribed by 
Sec. 440.73(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 559]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such requests 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin V potassium used in making the batch for potency, 
loss on drying, pH, penicillin V content, and crystallinity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The penicillin V potassium used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Reconstitute as directed in the labeling. Dilute an accurately measured 
representative portion of the suspension with 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to give a stock solution of 
convenient concentration.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the iodometric assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 unit of penicillin 
V per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the sample when reconstituted as directed in the labeling.
[42 FR 59865, Nov. 22, 1977, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.180  Penicillin G potassium oral dosage forms.



Sec. 440.180a  Penicillin G potassium tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin potassium tablets are composed 
of penicillin G potassium with or without one or more suitable and 
harmless buffer substances, diluents, binders, lubricants, colorings, 
and flavorings. Each tablet contains penicillin G potassium equivalent 
to 100,000 units, 200,000 units, 250,000 units, 400,000 units, 500,000 
units, 800,000 units or 1,000,000 units of penicillin G. Its potency is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of units of penicillin G that it is represented to 
contain. Its loss on drying is not more than 1 percent. The tablets 
shall disintegrate within 1 hour. The penicillin G potassium used 
conforms to the standards prescribed by Sec. 440.80a(a)(1), except 
sterility and pyrogens.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G potassium used in making the batch for potency, 
loss on drying, pH, penicillin G content, and crystallinity.
    (b) The batch:
    (1) If the person who requests certification is the manufacturer of 
the batch: Potency, loss on drying, and disintegration time of tablets 
collected during the time of tableting the batch; and, unless the 
tablets are packaged into dispensing-size containers immediately after 
they are compressed or the manufacturer has submitted to the 
Commissioner, and it has been accepted, information adequate to prove 
that such tests are not necessary, loss on drying of the tablets 
collected during each day of packaging the batch.
    (2) If the person who requests certification is not the manufacturer 
of the batch: Potency, loss on drying, and disintegration time of 
tablets collected during each day the tablets are being packaged into 
dispensing-size containers.
    (ii) Samples required:
    (a) The penicillin G potassium used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:

[[Page 560]]

    (1) If the person who requests certification is the manufacturer of 
the batch: A minimum of 36 tablets. If, after tableting, such person 
packaged the batch into dispensing-size containers: 20 tablets, 
collected at equal intervals during each day the tablets are being 
packaged, except that this sample is not required if the tablets are 
packaged immediately after they are compressed or if the manufacturer 
has been exempted by the Commissioner from such requirement.
    (2) If the person who requests certification is not the manufacturer 
of the batch (for the purposes of certification, a batch shall be that 
number of tablets filled by such person into dispensing-size containers 
during each day's packaging operations): A minimum of 36 tablets 
collected by taking single tablets at such intervals throughout each day 
of packaging the tablets so that the quantities packaged during the 
intervals are approximately equal.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to give a stock solution of convenient concentration. 
Blend for 3 to 5 minutes.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the iodometric assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 unit of penicillin 
G per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter.
[42 FR 59865, Nov. 22, 1977; 43 FR 3705, Jan. 27, 1978, as amended at 50 
FR 19919, May 13, 1985]



Sec. 440.180c  Penicillin G potassium capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G potassium capsules are 
composed of penicillin G potassium and a suitable and harmless diluent. 
Each capsule contains penicillin G potassium equivalent to 250,000 units 
or 400,000 units of penicillin G. Its potency is satisfactory if it is 
not less than 90 percent and not more than 120 percent of the number of 
units of penicillin G that it is represented to contain. Its loss on 
drying is not more than 1.5 percent. The penicillin G potassium used 
conforms to the standards prescribed by Sec. 440.80a(a)(1), except 
sterility and pyrogens.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G potassium used in making the batch for potency, 
loss on drying, pH, penicillin G content, and crystallinity.
    (b) The batch:
    (1) If the person who requests certification is the manufacturer of 
the batch: Potency and loss on drying of capsules collected during the 
time of encapsulating the batch; and, unless the capsules are packaged 
into dispensing-size containers immediately after they are encapsulated 
or the manufacturer has submitted to the Commissioner, and it has been 
accepted, information adequate to prove that such tests are not 
necessary, loss on drying of capsules collected during each day of 
packaging the batch.
    (2) If the person who requests certification is not the manufacturer 
of the batch: Potency and loss on drying of capsules collected during 
each day the capsules are being packaged into dispensing-size 
containers.
    (ii) Samples required:
    (a) The penicillin G potassium used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) If the person who requests certification is the manufacturer of 
the batch: A minimum of 30 capsules. If after encapsulating, such person 
packaged the batch into dispensing-size

[[Page 561]]

containers: 20 capsules collected at equal intervals during each day the 
capsules are being packaged, except that this sample is not required if 
the capsules are packaged immediately after they are filled or if the 
manufacturer has been exempted by the Commissioner from such 
requirement.
    (2) If the person who requests certification is not the manufacturer 
of the batch (for the purposes of certification, a batch shall be that 
number of capsules filed by such person into dispensing-size containers 
during each day's packaging operations): A minimum of 30 capsules 
collected by taking single capsules at such intervals throughout each 
day of packaging the capsules that the quantities packaged during the 
intervals are approximately equal.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 1 percent potassium phosphate buffer, 
pH 6.0 (solution 1), to give a stock solution of convenient 
concentration. Blend for 3 to 5 minutes.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the iodometric assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 unit of penicillin 
G per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[42 FR 59866, Nov. 22, 1977, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.180f  Penicillin G potassium for oral solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G potassium for oral solution 
contains penicillin G potassium and one or more suitable and harmless 
buffers, colorings, flavorings, diluents, and preservatives. Each 
milliliter contains penicillin G potassium equivalent to 20,000 units, 
25,000 units, 40,000 units, 50,000 units, 80,000 units, or 100,000 units 
of penicillin G. Its potency is satisfactory if it is not less than 90 
percent and not more than 130 percent of the number of units of 
penicillin G that it is represented to contain. Its moisture content is 
not more than 1 percent. When reconstituted as directed in the labeling, 
its pH is not less than 5.5 and not more than 7.5. The penicillin G 
potassium used conforms to the standards prescribed by 
Sec. 440.80a(a)(1), except sterility and pyrogens.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G potassium used in making the batch for potency, 
loss on drying, pH, penicillin G content, and crystallinity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The penicillin G potassium used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Reconstitute as directed in the labeling. Transfer an accurately 
measured representative portion into a suitable volumetric flask and 
dilute to volume with 1 percent potassium phosphate buffer, pH 6.0 
(solution 1).
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the iodometric assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 unit of penicillin 
G per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.

[[Page 562]]

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution obtained after reconstituting the drug as directed in the 
labeling.
[42 FR 59866, Nov. 22, 1977, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.180g  Penicillin G potassium tablets for solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G potassium tablets for 
solution are composed of penicillin G potassium. Each tablet contains 
penicillin G potassium equivalent to 100,000 units, 200,000 units, or 
250,000 units of penicillin G. The potency is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
units of penicillin G that it is represented to contain. Its loss on 
drying is not more than 1 percent. The penicillin G potassium used 
conforms to the standards prescribed by Sec. 440.80a(a)(1), except 
sterility and pyrogens.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G potassium used in making the batch for potency, 
loss on drying, pH, penicillin G content, and crystallinity.
    (b) The batch:
    (1) If the person who requests certification is the manufacturer of 
the batch: Potency and loss on drying of tablets collected during the 
time of tableting the batch; and, unless the tablets are packaged into 
dispensing-size containers immediately after they are compressed, or the 
manufacturer has submitted to the Commissioner, and it has been 
accepted, information adequate to prove that such tests are not 
necessary, loss on drying of the tablets collected during each day of 
packaging the batch.
    (2) If the person who requests certification is not the manufacturer 
of the batch: Potency and loss on drying of the tablets collected during 
each day the tablets are being packaged into dispensing-size containers.
    (ii) Samples required:
    (a) The penicillin G potassium used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) If the person who requests certification is the manufacturer of 
the batch: A minimum of 30 tablets. If, after tableting, such person 
packaged the batch into dispensing-size containers: 20 tablets collected 
at equal intervals during each day the tablets are packaged, except that 
this sample is not required if the tablets are packaged immediately 
after they are compressed or if the manufacturer has been exempted by 
the Commissioner from such requirement.
    (2) If the person who requests certification is not the manufacturer 
of the batch (for the purposes of certification, a batch shall be that 
number of tablets filed by such person into dispensing-size containers 
during each day's packaging operations): A minimum of 30 tablets 
collected by taking single tablets at such intervals throughout each day 
of packaging the tablets that the quantities packaged during the 
intervals are approximately equal.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to give a stock solution of convenient concentration. 
Blend for 3 to 5 minutes.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the iodometric assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 unit of penicillin 
G per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.

[[Page 563]]

    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[42 FR 59867, Nov. 22, 1977, as amended at 50 FR 19919, May 13, 1985]



                   Subpart C--Injectable Dosage Forms



Sec. 440.201  Sterile azlocillin sodium.

    The requirements for certification and the tests and methods of 
assay for sterile azlocillin sodium packaged for dispensing are 
described in Sec. 440.1a.
[47 FR 53349, Nov. 26, 1982]



Sec. 440.202  Sterile amdinocillin.

    The requirements for certification and the tests and methods of 
assay for sterile amdinocillin packaged for dispensing are described in 
Sec. 440.2a.
[50 FR 7766, Feb. 26, 1985]



Sec. 440.207  Sterile ampicillin trihydrate for suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile ampicillin trihydrate for 
suspension is a dry mixture of ampicillin trihydrate and one or more 
suitable and harmless buffer substances, stabilizers, suspending agents, 
and preservatives. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
ampicillin that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its loss on drying is not less than 11.4 percent and not 
more than 14.0 percent. When reconstituted as directed in the labeling, 
its pH is not less than 5.0 and not more than 7.0. The ampicillin 
trihydrate used conforms to the standards prescribed by 
Sec. 440.7a(a)(1) of this chapter.
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``sterile 
ampicillin for suspension.''
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The ampicillin trihydrate used in making the batch for potency, 
loss on drying, pH, ampicillin content, concordance, crystallinity, and 
identity.
    (b) The batch for potency, sterility, pyrogens, loss on drying, and 
pH.
    (ii) Samples required:
    (a) The ampicillin trihydrate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 12 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Reconstitute as directed in the labeling. Using a suitable hypodermic 
needle and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container, or, if the labeling specifies 
the amount of potency in a given volume of the resultant preparation, 
remove an accurately measured representative portion from each 
container. Dilute the resultant solution with 0.1M potassium phosphate 
buffer, pH 8.0 (solution 3), for the microbiological agar diffusion 
assay, or distilled water for the iodometric assay, to give a stock 
solution of convenient concentration.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the microbiological agar diffusion assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 3 to the reference concentration of 0.1 microgram of 
ampicillin per milliliter.
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, except in paragraph (d) of that section, add 3 drops of 1.2N 
hydrochloric acid to both the sample and working standard solutions 
after the addition of 0.01N iodine solution. Dilute an aliquot of the 
stock solution with distilled water to the prescribed concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
in lieu of (e)(1)(i)(a), prepare the sample for test as follows: From 
each of 10 immediate containers,

[[Page 564]]

aseptically transfer approximately 300 milligrams of sample into a 
sterile 500-milliliter Erlenmeyer flask containing approximately 400 
milliliters of diluting fluid D. Add at least 200,000 Levy units 1 
of penicillinase. Repeat the process using 10 additional containers. 
Swirl both of the stoppered flasks to completely solubilize the 
suspension prior to filtration and proceed as directed in paragraph 
(e)(1)(ii) of that section. If the formulation cannot be filtered, 
proceed as directed in Sec. 436.20(e)(2) of this chapter, except use 
medium B in lieu of medium A.
---------------------------------------------------------------------------

    1 One Levy unit of penicillinase inactivates 59.3 units 
of pencillin G in 1 hour at 25 deg. C. and at a pH of 7.0 in a phosphate 
buffered solution of a pure alkali salt of penicillin G when the 
substrate is in sufficient concentration to maintain a zero order 
reaction.
---------------------------------------------------------------------------

    (3) Pyrogens. Proceed as directed in Sec. 436.32(f) of this chapter, 
using a solution containing 20 milligrams of ampicillin per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution obtained when the product is reconstituted as directed in 
the labeling.
[39 FR 18976, May 30, 1974, as amended at 49 FR 3459, Jan. 27, 1984; 50 
FR 19918, 19919, May 13, 1985]



Sec. 440.209  Ampicillin sodium injectable dosage forms.



Sec. 440.209a  Sterile ampicillin sodium.

    The requirements for certification and the tests and methods of 
assay for sterile ampicillin sodium packaged for dispensing are 
described in Sec. 440.9a.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59867, Nov. 22, 1977. 
Redesignated at 52 FR 42288, Nov. 4, 1987]



Sec. 440.209b  Sterile ampicillin sodium and sulbactam sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ampicillin sodium and sulbactam sodium is 
a dry mixture of ampicillin sodium and sulbactam sodium in which the 
ratio of ampicillin to sulbactam is 2:1. Its ampicillin potency is not 
less than 563 micrograms of ampicillin per milligram on an anhydrous 
basis. It contains not less than 280 micrograms of sulbactam per 
milligram on an anhydrous basis. Its ampicillin sodium content is 
satisfactory if it contains not less than 90 percent and not more than 
115 percent of the number of milligrams of ampicillin that it is 
represented to contain. Its sulbactam sodium content is satisfactory if 
it contains not less than 90 percent and not more than 115 percent of 
the number of milligrams of sulbactam that it is represented to contain. 
It is sterile. It is nonpyrogenic. Its moisture content is not more than 
2.0 percent. The pH of an aqueous solution containing 10 milligrams of 
ampicillin and 5 milligrams of sulbactam per milliliter is not less than 
8.0 and not more than 10.0. It passes the identity test for ampicillin 
and sulbactam. The ampicillin sodium content conforms to the standards 
prescribed by Sec. 440.9a(a)(1) of this chapter. The sulbactam content 
conforms to the standards prescribed by Sec. 455.82a(a)(1) of this 
chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (A) The ampicillin sodium used in making the batch for potency, 
sterility, pyrogens, moisture, pH, crystallinity, and identity.
    (B) The sulbactam sodium used in making the batch for potency, 
sterility, pyrogens, moisture, crystallinity, and identity.
    (C) The batch for ampicillin potency, sulbactam potency, sterility, 
pyrogens, moisture, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The ampicillin sodium used in making the batch: 12 packages, 
each containing approximately 300 milligrams.
    (B) The sulbactam sodium used in making the batch: 12 packages, each 
containing approximately 300 milligrams.
    (C) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.

[[Page 565]]

    (2) For sterility testing: A minimum of 20 immediate containers 
collected at regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Ampicillin and sulbactam 
content. Proceed as directed in Sec. 436.216 of this chapter, operating 
isothermally at 25  deg.C, using an ultraviolet detection system 
operating at a wavelength of 230 nanometers, a column packed with 
microparticulate (3 to 10 micrometers in diameter) reversed phase 
packing material such as octadecyl hydrocarbon bonded silica, a flow 
rate of 2.0 milliliters per minute, and a known injection volume of 10 
microliters. Reagents, working standard and sample solutions, system 
suitability requirements, and calculations are as follows:
    (i) Reagents--(A) 1.0M Phosphoric acid. Prepare by diluting 67.5 
milliliters of reagent grade phosphoric acid (85 percent) in distilled 
water to 1 liter.
    (B) 0.005M Tetrabutylammonium hydroxide. Dilute 6.6 milliliters of 
tetrabutylammonium hydroxide (40 percent) to 1,800 milliliters with 
distilled water. Adjust the pH to 5.0 with 1.0M phosphoric acid and 
dilute with distill water to 2 liters.
    (C) Mobile phase. Mix 350 milliliters of acetonitrile with 1,650 
milliliters of 0.005M tetrabutylammonium hydroxide. Filter and degas the 
mobile phase just prior to its introduction into the chromatograph 
pumping system. (Slight adjustments in pH and/or acetonitrile content 
may be made to achieve the system suitability parameters defined in 
paragraph (b)(1)(iii) of this section.)
    (ii) Preparation of working standard and sample solutions--(A) 
Working standard solution. Accurately weigh a portion of the ampicillin 
working standard containing the equivalence of approximately 75 
milligrams of ampicillin activity and transfer into a 25-milliliter 
volumetric flask. Accurately weigh a portion of the sulbactam working 
standard containing 35 milligrams of sulbactam and transfer into the 25-
milliliter volumetric flask containing the ampicillin. Dissolve and 
dilute to volume with mobile phase. Further dilute 5 milliliters to 25 
milliliters with mobile phase.
    (B) Sample solution. Dissolve an accurately weighed sample in 
sufficient mobile phase to give a stock solution containing 1 milligram 
of sample per milliliter (estimated); and, also, if it is packaged for 
dispensing, reconstitute as directed in the labeling. Then using a 
suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is represented as a single-dose container, or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute with mobile phase to yield a 
solution containing about 0.30 milligram sulbactam and about 0.60 
milligram ampicillin per milliliter.
    (iii) System suitability requirements--(A) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.5 at 10 
percent of peak height in lieu of 5 percent of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 3,500 theoretical plates for 
sulbactam for a 30-centimeter column.
    (C) Resolution. Dissolve 17.5 milligrams of sulbactam in 50 
milliliters of 0.01N sodium hydroxide and let stand for 30 minutes. 
Adjust the pH of the solution to 5.0 with concentrated phosphoric acid. 
Transfer a 5-milliliter aliquot of the resulting solution to a 25-
milliliter volumetric flask, add 4.25 milliliters of acetonitrile, and 
dilute to volume with 0.005M tetrabutylammonium hydroxide as described 
in paragraph (b)(1)(i)(B) of this section. Transfer 2 milliliters of 
this solution to a 50-milliliter flask, add 30 milligrams of ampicillin 
potency, dissolve and dilute to volume with mobile phase. Use this 
solution to determine the resolution factor. The resolution (R) between 
the peaks for ampicillin and sulbactam alkaline degradation product is 
satisfactory if it is not less than 1.2.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent) of 5 replicate 
injections is satisfactory if it is not more than 2.0 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.216(b) of this chapter.

[[Page 566]]

Alternate chromatographic conditions are acceptable provided 
reproducibility and resolution are comparable to the system. However, 
the sample preparation described in paragraph (b)(1)(ii)(b) of this 
section should not be changed.
    (iv) Calculations. (A) Calculate the micrograms of ampicillin or 
sulbactam per milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.143

where:
Au=Area of the ampicillin or sulbactam peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
As=Area of the ampicillin or sulbactam peak in the 
          chromatogram of the ampicillin or sulbactam working standard;
Ps=Ampicillin or sulbactam activity in the ampicillin-
          sulbactam working standard solution in micrograms per 
          milliliter;
Cu=Milligrams of sample per milliliter of sample solution; 
          and
m=Percent moisture content of the sample.

    (B) Calculate the ampicillin or sulbactam content of the container 
as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.144

where:
Au=Area of the ampicillin or sulbactam peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
As=Area of the ampicillin or sulbactam peak in the 
          chromatogram of the ampicillin or sulbactam working standard;
Ps=Ampicillin or sulbactam activity in the ampicillin-
          sulbactam working standard solution in micrograms per 
          milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 20 milligrams of sulbactam and 40 milligrams 
of ampicillin per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams of ampicillin and 5 
milligrams of sulbactam per milliliter.
    (6) Identity. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the ampicillin-sulbactam working standard.
[52 FR 42288, Nov. 4, 1987, as amended at 54 FR 47205, Feb. 20, 1989; 55 
FR 11582, Mar. 29, 1990]



Sec. 440.210  Benzylpenicilloyl-polylysine injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Benzylpenicilloyl-polylysine injection is 
an aqueous solution of benzylpenicilloyl-polylysine. It contains one or 
more suitable and harmless buffers. Its benzylpenicilloyl content is 
satisfactory if it is not less than 5.4 x 10- 5M and 
not more than 7.0 x 10- 5M, except that for the 
issuance of a certificate for a batch, the benzylpenicilloyl content 
must be not less than 6.4 x 10- 5M. It is sterile. It 
is nonpyrogenic. Its pH is not less than 6.5 and not more than 8.5. The 
benzylpenicilloyl-polylysine concentrate used conforms to the standards 
prescribed by Sec. 440.10(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The benzylpenicilloyl-polylysine concentrate used in making the 
batch for percent benzylpenicilloyl substitution, benzylpenicilloyl 
content, penamaldate content, penicillenate content, and pH.
    (b) The batch for benzylpenicilloyl content, sterility, pyrogens, 
and pH.
    (ii) Samples required:
    (a) The benzylpenicilloyl-polylysine concentrate used in making the 
batch: 2 vials, each containing not less than 5 milliliters.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 60 immediate 
containers.

[[Page 567]]

    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Benzylpenicilloyl content. 
Proceed as directed in Sec. 440.10(b)(1)(ii) except in lieu of 
Sec. 440.10(b)(1)(ii)(b) prepare the sample solution as follows: Pool 
contents of 16 immediate containers. Dilute a 3.0-milliliter aliquot to 
10 milliliters with saline phosphate buffer, pH 7.6.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
preparing the sample solution as follows: Pool the contents of at least 
8 vials to obtain a minimum of 1.5 milliliters of the original 
preparation. Dilute the 1.5 milliliters to 50 milliliters with diluent 
2.
    (4) [Reserved]
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[39 FR 35347, Oct. 1, 1974, as amended at 42 FR 14094, Mar. 15, 1977; 50 
FR 19918, 19919, May 13, 1985]



Sec. 440.213  Sterile carbenicillin disodium.

    The requirements for certification and the tests and methods of 
assay for sterile carbenicillin disodium packaged for dispensing are 
described in Sec. 440.13a.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59867, Nov. 22, 1977]



Sec. 440.219  Dicloxacillin sodium monohydrate injectable dosage forms.



Sec. 440.219a  Sterile dicloxacillin sodium monohydrate.

    The requirements for certification and the tests and methods of 
assay for sterile dicloxacillin sodium monohydrate packaged for 
dispensing are described in Sec. 440.19a.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59867, Nov. 22, 1977]



Sec. 440.219b  Dicloxacillin sodium monohydrate for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Dicloxacillin sodium monohydrate for 
injection is a dry mixture of dicloxacillin sodium monohydrate and 
lidocaine hydrochloride packaged for dispensing. Its potency is 
satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of milligrams of dicloxacillin that it is 
represented to contain. It is sterile. It is nonpyrogenic. Its moisture 
content is not more than 5 percent. When reconstituted as directed in 
the labeling, its pH is not less than 4.5 and not more than 7.5. The 
dicloxacillin sodium monohydrate used conforms to the standards 
prescribed by Sec. 440.19a(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``dicloxacillin sodium for 
injection''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The dicloxacillin sodium monohydrate used in making the batch 
for potency, moisture, pH, organic chlorine content, free chloride 
content, crystallinity, and identity.
    (b) The batch for potency, sterility, pyrogens, moisture, and pH.
    (ii) Samples required:
    (a) The dicloxacillin sodium monohydrate used in making the batch: 
10 packages, each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Reconstitute as directed in the labeling. Using a suitable hypodermic 
needle and syringe remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency in a given volume of the resultant preparation, 
remove an accurately measured representative portion from each 
container. Dilute the sample thus obtained with sufficient 1.0 percent 
potassium phosphate buffer, pH 6.0 (solution 1), for the microbiological 
agar diffusion assay or in distilled water for the iodometric assay and 
hydroxylamine

[[Page 568]]

colorimetric assay, to give a stock solution of convenient 
concentration.
    (ii) Assay procedure. Use any of the following methods; however, the 
results obtained from the microbiological agar diffusion assay shall be 
conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 5 micrograms of 
dicloxacillin per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with distilled water 
to the prescribed concentration.
    (c) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter, except dilute an aliquot of the stock 
solution with distilled water to the prescribed concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 20 milligrams of dicloxacillin per 
milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the product reconstituted as directed in the labeling.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59867, Nov. 22, 1977; 50 
FR 19918, 19919, May 13, 1985]



Sec. 440.229  Hetacillin potassium injectable dosage forms.



Sec. 440.229a  Sterile hetacillin potassium.

    The requirements for certification and the tests and methods of 
assay for sterile hetacillin potassium packaged for dispensing are 
described in Sec. 440.29a.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59867, Nov. 22, 1977]



Sec. 440.229b  Hetacillin potassium for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Hetacillin potassium for injection is a 
dry mixture of hetacillin potassium and lidocaine hydrochloride. Its 
potency is satisfactory if it contains not less than 90 percent and not 
more than 120 percent of the number of milligrams of ampicillin that it 
is represented to contain. It is sterile and nonpyrogenic. Its moisture 
content is not more than 1.0 percent. When reconstituted as directed in 
its labeling, its pH is not less than 7.0 and not more than 9.0. The 
hetacillin potassium used conforms to the requirements of 
Sec. 440.29a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The hetacillin potassium used in making the batch for potency, 
moisture, pH, hetacillin content, identity, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, moisture, and pH.
    (ii) Samples required:
    (a) The hetacillin potassium used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers, except if each contains less than 450 milligrams of 
ampicillin, a minimum of 16 immediate containers.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed for 
ampicillin in Sec. 436.105 of this chapter, using the ampicillin working 
standard as the standard of comparison and preparing the sample for 
assay as follows: Reconstitute as directed in the labeling. Using a 
suitable hypodermic needle and syringe, remove the withdrawable contents 
from each container represented as a single-dose container; or, if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, withdraw an accurately measured representative 
portion from each container. Dilute the sample thus obtained with 
sufficient 0.1M potassium phosphate buffer, pH 8.0 (solution

[[Page 569]]

3), to give a stock solution of convenient concentration. Further dilute 
the stock solution with solution 3 to the reference concentration of 0.1 
microgram of ampicillin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing the equivalent of 18 milligrams of 
ampicillin per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the product reconstituted as directed in the labeling.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59867, Nov. 22, 1977; 50 
FR 19918, 19919, May 13, 1985]



Sec. 440.236  Methicillin sodium monohydrate for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Methicillin sodium monohydrate for 
injection is methicillin sodium monohydrate with or without one or more 
suitable and harmless preservatives and the buffer sodium citrate in a 
quantity not less than 4 percent and not more than 5 percent by weight 
of its total solids (such sodium citrate conforms to the standards 
prescribed therefor by the U.S.P.). Its potency is satisfactory if it is 
not less than 90 percent and not more than 115 percent of the number of 
milligrams of methicillin that it is represented to contain. It is 
sterile. It is nonpyrogenic. Its pH in an aqueous solution containing 10 
milligrams per milliliter is not less than 6.0 and not more than 8.5. 
Its moisture content is not more than 6.0 percent. The methicillin 
sodium monohydrate used conforms to the standards prescribed by 
Sec. 440.36a(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``methicillin sodium for 
injection''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this subchapter, each such 
request shall contain:
    (i) Results of tests and assays on:
    (a) The methicillin sodium monohydrate used in making the batch for 
potency, moisture, pH, methicillin content, crystallinity, and identity.
    (b) The batch for potency, sterility, pyrogens, pH, and moisture.
    (ii) Samples required:
    (a) The methicillin sodium monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams, plus one package 
containing approximately 2 grams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Reconstitute as directed in the labeling. Using a suitable hypodermic 
needle and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency in a given volume of the resultant preparation, 
remove an accurately measured representative portion from each 
container. Dilute the portion thus obtained with 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to give a stock solution of 
convenient concentration.
    (ii) Assay procedure. Use either of the following methods; however, 
the results obtained from the microbiological agar diffusion assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this subchapter, diluting an aliquot of the stock 
solution with solution 1 to the reference concentration of 10 micrograms 
of methicillin per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
subchapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this 
subchapter, using the method described in paragraph (e)(1) of that 
section.

[[Page 570]]

    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 60 milligrams of methicillin per milliliter.
    (4) [Reserved]
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per millilter.
    (6) Moisture. Proceed as directed in Sec. 436.201 of this 
subchapter.
[39 FR 18976, May 30, 1974, as amended at 40 FR 15089, Apr. 4, 1975; 42 
FR 59868, Nov. 22, 1977; 49 FR 5096, Feb. 10, 1984; 50 FR 19918, 19919, 
May 13, 1985]



Sec. 440.237  Sterile mezlocillin sodium monohydrate.

    The requirements for certification and the tests and methods of 
assay for sterile mezlocillin sodium monohydrate packaged for dispensing 
are described in Sec. 440.37a.
[46 FR 58299, Dec. 1, 1981]



Sec. 440.241  Nafcillin sodium injectable dosage forms.



Sec. 440.241a  Nafcillin sodium monohydrate for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nafcillin sodium monohydrate for 
injection is a dry mixture of nafcillin sodium monohydrate and a 
suitable buffer substance. Its potency is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the number of 
milligrams of nafcillin that it is represented to contain. It is 
sterile. It is nonpyrogenic. Its moisture content is not less than 3.5 
and not more than 5.3 percent. When reconstituted as directed in the 
labeling, the pH is not less than 6.0 and not more than 8.5. The 
nafcillin sodium monohydrate used conforms to the requirements of 
Sec. 440.41a(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``nafcillin sodium for 
injection''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The nafcillin sodium monohydrate used in making the batch for 
potency, moisture, pH, crystallinity, nafcillin content, and identity.
    (b) The batch for potency, sterility, pyrogens, moisture, and pH.
    (ii) Samples required:
    (a) The nafcillin sodium monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 12 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Reconstitute as directed in the labeling. Using a suitable hypodermic 
needle and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency in a given volume of the resultant preparation 
remove an accurately measured representative portion from each 
container. Dilute the sample thus obtained with 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to the reference concentration of 
2.0 micrograms of nafcillin per milliliter (estimated) for the 
microbiological agar diffusion assay and to the prescribed concentration 
for the iodometric assay.
    (ii) Assay procedures. Assay for potency by either of the following 
methods; however, the results obtained from the microbiological agar 
diffusion assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter.
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 80 milligrams of nafcillin per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.

[[Page 571]]

    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution obtained when the product is reconstituted as directed in 
the labeling.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59868, Nov. 22, 1977; 45 
FR 22922, Apr. 4, 1980; 47 FR 22515, May 25, 1982; 50 FR 19919, May 13, 
1985. Redesignated at 55 FR 277, Jan. 4, 1990]



Sec. 440.241b  Nafcillin sodium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nafcillin sodium injection is a frozen, 
aqueous, iso-osmatic solution of nafcillin sodium which may contain one 
or more suitable and harmless buffer substances and a tonicity adjusting 
agent. Each milliliter contains nafcillin sodium equivalent to 20 or 40 
milligrams of nafcillin. Its nafcillin content is satisfactory if it is 
not less than 90 percent and not more than 120 percent of the number of 
milligrams of nafcillin that it is represented to contain. It is 
sterile. It is nonpyrogenic. Its pH is not less than 6.0 and not more 
than 8.5. The nafcillin sodium monohydrate used conforms to the 
standards prescribed by Sec. 440.41(a)(1).
    (2) Labeling. it shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. In addition, this drug shall 
be labeled ``nafcillin sodium injection.''
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The nafcillin sodium monohydrate used in making the batch for 
potency, moisture, pH, crystallinity, nafcillin content, and identity.
    (B) The batch for nafcillin content, sterility, pyrogens, and pH.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research:
    (A) The nafcillin sodium monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Nafcillin content. Proceed as directed in Sec. 440.241a(b)(1), 
except use the thawed solution.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
except inject a sufficient volume of the undiluted solution to deliver 
80 milligrams of nafcillin per kilogram.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[55 FR 277, Jan. 4, 1990]



Sec. 440.249  Oxacillin sodium injectable dosage forms.



Sec. 440.249a  Oxacillin sodium monohydrate for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxacillin sodium monohydrate for 
injection is a dry mixture of oxacillin sodium monohydrate and one or 
more buffer substances, with or without trisodium ethylenediamine 
tetraacetic acid, and with or without one or more suitable and harmless 
preservatives. Its potency is satisfactory if it is not less than 90 
percent and not more than 115 percent of the number of milligrams of 
oxacillin that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its moisture content is not more than 6.0 percent. Its pH 
in an aqueous solution containing 30 milligrams per milliliter is not 
less than 6.0 and not more than 8.5. The oxacillin sodium monohydrate 
used conforms to the standards prescribed by Sec. 440.49a(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``oxacillin sodium for 
injection''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oxacillin sodium monohydrate used in making the batch

[[Page 572]]

for potency, moisture, pH, oxacillin content, crystallinity, and 
identity.
    (b) The batch for potency, sterility, pyrogens, moisture, and pH.
    (ii) Samples required:
    (a) The oxacillin sodium monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation, or 40 immediate 
containers if each contains less than 600 milligrams.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Reconstitute as directed in the labeling. Then using a suitable 
hypodermic needle and syringe, remove all of the withdrawable contents 
if it is represented as a single-dose container, or, if the labeling 
specifies the amount of potency in a given volume of the resultant 
preparation, remove an accurately measured representative portion from 
each container. Dilute with 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to give a stock solution of convenient concentration.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the microbiological agar diffusion assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 5 micrograms of 
oxacillin per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 20 milligrams of oxacillin per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 30 milligrams per milliliter.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59868, Nov. 22, 1977; 50 
FR 19918, 19919, May 13, 1985. Redesignated at 55 FR 279, Jan. 4, 1990]



Sec. 440.249b  Oxacillin sodium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxacillin sodium injection is a frozen 
aqueous, iso-osmotic solution of oxacillin sodium which may contain one 
or more suitable and harmless buffer substances and a tonicity adjusting 
agent. Each milliliter contains oxacillin sodium equivalent to 20 or 40 
milligrams of oxacillin. Its oxacillin content is satisfactory if it is 
not less than 90 percent and not more than 115 percent of the number of 
milligrams of oxacillin that it is represented to contain. It is 
sterile. It is nonpyrogenic. Its pH is not less than 6.0 and not more 
than 8.5. The oxacillin sodium monohydrate used conforms to the 
standards prescribed by Sec. 440.49(a)(1), except that the pH of an 
aqueous solution containing 30 milligrams per milliliter is not less 
than 4.0 and not more than 7.0.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. In addition, this drug shall 
be labeled ``oxacillin sodium injection''.
    (3) Requests for certification: samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The oxacillin sodium monohydrate used in making the batch for 
potency, moisture, pH, oxacillin content, crystallinity, and identity.
    (B) The batch for oxacillin content, sterility, pyrogens, and pH.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research:
    (A) The oxacillin sodium monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (B) The batch:

[[Page 573]]

    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the sample as directed in the 
labeling. he sample solution used for testing must be at room 
temperature.
    (1) Oxacillin content. Proceed as directed in Sec. 440.249a(b)(1), 
except use the thawed solution.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
except inject a sufficient volume of the undiluted solution to deliver 
20 milligrams of oxacillin per kilogram.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[55 FR 279, Jan. 4, 1990; 55 FR 2481, Jan. 24, 1990]



Sec. 440.255  Penicillin G benzathine injectable dosage forms.



Sec. 440.255b  Sterile penicillin G benzathine suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile penicillin G benzathine 
suspension is an aqueous suspension of penicillin G benzathine and one 
or more suitable suspending or dispersing agents, buffer substances, and 
preservatives. Each container or each milliliter contains penicillin G 
benzathine equivalent to not less than 300,000 units of penicillin G. 
Its potency is satisfactory if it is not less than 90 percent and not 
more than 115 percent of the number of units of penicillin G that it is 
represented to contain. It is sterile. It is nonpyrogenic. Its pH is not 
less than 5.0 and not more than 7.5. The penicillin G benzathine used 
conforms to the standards prescribed by Sec. 440.55a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G benzathine used in making the batch for 
potency, moisture, pH, penicillin G content, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, and pH.
    (ii) Samples required:
    (a) The penicillin G benzathine used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filing operation.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the iodometric 
assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Using a suitable hypodermic needle and syringe, remove all of the 
withdrawable contents if it is represented as a single-dose container; 
or, if the labeling specifies the amount of potency in a given volume, 
remove an accurately measured representative portion from each 
container. Dilute the portion thus obtained with sufficient absolute 
methyl alcohol to give a solution of convenient concentration. 
Immediately further dilute with 1 percent potassium phosphate buffer, pH 
6.0 (solution 1), to the reference concentration of 1.0 unit of 
penicillin G per milliliter (estimated).
    (ii) Iodometric assay. Using a suitable hypodermic needle and 
syringe, remove all of the withdrawable contents if it is represented as 
a single-dose container; or if the labeling specifies the amount of 
potency in a given volume, remove an accurately measured representative 
portion from each container. Using the sample thus obtained, proceed as 
directed in Sec. 436.204(b)(2) of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section, except 
use medium C in lieu of medium A, and medium F in lieu of

[[Page 574]]

medium E. During the period of incubation, shake the tubes at least once 
daily.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(d) of this chapter, 
using a solution containing 4,000 units of penicillin G per milliliter.
    (4) [Reserved]
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[42 FR 59868, Nov. 22, 1977, as amended at 43 FR 9799, Mar. 10, 1978; 50 
FR 19918, 19919, May 13, 1985]



Sec. 440.255c  Sterile penicillin G benzathine-penicillin G procaine suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile penicillin G benzathine-
penicillin G procaine suspension is an aqueous mixture of penicillin G 
benzathine and penicillin G procaine with or without suitable and 
harmless buffer substances, suspending agents, and preservatives. Each 
container or each milliliter contains penicillin G benzathine and 
penicillin G procaine each equivalent to not less than 150,000 units of 
penicillin G. Its penicillin G benzathine content is satisfactory if it 
is not less than 90 percent and not more than 115 percent of the number 
of units of penicillin G that it is represented to contain. Its 
penicillin G procaine content is satisfactory if it is not less than 90 
percent and not more than 115 percent of the number of units of 
penicillin G that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its pH is not less than 5.0 and not more than 7.5. The 
penicillin G benzathine used conforms to the standards prescribed by 
Sec. 440.55a (a)(1). The penicillin G procaine used conforms to the 
standards prescribed by Sec. 440.74a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G benzathine used in making the batch for 
potency, moisture, pH, penicillin G content, and crystallinity.
    (b) The penicillin G procaine used in making the batch for potency, 
moisture, pH, penicillin G content, and crystallinity.
    (c) The batch for penicillin G benzathine content, penicillin G 
procaine content, sterility, pyrogens, and pH.
    (ii) Samples required:
    (a) The penicillin G benzathine used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (b) The penicillin G procaine used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (c) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regualr intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Total potency. 
Assay for total potency by either of the following methods; however, the 
results obtained from the iodometric assay shall be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Using a suitable hypodermic needle and syringe, place one dose of the 
drug in a 100-milliliter volumetric flask and add sufficient methyl 
alcohol to dissolve the benzathine penicillin G. Dilute to volume with 1 
percent potassium phosphate buffer, pH 6.0 (solution 1), and shake well. 
Immediately further dilute an aliquot with solution 1 to the reference 
concentration of 1.0 unit of penicillin G per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, preparing the sample as follows: Using a suitable hypodermic 
needle and syringe, withdraw 2 one-dose portions of sample. Place one 
portion into an appropriate-sized volumetric flask and add 20 
milliliters of 0.5N NaOH for each 300,000 units of benzathine penicillin 
G, mix well, being sure that all penicillin is in solution, and allow to 
stand for 15 minutes. Add 1 milliliter of 1.2N HCl for each 2 
milliliters of 0.5N NaOH,

[[Page 575]]

mix, and dilute with distilled water to obtain a concentration of 2,000 
units per milliliter. Dilute the other portion, which is to be used as 
the blank solution, with 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to give a concentration of approximately 2,000 units per 
milliliter.
    (ii) Penicillin G procaine content--(a) Reagents--(1) Sodium nitrite 
solution. Dissolve 0.1 gram of sodium nitrite in 100 milliliters of 
distilled water. Prepare a fresh solution every week and store under 
refrigeration.
    (2) Ammonium sulfamate solution. Dissolve 0.5 gram of ammonium 
sulfamate in 100 milliliters of distilled water and store under 
refrigeration.
    (3) N-(1-naphthyl)-ethylenediamine solution. Dissolve 0.1 gram of N-
(1-naphthyl) - ethylenediamine dihydrochloride in 100 milliliters of 
distilled water. Prepare fresh solutions every week and store under 
refrigeration.
    (4) Standard procaine solution. Prepare a standard solution 
containing 27.55 milligrams of procaine hydrochloride U.S.P. in a liter 
of distilled water (each milliliter of the standard solution is 
equivalent to 60 units of penicillin G procaine).
    (b) Preparation of sample solution. Using a suitable hypodermic 
needle and syringe, withdraw a one-dose portion of the sample and place 
it into an appropriate-sized volumetric flask. Add 20 milliliters of 
0.5N NaOH for each 300,000 units of penicillin G benzathine, mix well, 
being sure that all penicillin is in solution, and allow to stand for 15 
minutes. Add 1 milliliter of 1.2N HC1 for each 2 milliliters of 0.5N 
NaOH, mix, and dilute with distilled water to obtain a concentration of 
60 units of penicillin G procaine per milliliter. Transfer a 3.0-
milliliter aliquot of this solution to a 50-milliliter volumetric flask 
and add 2 milliliters of water to give a volume of 5 milliliters.
    (c) Procedure. Transfer respectively, 1.0, 2.0, 3.0, 4.0, and 5.0 
milliliters of the standard procaine solution to each of five 50-
milliliter volumetric flasks and transfer 5.0 milliliters of distilled 
water to a sixth 50-milliliter volumetric flask. Add 4.0, 3.0, 2.0, and 
1.0 milliliters of water to the first four flasks, respectively, to give 
each a volume of 5 milliliters. To each flask of the standard and sample 
solutions, add 0.5 milliliter of 4N HC1, 1.0 milliliter of sodium 
nitrite solution, 1.0 milliliter of ammonium sulfamate solution, and 1.0 
milliliter of N-(1-naphthyl)-ethylenediamine solution. Mix and wait two 
minutes after each addition. Dilute each flask to volume with distilled 
water. Using a suitable photoelectric colorimeter, determine the 
absorbancy of each solution at 550 nanometers. The instrument is 
balanced so that the zero concentration reads 0 absorbancy. Plot the 
standard curve on coordinate graph paper. Obtain the procaine penicillin 
content of the solution for assay directly from the point on the 
standard curve corresponding to its absorbancy.
    (iii) Penicillin G benzathine content. The sum of the penicillin G 
procaine content determined as directed in paragraph (b)(1)(ii) of this 
section subtracted from the total potency determined as directed in 
paragraph (b)(1)(i) of this section represents the penicillin G 
benzathine content.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section, except 
use medium C in lieu of medium A, medium F in lieu of medium E, and 
during the period of incubation, shake the tubes at least once daily.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(d) of this chapter, 
using a solution containing 4,000 units of penicillin G per milliliter.
    (4) [Reserved]
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted aqueous suspension.
[42 FR 59868, Nov. 22, 1977, as amended at 43 FR 9799, Mar. 10, 1978; 50 
FR 19918, 19919, May 13, 1985]



Sec. 440.255d  Sterile penicillin G benzathine for suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile penicillin G benzathine for 
suspension is a dry mixture of penicillin G benzathine and one or more 
suitable suspending or dispersing agents, and with or without one or 
more suitable preservatives and buffer substances. Its potency is 
satisfactory if it is not less than 90 percent and not

[[Page 576]]

more than 115 percent of the number of units of penicillin G that it is 
represented to contain. It is sterile. It is nonpyrogenic. Its moisture 
content is not less than 5.0 percent and not more than 8.0 percent. When 
reconstituted as directed in the labeling, its pH is not less than 5.0 
and not more than 7.5. The penicillin G benzathine used conforms to the 
standards prescribed by Sec. 440.55a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G benzathine used in making the batch for 
potency, moisture, pH, penicillin G content, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, moisture, and pH.
    (ii) Samples required:
    (a) The penicillin G benzathine used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) If the batch is packaged for repacking:
    (i) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (ii) For sterility testing: 20 packages, each containing 
approximately 600 milligrams.
    (2) If the batch is packaged for dispensing:
    (i) For all tests except sterility: A minimum of 10 immediate 
containers.
    (ii) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the iodometric 
assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Using a suitable hypodermic 
needle and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency in a given volume of the resultant preparation, 
remove an accurately measured representative portion from each 
container. Dissolve the portion thus obtained with sufficient absolute 
methyl alcohol to give a solution of convenient concentration. 
Immediately, further dilute with 1 percent potassium phosphate buffer, 
pH 6.0 (solution 1), to the reference concentration of 1.0 unit of 
penicillin G per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, preparing the sample as follows: Reconstitute as directed in 
the labeling. Using a suitable hypodermic needle and syringe, remove all 
of the withdrawable contents if it is represented as a single-dose 
container; or, if the labeling specifies the amount of potency in a 
given volume of the resultant preparation, remove an accurately measured 
representative portion from each container. Using the sample thus 
obtained, proceed as directed in Sec. 436.205(b)(2) of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section, except 
use medium C in lieu of medium A, and medium F in lieu of medium E. 
During the period of incubation shake the tubes at least once daily.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(d) of this chapter, 
using a solution containing 4,000 units of penicillin G per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the suspension obtained when the product is reconstituted as directed in 
the labeling.
[42 FR 59869, Nov. 22, 1977, as amended at 50 FR 19918, 19919, May 13, 
1985]



Sec. 440.274  Penicillin G procaine injectable dosage forms.



Sec. 440.274a  Sterile penicillin G procaine with aluminum stearate suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality,

[[Page 577]]

and purity. Sterile penicillin G procaine with aluminum stearate 
suspension is penicillin G procaine in a refined vegetable oil with one 
or more suitable and harmless dispersing agents and hardening agents. 
Each milliliter contains penicillin G procaine equivalent to 300,000 
units of penicillin G. Its potency is satisfactory if it is not less 
than 90 percent and not more than 115 percent of the number of units of 
penicillin G that it is represented to contain. It is sterile. Its 
moisture content is not more than 1.4 percent. The penicillin G procaine 
used conforms to the standards prescribed by Sec. 440.74a(a)(1). If the 
hardening agent is a refined hydrogenated and deodorized peanut oil, it 
is free from rancidity; it has an iodine value of not more than 10; its 
free fatty acid content as oleic acid is not more than one-tenth of 1 
percent and its melting point is 64 deg. Cplus-minus2 deg. C.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G procaine used in making the batch for potency, 
pyrogens, moisture, pH, penicillin G content, and crystallinity.
    (b) The batch for potency, sterility, and moisture.
    (ii) Samples required:
    (a) The penicillin G procaine used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 5 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place an accurately measured representative portion of the sample or the 
entire contents of a single-dose container into a 50-milliliter 
volumetric flask. Add 4 milliliters of chloroform and mix thoroughly. 
Dilute to volume with absolute ethyl alcohol. Mix well. Immediately 
further dilute with sufficient 1.0 percent potassium phosphate buffer, 
pH 6.0 (solution 1), to the reference concentration of 1 unit of 
penicillin G per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section, except 
use medium B in lieu of medium A.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[42 FR 59870, Nov. 22, 1977, as amended at 50 FR 19919, May 13, 1985]



Sec. 440.274b  Sterile penicillin G procaine suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile penicillin G procaine suspension 
is an aqueous mixture of penicillin G procaine and one or more suitable 
suspending or dispersing agents, buffer substances, and preservatives. 
It may contain procaine hydrochloride in a concentration not exceeding 
2.0 percent and one or more suitable stabilizing agents. Each container 
or each milliliter contains penicillin G procaine equivalent to not less 
than 300,000 units of penicillin G. Its potency is satisfactory if it is 
not less than 90 percent and not more than 115 percent of the number of 
units of penicillin G that it is represented to contain. It is sterile. 
It is nonpyrogenic. Its pH is not less than 5.0 and not more than 7.5. 
The penicillin G procaine used conforms to the standards prescribed by 
Sec. 440.74a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G procaine used in making the batch for potency, 
moisture, pH, penicillin G content, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, and pH.
    (ii) Samples required:
    (a) The penicillin G procaine used in making the batch: 10 packages, 
each containing approximately 300 milligrams.

[[Page 578]]

    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the iodometric 
assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Using a suitable hypodermic needle and syringe, remove all of the 
withdrawable contents, if it is represented as a single-dose container; 
or, if the labeling specifies the amount of potency in a given volume, 
remove an accurately measured representative portion from each 
container. Dissolve the sample thus obtained in 50 to 100 milliliters of 
absolute methyl alcohol and add sufficient 1 percent potassium phosphate 
buffer, pH 6.0 (solution 1), to give a stock solution of convenient 
concentration. Immediately further dilute an aliquot of the stock 
solution with solution 1 to the reference concentration of 1.0 unit of 
penicillin G per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, preparing the sample as follows: Using a suitable hypodermic 
needle and syringe, remove all of the withdrawable contents, if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency in a given volume, remove an accurately measured 
representative portion from each container. Dissolve and dilute the 
sample thus obtained with 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to the prescribed concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
add sufficient penicillinase to diluting fluid A and swirl the flask to 
completely solubilize the procaine penicillin before filtration. If the 
product contains lecithin, use diluting fluid D in lieu of diluting 
fluid A. If the product contains sodium carboxymethylcellulose, add 
sufficient sterile carboxymethylcellulase to diluting fluid A or D to 
completely solubilize the sodium carboxymethylcellulose before 
filtration. If the preparation contains homogenizers or suspending 
agents that prevent solubilization, proceed as directed in paragraph 
(e)(2) of that section, except use medium B in lieu of medium A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(h) of this chapter, 
using a solution containing 2,000 units of penicillin G per milliliter.
    (4) [Reserved]
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted drug.
[42 FR 59870, Nov. 22, 1977, as amended at 43 FR 9799, Mar. 10, 1978; 45 
FR 22922, Apr. 4, 1980; 50 FR 19918, 19919, May 13, 1985]



Sec. 440.274c  Sterile penicillin G procaine for suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile penicillin G procaine for 
suspension is a dry mixture of penicillin G procaine and one or more 
suitable suspending or dispersing agents, buffer substances, and 
preservatives. Its potency is satisfactory if it is not less than 90 
percent and not more than 115 percent of the number of units of 
penicillin G that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its moisture content is not less than 2.8 and not more 
than 4.2 percent. When reconstituted as directed in the labeling, its pH 
is not less than 5.0 and not more than 7.5. The penicillin G procaine 
used conforms to the standards prescribed by Sec. 440.74a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G procaine used in making the batch for potency, 
moisture, pH, penicillin G content, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, moisture, and pH.
    (ii) Samples required:

[[Page 579]]

    (a) The penicillin G procaine used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch:
    (1) If the batch is packaged for repacking:
    (i) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (ii) For sterility testing: 20 packages, each containing 
approximately 600 milligrams.
    (2) If the batch is packaged for dispensing:
    (i) For all tests except sterility: A minimum of 10 immediate 
containers.
    (ii) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the iodometric 
assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
If it is packaged for repacking, dissolve an accurately weighed sample, 
equivalent to one dose, in 50 to 100 milliliters of absolute methyl 
alcohol and add sufficient 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to give a stock solution of convenient concentration. If 
it is packaged for dispensing, reconstitute as directed in the labeling. 
Then using a suitable hypodermic needle and syringe, remove all of the 
withdrawable contents, if it is represented as a single-dose container; 
or, if the labeling specifies the amount of potency in a given volume of 
the resultant preparation, remove an accurately measured representative 
portion from each container. Dissolve the sample thus obtained in 50 to 
100 milliliters of absolute methyl alcohol and add sufficient 1 percent 
potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration. Immediately further dilute an 
aliquot of the stock solution with solution 1 to the reference 
concentration of 1.0 unit of penicillin G per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, preparing the sample as follows: If it is packaged for 
repacking, dissolve and dilute an accurately weighed sample, equivalent 
to one dose, with 1 percent potassium phosphate buffer, pH 6.0 (solution 
1), to the prescribed concentration. If it is packaged for dispensing, 
reconstitute as directed in the labeling. Then using a suitable 
hypodermic needle and syringe, remove all of the withdrawable contents, 
if it is represented as a single-dose container; or, if the labeling 
specifies the amount of potency in a given volume, remove an accurately 
measured representative portion from each container. Dissolve and dilute 
the sample thus obtained with 1 percent potassium phosphate buffer, pH 
6.0 (solution 1), to the prescribed concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
add sufficient penicillinase to diluting fluid A and swirl the flask to 
completely solubilize the procaine penicillin before filtration. If the 
product contains lecithin, use diluting fluid D in lieu of diluting 
fluid A. If the product contains sodium carboxymethylcellulose, add 
sufficient sterile carboxymethylcellulase to diluting fluid A or D to 
completely solubilize the sodium carboxymethylcellulose before 
filtration. If the preparation contains homogenizers or suspending 
agents that prevent solubilization, proceed as directed in paragraph 
(e)(2) of that section, except use medium B in lieu of medium A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(h) of this chapter, 
using a solution containing 2,000 units of penicillin G per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the suspension obtained when reconstituted as directed in the labeling.
[42 FR 59871, Nov. 22, 1977, as amended at 45 FR 22922, Apr. 4, 1980; 50 
FR 19918, 19919, May 13, 1985]

[[Page 580]]



Sec. 440.280  Penicillin G potassium injectable dosage forms.



Sec. 440.280a  Sterile penicillin G potassium.

    The requirements for certification and the tests and methods of 
assay for sterile penicillin G potassium packaged for dispensing are 
described in Sec. 440.80a.
[39 FR 18976, May 30, 1974, as amended at 42 FR 59871, Nov. 22, 1977]



Sec. 440.280b  Penicillin G potassium for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G potassium for injection is a 
dry mixture of penicillin G potassium and the buffer sodium citrate in a 
quantity not less than 4.0 percent and not more than 5.0 percent by 
weight of its total solids. It may contain citric acid in a quantity not 
more than 0.15 percent of its total solids in place of a corresponding 
amount of sodium citrate. Its potency is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the number of units of 
penicillin G that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its loss on drying is not more than 1.5 percent. Its pH is 
not less than 6.0 and not more than 8.5. If penicillin G potassium 
buffered is used, it conforms to the standards prescribed by 
Sec. 440.1080(a)(1). If penicillin G potassium is used, it conforms to 
the standards prescribed by Sec. 440.80a(a)(1) and the sodium citrate 
and citric acid conforms to the standards prescribed by the U.S.P.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G potassium used in making the batch for potency, 
loss on drying, pH, penicillin G content, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, loss on drying, and 
pH.
    (ii) Samples required:
    (a) The penicillin G potassium, buffered, used in making the batch: 
10 packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Reconstitute as directed in the labeling. Then, using a suitable 
hypodermic needle and syringe, remove all of the withdrawable contents 
if it is represented as a single-dose container; or, if the labeling 
specifies the amount of potency in a given volume of the resultant 
preparation, remove or expel an accurately measured representative 
portion from each container. Dilute with solution 1 to give a stock 
solution of convenient concentration.
    (ii) Assay procedures. Assay for potency by any of the following 
methods; however, the results obtained from the iodometric assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 unit of penicillin 
G per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (c) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the prescribed concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 20,000 units of penicillin G per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 60 milligrams per milliliter or, if the 
diluent is included in a disposable syringe combination, use the 
solution obtained when the drug is

[[Page 581]]

reconstituted as directed in the labeling.
[42 FR 59871, Nov. 22, 1977, as amended at 43 FR 9799, Mar. 10, 1978; 45 
FR 22922, Apr. 4, 1980; 50 FR 19918, 19919, May 13, 1985]



Sec. 440.280c  Penicillin G potassium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G potassium injection is a 
frozen, aqueous, iso-osmotic solution of penicillin G potassium which 
may contain one or more suitable and harmless buffer substances and a 
tonicity adjusting agent. Each milliliter contains penicillin G 
potassium equivalent to 20,000, 40,000, or 60,000 units of penicillin G. 
Its penicillin G content is satisfactory if it is not less than 90 
percent and not more than 115 percent of the number of units of 
penicillin G that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its pH is not less than 5.5 and not more than 8.0. The 
penicillin G potassium used conforms to the standards prescribed by 
Sec. 440.80(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. In addition, this drug shall 
be labeled ``penicillin G potassium injection''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The penicillin G potassium used in making the batch for potency, 
loss on drying, pH, penicillin G content, and crystallinity.
    (B) The batch for penicillin G content, sterility, pyrogens, and pH.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research:
    (A) The penicillin G potassium used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Penicillin G content. Proceed as directed in Sec. 440.280b(b)(1) 
of this chapter, except use the thawed solution.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
except inject a sufficient volume of the undiluted solution to deliver 
20,000 units of penicillin G per kilogram.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[55 FR 38675, Sept. 20, 1990]



Sec. 440.281  Pencillin G sodium injectable dosage forms.



Sec. 440.281a  Sterile penicillin G sodium.

    The requirements for certification and the tests and methods of 
assay for sterile penicillin G sodium packaged for dispensing are 
described in Sec. 440.81a.
[42 FR 59872, Nov. 22, 1977]



Sec. 440.281b  Penicillin G sodium for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G sodium for injection is a 
dry mixture of penicillin G sodium and the buffer sodium citrate in a 
quantity not less than 4.0 percent and not more than 5.0 percent by 
weight of its total solids. It may contain citric acid in a quantity not 
more than 0.15 percent of its total solids in place of a corresponding 
amount of sodium citrate. Its potency is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the number of units of 
penicillin G that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its loss on drying is not more than 1.5 percent. Its pH is 
not less than 6.0 and not more than 7.5. The penicillin G sodium, 
buffered, used conforms to the standards prescribed by 
Sec. 440.1081a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 582]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G sodium, buffered, used in making the batch for 
potency, loss on drying, pH, penicillin G content, crystallinity, and 
heat stability.
    (b) The batch for potency, sterility, pyrogens, loss on drying, and 
pH.
    (ii) Samples required:
    (a) The penicillin G sodium, buffered, used in making the batch: 10 
packages, each containing approximately 60 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Reconstitute as directed in the labeling. Then using a suitable 
hypodermic needle and syringe, remove all of the withdrawable contents 
if it is represented as a single-dose container; or, if the labeling 
specifies the amount of potency in a given volume of the resultant 
preparation, remove an accurately measured representative portion from 
each container. Dilute with solution 1 to give a stock solution of 
convenient concentration.
    (ii) Assay procedures. Assay for potency by any of the following 
methods; however, the results obtained from the iodometric assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 unit of penicillin 
G per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (c) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the prescribed concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 20,000 units of penicillin G per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed Sec. 436.202 of this chapter, using an 
aqueous solution containing 60 milligrams per milliliter.
[42 FR 59872, Nov. 22, 1977; 43 FR 2393, Jan. 17, 1978, as amended at 45 
FR 22922, Apr. 4, 1980; 50 FR 19918, 19919, May 13, 1985]



Sec. 440.283  Sterile piperacillin sodium.

    The requirements for certification and the tests and methods of 
assay for sterile piperacillin sodium packaged for dispensing are 
described in Sec. 440.83a
[47 FR 15770, Apr. 13, 1982]



Sec. 440.290  Ticarcillin disodium injectable dosage forms.



Sec. 440.290a  Sterile ticarcillin disodium.

    The requirements for certification and the tests and methods of 
assay for sterile ticarcillin disodium packaged for dispensing are 
described in Sec. 440.90a.
[43 FR 9800, Mar. 10, 1978. Redesignated at 50 FR 33518, Aug. 20, 1985]



Sec. 440.290b  Sterile ticarcillin disodium and clavulanate potassium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ticarcillin disodium and clavulanate 
potassium is a dry mixture of ticarcillin disodium and clavulanate 
potassium, in which the ratio of ticarcillin to clavulanic acid is 15:1 
or 30:1. Its ticarcillin potency is not less than 755 micrograms of 
ticarcillin per milligram on an anhydrous basis if the ratio is 30:1 and 
733 micrograms of ticarcillin per milligram on an anhydrous basis if the 
ratio is 15:1. Its ticarcillin disodium content is satisfactory if it 
contains not less than 90 percent and not more than 115 percent of the 
number of milligrams of ticarcillin that it is represented to contain. 
Its clavulanate potassium content is satisfactory if it contains not 
less than 85 percent and not more than 120 percent of the number of 
milligrams of

[[Page 583]]

clavulanic acid that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its moisture content is not more than 4.2 percent. Its pH 
of an aqueous solution containing 100 milligrams per milliliter is not 
less than 5.5 and not more than 7.5. The ticarcillin disodium conforms 
to the standards prescribed by Sec. 440.90a(a)(1) except that it 
contains not less than 840 micrograms of ticarcillin per milligram on an 
anhydrous basis. The clavulanate potassium conforms to the standards 
prescribed by Sec. 455.15a(a)(1) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The ticarcillin disodium used in making the batch for potency, 
sterility, pyrogens, moisture, pH, and identity.
    (b) The clavulanate potassium used in making the batch for potency, 
sterility, pyrogens, moisture, pH, identity, and clavam-2-carboxylate 
content.
    (c) The batch for ticarcillin potency, ticarcillin content, 
clavulanic acid content, sterility, pyrogens, moisture, and pH.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The ticarcillin disodium used in making the batch: 12 packages, 
each containing approximately 300 milligrams.
    (b) The clavulanate potassium used in making the batch: 12 packages, 
each containing approximately 300 milligrams.
    (c) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: A minimum of 20 immediate containers 
collected at regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Ticarcillin and clavulanic acid 
contents. Determine micrograms of ticarcillin per milligram of sample 
and milligrams of both ticarcillin and clavulanic acid per container. 
Proceed as directed in Sec. 436.355 of this chapter, using ambient 
temperature, an ultraviolet detection system operating at a wavelength 
between 220 and 230 nanometers, and a column packed with 
microparticulate (3 to 10 micrometers in diameter) reversed phase 
packing material such as octadecyl silane bonded silicas. Reagents, 
working standard and sample solutions, system suitability requirements, 
and calculations for ticarcillin or clavulanic acid content are as 
follows:
    (i) Reagents--(a) 0.1M Monobasic sodium phosphate buffer solution, 
pH 4.3. Transfer 13.8 grams of monobasic sodium phosphate monohydrate to 
a 1-liter volumetric flask and dissolve in 900 milliliters of distilled 
water. Adjust the pH to 4.30.1 with 18N phosphoric acid or 
10N sodium hydroxide. Dilute to volume with distilled water. Mix well.
    (b) Mobile phase. Mix acetonitrile: 0.1M monobasic sodium phosphate 
buffer solution, pH 4.3 (5:95 v/v) and mix for no less than two minutes. 
Degas by passing through a 0.5-micrometer filter with vacuum. The mobile 
phase may be sparged with the helium through a 2-micrometer metal filter 
for the duration of the analysis. Adjust the ratio of acetonitrile to 
aqueous buffer as necessary to obtain satisfactory separation of the 
peaks.
    (c) Diluent. 0.05M monobasic sodium phosphate buffer solution, pH 
6.4 Transfer 6.9 grams of monobasic sodium phosphate to a 1-liter 
volumetric flask and dissolve in 900 milliliters of water. Adjust the pH 
to 6.4 with sodium hydroxide (10N). Dilute to volume with distilled 
water. Mix well. Use this diluent to prepare the working standard and 
sample solutions described in paragraph (b)(1)(ii) of this section.
    (ii) Working standard and sample solutions--(a) Preparation of 
working standard solution. Accurately weigh a quantity of the 
ticarcillin working standard containing the equivalent of approximately 
90 milligrams of ticarcillin activity and transfer into a 100-milliliter 
volumetric flask. Prepare a solution of the clavulanic acid working 
standard containing the equivalent of 30 milligrams or 60 milligrams of 
clavulanic acid activity in a 100-milliliter volumetric flask. Dissolve 
and dilute to

[[Page 584]]

volume with diluent. Transfer 10 milliliters of this solution into the 
flask containing the ticarcillin standard. Dilute the combined standard 
solution to volume with diluent. Mix. Use within 8 hours after 
preparation.
    (b) Preparation of sample solutions--(1) Ticarcillin potency 
(micrograms of ticarcillin per milligram). Accurately weigh the total 
contents of a container and dissolve with sufficient diluent to obtain a 
stock solution containing approximately 30 milligrams of ticarcillin per 
milliliter. Further dilute this solution with diluent to obtain a final 
concentration of 0.9 milligrams of ticarcillin per milliliter 
(estimated).
    (2) Ticarcilin and clavulanic acid content (milligrams of 
ticarcillin and clavulanic acid per container). Reconstitute the 
container with an appropriate volume of distilled water. Using a 
suitable hypodermic syringe, remove all of the withdrawable contents. 
Dilute with diluent to obtain a stock solution containing approximately 
30 milligrams of ticarcillin per milliliter and 1 or 2 milligrams of 
clavulanic acid per milliliter. Further dilute this solution with the 
diluent to obtain a final concentration of 0.9 milligram of ticarcillin 
per milliliter. The final solution will contain either 0.03 or 0.06 
milligram of clavulanic acid per milliliter (estimated) depending on the 
initial ticarcillin to clavulanic acid ratio.
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 2.0.
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,000 theoretical plates in a 25-
centimeter column.
    (c) Resolution factor. The resolution factor (R) between the 
clavulanic acid and ticarcillin peaks is satisfactory if it is not less 
than 5.0.
    (d) Coefficient of variation. The coefficient of variation (SR 
in percent) is satisfactory if it is not more than 2.0 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.355(b) of this chapter.
    (iv) Calculations. (a) Calculate the micrograms of ticarcillin per 
milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.145

where:
Au=Area of the ticarcillin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the ticarcillin peak in the chromatogram of the 
          ticarcillin working standard;
Ps=Ticarcillin activity in the ticarcillin working standard 
          solution in micrograms of anhydrous ticarcillin free acid per 
          milliliter; and
Cu=Milligrams of sample per millilter of sample solution.

    (b) Calculate the ticarcillin or clavulanic acid anhydrous free acid 
content of the container as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.045

where:
Au=Area of the ticarcillin or clavulanic acid peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
As=Area of the ticarcillin or clavulanic acid peak in the 
          chromatogram of the ticarcillin or clavulanic acid working 
          standard;
Ps=Anhydrous ticarcillian or clavulanic free acid activity in 
          the ticarcillin-clavulanic acid working standard solution in 
          micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 100 milligrams of ticarcillin per 
milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 100 milligrams per milliliter.
[50 FR 34418, Aug. 20, 1985; 50 FR 42156, Oct. 18, 1985; 50 FR 43384, 
Oct. 25, 1985; 50 FR 45403, Oct. 31, 1985, as amended at 55 FR 11582, 
Mar. 29, 1990]

[[Page 585]]



Sec. 440.290c  Ticarcillin disodium and clavulanate potassium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ticarcillin disodium and clavulanate 
potassium injection is a frozen, aqueous, isoosmotic solution of 
ticarcillin disodium and clavulanate potassium with one or more suitable 
and harmless buffer substances. The ratio of ticarcillin to clavulanic 
acid is 30:1. Each milliliter contains ticarcillin disodium equivalent 
to 30 milligrams of ticarcillin and clavulanate potassium equivalent to 
1 milligram of clavulanic acid. Its ticarcillin content is satisfactory 
if it is not less than 90 percent and not more than 115 percent of the 
number of milligrams of ticarcillin that it is represented to contain. 
Its clavulanate potassium content is satisfactory if it contains not 
less than 85 percent and not more than 120 percent of the number of 
milligrams of clavulanic acid that it is represented to contain. It is 
sterile. It is nonpyrogenic. Its pH is not less than 5.5 and not more 
than 7.5. It passes the identity test. The ticarcillin monosodium 
monohydrate used conforms to the standards prescribed by 
Sec. 440.91(a)(1). The clavulanate potassium used conforms to the 
standards prescribed by Sec. 455.15(a)(1) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The ticarcillin monosodium monohydrate used in making the batch 
for potency, moisture, pH, identity, and crystallinity.
    (B) The clavulanate potassium used in making the batch for potency, 
moisture, pH, identity, and clavam-2-carboxylate content.
    (C) The batch for ticarcillin content, clavulanic acid content, 
sterility, pyrogens, pH, and identity.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research:
    (A) The ticarcillin monosodium monohydrate used in making the batch: 
12 packages, each containing approximately 300 milligrams.
    (B) The clavulanate potassium used in making the batch: 12 packages, 
each containing approximately 300 milligrams.
    (C) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Ticarcillin and clavulanic acid contents. Proceed as directed in 
Sec. 440.290b(b)(1), except use the thawed solution and prepare the 
sample solution and calculate the ticarcillin and clavulanic acid 
content as follows:
    (i) Preparation of sample solution. Using a suitable hypodermic 
needle and syringe, remove an accurately measured representative portion 
from each container immediately after thawing and reaching room 
temperature. Dilute with diluent (described in 
Sec. 440.290b(b)(1)(i)(c)) to obtain a solution containing approximately 
0.9 milligram of ticarcillin activity per milliliter (estimated). This 
solution will contain approximately 0.03 milligram of clavulanic acid 
per milliliter. Introduce the sample into the chromatograph in a timely 
manner.
    (ii) Calculations. Calculate the ticarcillin or clavulanic acid 
concentration as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.046

where:
Au=Area of the ticarcillin or clavulanic acid peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
As=Area of the ticarcillin or clavulanic acid peak in the 
          chromatogram of the ticarcillin or clavulanic acid working 
          standard;
Ps=Ticarcillin or clavulanic acid activity in the 
          ticarcillin-clavulanic acid working standard solution in 
          micrograms per milliliter; and
d=Dilution factor of the sample.


[[Page 586]]


    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in Sec. 436.20(e)(1).
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
except inject a sufficient volume of the undiluted solution to deliver 
100 milligrams of ticarcillin per kilogram.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (5) Identity. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the ticarcillin and clavulanic acid working 
standard.
[55 FR 5840, Feb. 20, 1990]



                        Subparts D-J  [Reserved]



Subpart K--Bulk Drug Formulations for Repacking or for Manufacturing Use



Sec. 440.1080a  Sterile penicillin G potassium buffered.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G potassium, buffered, is a 
dry mixture of penicillin G potassium and the buffer sodium citrate in a 
quantity not less than 4.0 percent and not more than 5.0 percent by 
weight of its total solids. It may contain citric acid in a quantity not 
more than 0.15 percent of its total solids in place of a corresponding 
amount of sodium citrate. The sodium citrate and citric acid used in 
making the batch must conform to all U.S.P. specifications. It is so 
purified and dried that:
    (i) Its potency is not less than 1,355 units and not more than 1,595 
units per milligram.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its loss on drying is not more than 1.5 percent.
    (vi) Its pH is not less than 6.0 and not more than 8.5.
    (vii) Its penicillin G content is not less than 76.3 percent and not 
more than 89.8 percent.
    (viii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, pH, penicillin G content, and crystallinity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 600 milligrams.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Dissolve an accurately weighed sample in sufficient 1.0 percent 
potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration.
    (ii) Assay procedures. Assay for potency by any of the following 
methods; however, the results obtained from the iodometric assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 unit of penicillin 
G per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (c) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the prescribed concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 20,000 units of penicillin G per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.

[[Page 587]]

    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 60 milligrams per milliliter.
    (7) Penicillin G content. Proceed as directed in Sec. 436.316 of 
this chapter.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[42 FR 59872, Nov. 22, 1977, as amended at 45 FR 22922, Apr. 4, 1980; 50 
FR 19918, 19919, May 13, 1985]



Sec. 440.1081a  Sterile penicillin G sodium, buffered.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G sodium, buffered, is a dry 
mixture of penicillin G sodium and the buffer sodium citrate in a 
quantity not less than 4.0 percent and not more than 5.0 percent by 
weight of its total solids. It may contain citric acid in a quantity not 
more than 0.15 percent of its total solids in place of a corresponding 
amount of sodium citrate. The sodium citrate and citric acid used in 
making the batch must conform to all U.S.P. specifications. It is so 
purified and dried that:
    (i) Its potency is not less than 1,420 units and not more than 1,667 
units per milligram.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its loss on drying is not more than 1.5 percent.
    (vi) Its pH is not less than 6.0 and not more than 7.5.
    (vii) Its penicillin G content is not less than 80 percent and not 
more than 93.8 percent.
    (viii) It is crystalline.
    (ix) It passes the test for heat stability if it does not show a 
loss of more than 10 percent of its original potency.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, pH, penicillin G content, crystallinity, and 
heat stability.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 600 milligrams.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Dissolve an accurately weighed sample in sufficient 1.0 percent 
potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration.
    (ii) Assay procedures. Assay for potency by any of the following 
methods; however, the results obtained from the iodometric assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 unit of penicillin 
G per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, diluting an aliquot of the stock solution with solution 1 to 
the prescribed concentration.
    (c) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the prescribed concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 20,000 units of penicillin G per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 60 milligrams per milliliter.
    (7) Penicillin G content. Proceed as directed in Sec. 436.316 of 
this chapter.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (9) Heat stability. Proceed as directed in Sec. 436.214 of this 
chapter.
[42 FR 59873, Nov. 22, 1977; 43 FR 2393, Jan. 17, 1978, as amended at 45 
FR 22922, Apr. 4, 1980; 50 FR 19918, 19919, May 13, 1985]

[[Page 588]]



PART 441--PENEM ANTIBIOTIC DRUGS--Table of Contents




                          Subpart A--Bulk Drugs

Sec.
441.20a  Sterile imipenem monohydrate.

                          Subpart B  [Reserved]

                   Subpart C--Injectable Dosage Forms

441.220  Imipenem monohydrate-cilastatin sodium injectable dosage forms.
441.220a  Sterile imipenem monohydrate-cilastatin sodium.
441.220b  Imipenem monohydrate-cilastatin sodium for injection.

    Authority:  21 U.S.C. 357.



                          Subpart A--Bulk Drugs



Sec. 441.20a  Sterile imipenem monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Imipenem monohydrate is the monohydrate 
form of [5R-[5, 6, (R*)]]-6-(1-hydroxyethyl)-3-[[2-
[(iminomethyl) amino]ethyl]thio]-7-oxo-1-azabicyclo[3.2.0]-hept-2-ene-2-
carboxylic acid. It is a white to tan colored powder. It is so purified 
and dried that:
    (i) Its potency is not less than 900 micrograms and not more than 
1,050 micrograms of imipenem per milligram on an anhydrous basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its loss on drying is not less than 5.0 percent and not more 
than 8.0 percent.
    (v) Its specific rotation in an aqueous solution containing 5 
milligrams of imipenem per milliliter at 25  deg.C is +85 deg. to 
+95 deg. on an anhydrous basis.
    (vi) It gives a positive identity test.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, specific rotation, identity, and 
crystallinity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (b) For sterility testing: 20 packages, each containing equal 
portions of approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using a column heater which will maintain 
a 50  deg.C column temperature, and ultraviolet detection system 
operating at a wavelength of 254 nanometers, a column packed with 
microparticulate (3 to 10 micrometers in diameter) reversed phase 
packing material such as octyl or octadecyl hydrocarbon bonded silicas, 
a flow rate of 2.0 milliliters per minute, and a known injection volume 
of 10 microliters. Reagents, working standard and sample solutions, 
system suitability requirements, and calculations are as follows:
    (i) Reagents--(a) Phosphate buffer, 0.001M. Dissolve 272 milligrams 
of monobasic potassium phosphate in 1,800 milliliters of deionized 
water. Adjust the pH to 6.8 with 0.5N sodium hydroxide or dilute 
phosphoric acid. Dilute to 2,000 milliliters with deionized water and 
filter prior to use.
    (b) Mobile phase. Dissolve 2.0 grams of 1-hexanesulfonic acid, 
sodium salt in 800 milliliters of phosphate buffer, 0.001M. Adjust the 
pH to 6.8 with 0.5N sodium hydroxide or dilute phosphoric acid and 
dilute to 1,000 milliliters with phosphate buffer, 0.001M. Filter and 
degas the mobile phase just prior to its introduction into the 
chromatograph pumping system.
    (c) 0.1 Percent bicarbonate solution. Dissolve 50 milligrams of 
sodium bicarbonate in 40 milliliters of phosphate buffer, 0.001M, and 
dilute to 50 milliliters with phosphate buffer, 0.001M.
    (d) 0.9 Percent saline solution. Dissolve 9.0 grams of sodium 
chloride in 800 milliliters of deionized water and dilute to 1.0 liter 
with deionized water.
    (ii) Preparations of working standard and sample solutions--(a) 
Working standard solution. Accurately weigh approximately 25 milligrams 
of the imipenem working standard into a 50-milliliter volumetric flask. 
Immediately prior to

[[Page 589]]

analysis, add 10 milliliters of 0.9 percent saline solution and 1 
milliliter of 0.1 percent bicarbonate solution. Add phosphate buffer, 
0.001M, and shake until dissolved. Sonicate, if necessary, but for no 
longer than 1 minute. Dilute to volume with phosphate buffer, 0.001M, to 
obtain a solution containing approximately 500 micrograms of imipenem 
per milliliter. Mix well and inject immediately.
    (b) Sample solution. Dissolve an accurately weighed portion 
(approximately 25 milligrams) of the sample with 10 milliliters of 0.9 
percent saline solution and 1 milliliter of 0.1 percent bicarbonate 
solution in a 50-milliliter volumetric flask. Dilute the sample solution 
to volume with phosphate buffer, 0.001M, to obtain a solution containing 
500 micrograms of imipenem per milliliter (estimated).
    (iii) System suitability requirements--
    (a) Tailing factor. The tailing factor (T) is satisfactory if it is 
not more than 1.5 at 10 percent of peak height in lieu of 5 percent of 
peak height.
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 600 theoretical plates for a 30-
centimeter column.
    (c) Resolution. The resolution (R) between the peaks for thienamycin 
and imipenem is satisfactory if it is not less than 2.0.
    (d) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SRinpercent) of 5 replicate 
injections is satisfactory if it is not more than 2.0 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.216(b) of this chapter. Alternate chromatographic 
conditions are acceptable provided reproducibility and resolution are 
comparable to the system. However, the sample preparation described in 
paragraph (b)(1)(ii)(b) of this section should not be changed.
    (iv) Calculations. Calculate the micrograms of imipenem per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.146

where:
Au=Area of the imipenem peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the imipenem peak in the chromatogram of the 
          imipenem working standard;
Ps=Anhydrous imipenem activity in the imipenem working 
          standard solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of sample solution; 
          and
L=Percent loss on drying of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 5.0 milligrams of imipenem per milliliter, 
except inject 10 milliliters per kilogram of rabbit weight.
    (4) Loss on drying. Proceed as directed in Sec. 436.200(i) of this 
chapter.
    (5) Specific rotation. Dilute an accurately weighed sample with 
sufficient pH 7.0 phosphate buffer to give a concentration of 
approximately 5.0 milligrams of imipenem per milliliter. Proceed as 
directed in Sec. 436.210 of this chapter, using a 1.0-decimeter 
polarimeter tube. To prepare the pH 7.0 phosphate buffer, transfer 5 
grams of monobasic potassium phosphate and 11 grams of dibasic potassium 
phosphate to a 1.0-liter volumetric flask. Dissolve and dilute to volume 
with distilled water.
    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation described in paragraph (b)(2) of that 
section.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[51 FR 11573, Apr. 4, 1986; 51 FR 16517, May 5, 1986, as amended at 55 
FR 11582, Mar. 29, 1990; 59 FR 8133, Feb. 18, 1994]



                          Subpart B--[Reserved]



                   Subpart C--Injectable Dosage Forms



Sec. 441.220  Imipenem monohydrate-cilastatin sodium injectable dosage forms.



Sec. 441.220a  Sterile imipenem monohydrate-cilastatin sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Imipenem monohydrate-

[[Page 590]]

cilastatin sodium is a dry mixture of imipenem monohydrate and 
cilastatin sodium packaged for dispensing. Its potency is satisfactory 
if it contains not less than 400 micrograms of imipenem and not less 
than 400 micrograms of cilastatin per milligram. Its imipenem content is 
satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of milligrams of imipenem that it is represented 
to contain. Its cilastatin content is satisfactory if it is not less 
than 90 percent and not more than 115 percent of the number of 
milligrams of cilastatin that it is represented to contain. It is 
sterile. It is nonpyrogenic. Its loss on drying is not more than 3.5 
percent. When reconstituted as directed in the labeling, its pH is not 
less than 6.0 and not more than 7.5. The imipenem monohydrate used 
conforms to the standards prescribed by Sec. 441.20a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The imipenem monohydrate used in making the batch for potency, 
sterility, pyrogens, loss on drying, specific rotation, identity, and 
crystallinity.
    (B) The batch for imipenem potency, cilastatin potency, imipenem 
content, cilastatin content, sterility, pyrogens, loss on drying, and 
pH.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The imipenem monohydrate used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Imipenem and cilastatin potency 
and content. Determine the potency of the sample in micrograms per 
milligram of both imipenem and cilastatin and the milligrams of both 
imipenem and cilastatin per container. Proceed as directed in 
Sec. 441.20a(b)(1), preparing the cilastatin reference standard 
solution, the sample solution and calculating the imipenem and 
cilastatin potency and content as follows:
    (i) Cilastatin reference standard. Accurately weigh approximately 25 
milligrams of the cilastatin reference standard into a 50-milliliter 
volumetric flask. Immediately prior to analysis, add 10 milliliters of a 
0.9 percent saline solution and 1.0 milliliter of a 0.1 percent 
bicarbonate solution. Add phosphate buffer, 0.001M, and shake until 
dissolved. Sonicate, if necessary, but no longer than 1 minute. Dilute 
to volume with phosphate buffer, 0.001M, to obtain a solution containing 
approximately 500 micrograms of cilastatin per milliliter. Mix well and 
inject immediately.
    (ii) Preparation of sample solutions--(A) Imipenem and cilastatin 
potency (micrograms of imipenem and cilastatin per milligram). Remove 
the metal seal from each of 10 containers and determine the gross weight 
in grams. Dissolve and wash out the entire contents of each container 
with a 0.9 percent saline solution into an appropriate size volumetric 
flask to give a concentration of 5 milligrams per milliliter each of 
imipenem and cilastatin. Further dilute with phosphate buffer, 0.001M, 
to obtain a solution containing 500 micrograms each of imipenem and 
cilastatin per milliliter (estimated). Wash each stopper and container 
with small quantities of acetone or methanol three times being careful 
not to wet the container labeling. Allow the containers to air dry about 
3 hours or to constant weight. Weigh each container and stopper to 
determine tare weight in grams.
    (B) Imipenem and cilastatin content (milligrams of imipenem and 
cilastatin per container). Reconstitute the sample as directed in the 
labeling, except use a 0.9 percent saline solution as the reconstituting 
fluid. Then, using a suitable hypodermic needle and syringe, remove all 
of the withdrawable contents if it is represented as a single-dose 
container; or, if the labeling specifies the amount of potency in a 
given volume of the resultant preparation, remove an accurately measured 
representative portion from each container. Accurately dilute

[[Page 591]]

the solution thus obtained in a suitable volumetric flask with 
sufficient 0.9 percent saline solution to obtain a stock solution 
containing about 2,500 micrograms of imipenem and 2,500 micrograms of 
cilastatin per milliliter. Transfer a 10-milliliter aliquot of this 
solution to a 50-milliliter volumetric flask and dilute to volume with 
phosphate buffer, 0.001M, to obtain a solution containing 500 micrograms 
of imipenem and 500 micrograms of cilastatin per milliliter (estimated).
    (iii) Calculations--(A) Imipenem and cilastatin potency. Calculate 
the micrograms of imipenem and cilastatin per milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.147

where:
AU=Area of the imipenem or cilastatin peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
AS=Area of the imipenem or cilastatin peak in the 
          chromatogram of the imipenem or cilastatin working standard;
PS=Anhydrous imipenem or cilastatin activity in the 
          respective working standard solution in micrograms per 
          milliliter;
d=Dilution factor of the 10 samples; and
WS=Net contents of 10 containers in grams (gross weight of 10 
          containers in grams--tare weight of 10 containers in grams).

    (B) Imipenem and cilastatin content. Calculate the imipenem or 
cilastatin content of the container as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.148

where:
AU=Area of the imipenem or cilastatin peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
AS=Area of the imipenem or cilastatin peak in the 
          chromatogram of the imipenem or cilastatin working standard;
PS=Anhydrous imipenem or cilastatin activity in the imipenem 
          or cilastatin working standard solution in micrograms per 
          milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in Sec. 436.20(e)(1).
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 5.0 milligrams of imipenem per milliliter 
except inject 10 milliliters per kilogram of rabbit weight.
    (4) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter.
[58 FR 26670, May 4, 1993]



Sec. 441.220b  Imipenem monohydrate-cilastatin sodium for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Imipenem monohydrate-cilastatin sodium is 
a dry mixture of imipenem monohydrate, cilastatin sodium, and sodium 
bicarbonate packaged for dispensing. Its potency is satisfactory if it 
contains not less than 400 micrograms of imipenem and not less than 400 
micrograms of cilastatin per milligram. Its imipenem content is 
satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of milligrams of imipenem that it is represented 
to contain. Its cilastatin content is satisfactory if it is not less 
than 90 percent and not more than 115 percent of the number of 
milligrams of cilastatin that it is represented to contain. It is 
sterile. It is nonpyrogenic. Its loss on drying is not more than 3.5 
percent. When reconstituted as directed in the labeling, its pH is not 
less than 6.5 and not more than 8.5. The imipenem monohydrate used 
conforms to the standards prescribed by Sec. 441.20a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The imipenem monohydrate used in making the batch for potency, 
sterility, pyrogens, loss on drying, specific rotation, identity, and 
crystallinity.
    (b) The batch for imipenem potency, cilastatin potency, imipenem 
content, cilastatin content, sterility, pyrogens, loss on drying, and 
pH.

[[Page 592]]

    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research
    (a) The imipenem used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Imipenem and cilastatin potency 
and content. Determine the potency of the sample in micrograms per 
milligram of both imipenem and cilastatin and the milligrams of both 
imipenem and cilastatin per container. Proceed as directed in 
Sec. 441.20a(b)(1) of this chapter, preparing the cilastatin reference 
standard solution, the sample solution and calculating the imipenem and 
cilastatin potency and content as follows:
    (i) Cilastatin reference standard. Accurately weigh approximately 25 
milligrams of the cilastatin reference standard into a 50-milliliter 
volumetric flask. Immediately prior to analysis, add 10 milliliters of 
0.9 percent saline solution and 1 milliliter of 0.1 percent bicarbonate 
solution. Add phosphate buffer, 0.001M, and shake until dissolved. 
Sonicate, if necessary, but for no longer than 1 minute. Dilute to 
volume with phosphate buffer, 0.001M, to obtain a solution containing 
approximately 500 micrograms of cilastatin per milliliter. Mix well and 
inject immediately.
    (ii) Preparation of sample solutions--(a) Imipenem and cilastatin 
potency (micrograms of imipenem and cilastatin per milligram). Remove 
the metal seal from each of 10 containers and determine gross weight in 
grams. Dissolve and wash out the entire contents of each container with 
0.9 percent saline into an appropriate size volumetric flask to give a 
concentration of 5 milligrams per milliliter each of imipenem and 
cilastatin. Further dilute with phosphate buffer, 0.001 M, to obtain a 
solution containing 500 micrograms each of imipenem and cilastatin per 
milliliter (estimated). Wash each stopper and container with small 
quantities of acetone or methanol three times being careful not to wet 
the container labeling. Allow the containers to air dry about 3 hours or 
to constant weight. Weigh each container and stopper to determine tare 
weight in grams.
    (b) Imipenem and cilastatin content (milligrams of imipenem and 
cilastatin per container). Reconstitute the sample as directed in the 
labeling, except use 0.9 percent saline solution as the reconstituting 
fluid. Then, using a suitable hypodermic needle and syringe, remove all 
of the withdrawable contents if it is represented as a single-dose 
container; or, if the labeling specifies the amount of potency in a 
given volume of the resultant preparation, remove an accurately measured 
representative portion from each container. Accurately dilute the 
solution thus obtained in a suitable volumetric flask with sufficient 
0.9. percent saline solution to obtain a stock solution containing about 
2,500 micrograms of imipenem and 2,500 micrograms of cilastatin per 
milliliter. Transfer a 10-milliliter aliquot of this solution to a 50-
milliliter volumetric flask and dilute to volume with phosphate buffer, 
0.001M, to obtain a solution containing 500 micrograms of imipenem and 
500 micrograms of cilastatin per milliliter (estimated).
    (iii) Calculations--(a) Calculate the micrograms of imipenem and 
cilastatin per milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.149

where:
Au=Area of the imipenem or cilastatin peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
As=Area of the imipenem or cilastatin peak in the 
          chromatogram of the imipenem or cilastatin acid working 
          standard;
Ps=Anhydrous imipenem or cilastatin activity in the 
          respective working standards solutions in micrograms per 
          milliliter;
d=Dilution factor for the 10 samples; and
Ws=Net contents of 10 containers in grams (gross weight of 10 
          containers in grams-tare weight of 10 containers in grams).

    (b) Calculate the imipenem or cilastatin content of the container as 
follows:

[[Page 593]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.150


where:
Au=Area of the imipenem or cilastatin peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
As=Area of the imipenem or cilastatin peak in the 
          chromatogram of the imipenem or cilastatin working standard:
Ps=Anhydrous imipenem or cilastatin activity in the imipenem 
          or cilastatin working standard solution in micrograms per 
          milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 5.0 milligrams of imipenem per milliliter 
except inject 10 milliliters per kilogram of rabbit weight.
    (4) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter.
[51 FR 11573, Apr. 4, 1986; 51 FR 22275, June 19, 1986, as amended at 55 
FR 11582, Mar. 29, 1990. Redesignated at 58 FR 26669, May 4, 1993]



PART 442--CEPHA ANTIBIOTIC DRUGS--Table of Contents




                          Subpart A--Bulk Drugs

Sec.
442.4  Cefaclor monohydrate.
442.6  Cefadroxil monohydrate.
442.7  Cefadroxil hemihydrate.
442.8a  Sterile cefamandole nafate.
442.9a  Sterile cefamandole sodium.
442.10  Cefazolin.
442.11a  Sterile cefazolin sodium.
442.12  Cefoperazone sodium.
442.12a  Sterile cefoperazone sodium.
442.13  Cefotaxime sodium.
442.13a  Sterile cefotaxime sodium.
442.14  Cefoxitin sodium.
442.14a  Sterile cefoxitin sodium.
442.15  Cefixime trihydrate.
442.16  Ceftazidime pentahydrate.
442.16a  Sterile ceftazidime pentahydrate.
442.17  Ceftizoxime sodium.
442.17a  Sterile ceftizoxime sodium.
442.18  Cefuroxime sodium.
442.18a  Sterile cefuroxime sodium.
442.19  Cefuroxime axetil.
442.20a  Sterile cefonicid sodium.
442.21  Cephaloglycin dihydrate.
442.22a  Sterile cefmenoxime hydrochloride.
442.23a  Sterile cephaloridine.
442.25a  Sterile cephalothin sodium.
442.27  Cephalexin monohydrate.
442.28  Cephalexin hydrochloride monohydrate.
442.29a  Sterile cephapirin sodium.
442.40  Cephradine.
442.40a  Sterile cephradine.
442.41  Cephradine dihydrate.
442.50a  Sterile ceforanide.
442.52  Cefotetan.
442.53a  Sterile cefotetan disodium.
442.54  Cefpodoxime proxetil.
442.55  Ceftriaxone sodium.
442.55a  Sterile ceftriaxone sodium.
442.58a  Sterile cefotiam dihydrochloride.
442.60  Cefpiramide.
442.69  Cefmetazole.
442.70a  Sterile cefmetazole sodium.
442.80  Cefprozil.

                      Subpart B--Oral Dosage Forms

442.104  Cefaclor monohydrate oral dosage forms.
442.104a  Cefaclor monohydrate capsules.
442.104b  Cefaclor monohydrate for oral suspension.
442.106  Cefadroxil monohydrate oral dosage forms.
442.106a  Cefadroxil monohydrate capsules.
442.106b  Cefadroxil monohydrate tablets.
442.106c  Cefadroxil monohydrate for oral suspension.
442.107  Cefadroxil hemihydrate oral dosage forms.
442.107a  Cefadroxil hemihydrate capsules.
442.107b  Cefadroxil hemihydrate tablets.
442.115  Cefixime trihydrate oral dosage forms.
442.115a  Cefixime trihydrate for oral suspension.
442.115b  Cefixime trihydrate tablets.
442.119  Cefuroxime axetil oral dosage forms.
442.119a  Cefuroxime axetil tablets.
442.119b  Cefuroxime axetil for oral suspension.
442.121  Cephaloglycin dihydrate oral dosage forms.
442.121a  Cephaloglycin dihydrate capsules.
442.121b  Cephaloglycin dihydrate for oral suspension.
442.127  Cephalexin monohydrate oral dosage forms.
442.127a  Cephalexin monohydrate tablets.
442.127b  Cephalexin monohydrate capsules.
442.127c  Cephalexin monohydrate for oral suspension.
442.128  Cephalexin hydrochloride monohydrate tablets.
442.140a  Cephradine for oral suspension.
442.140b  Cephradine capsules.
442.140c  Cephradine tablets.
442.141  Cephradine dihydrate capsules.

[[Page 594]]

442.154  Cefpodoxime proxetil oral dosage forms.
442.154a  Cefpodoxime proxetil tablets.
442.154b  Cefpodoxime proxetil granules for oral suspension.
442.180  Cefprozil oral dosage forms.
442.180a  Cefprozil tablets.
442.180b  Cefprozil for oral suspension.

                   Subpart C--Injectable Dosage Forms

442.208  Cefamandole nafate for injection.
442.209  Cefamandole sodium for injection.
442.211  Cefazolin sodium injectable dosage forms.
442.211a  Sterile cefazolin sodium.
442.211b  Cefazolin sodium injection.
442.212  Cefoperazone injectable dosage forms.
442.212a  Sterile cefoperazone sodium.
442.212b  Cefoperazone sodium injection.
442.213  Cefotaxime injectable dosage forms.
442.213a  Sterile cefotaxime sodium.
442.213b  Cefotaxime sodium injection.
442.214  Cefoxitin injectable dosage forms.
442.214a  Sterile cefoxitin sodium.
442.214b  Cefoxitin sodium injection.
442.216  Ceftazidime injectable dosage forms.
442.216a  Ceftazidime pentahydrate for injection.
442.216b  Ceftazidime sodium injection.
442.217  Ceftizoxime injectable dosage forms.
442.217a  Sterile ceftizoxime sodium.
442.217b  Ceftizoxime sodium injection.
442.218  Cefuroxime injectable dosage forms.
442.218a  Sterile cefuroxime sodium.
442.218b  Cefuroxime sodium injection.
442.220  Sterile cefonicid sodium.
442.222  Cefmenoxime hydrochloride for injection.
442.223  Sterile cephaloridine.
442.225  Cephalothin sodium injectable dosage forms.
442.225a  Sterile sodium cephalothin.
442.225b  Cephalothin sodium injection.
442.225c  Cephalothin sodium for injection.
442.229  Sterile cephapirin sodium.
442.240  Cephradine injectable dosage forms.
442.240a  Cephradine for injection.
442.240b  Sterile cephradine.
442.250  Ceforanide for injection.
442.253  Cefotetan injectable dosage forms.
442.253a  Sterile cefotetan disodium.
442.253b  Cefotetan sodium injection.
442.255  Ceftriaxone injectable dosage forms.
442.255a  Sterile ceftriaxone sodium.
442.255b  Ceftriaxone sodium injection.
442.258  Cefotiam dihydrochloride for injection.
442.260  Cefpiramide sodium for injection.
442.270  Cefmetazole injectable dosage forms.
442.270a  Sterile cefmetazole sodium.
442.270b  Cefmetazole sodium injection.

    Authority:  21 U.S.C. 357.

    Source:  39 FR 19040, May 30, 1974, unless otherwise noted.



                          Subpart A--Bulk Drugs



Sec. 442.4  Cefaclor monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefaclor monohydrate is the monohydrate 
form of (6R, 7R)-7-[(R)-2-amino-2-phenylacetamido]-3-chloro-8-oxo-5-
thia-1-azabicyclo [4.2.0]oct-2-ene-2-carboxylic acid. It is so purified 
and dried that:
    (i) Its potency is not less than 860 micrograms and not more than 
1,050 micrograms of cefaclor per milligram on an ``as is'' basis.
    (ii) Its moisture content is not less than 3.0 percent and not more 
than 8.0 percent.
    (iii) Its pH in an aqueous suspension containing 25 milligrams per 
milliliter is not less than 3.0 and not more than 4.5.
    (iv) It gives a positive identity test.
    (v) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the hydroxylamine 
colorimetric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to obtain a stock solution 
containing 1 milligram of cefaclor per milliliter (estimated). Further 
dilute an aliquot of the stock solution with solution 1 to the reference 
concentration of 5.0 micrograms of cefaclor per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, except prepare the working 
standard and sample solutions

[[Page 595]]

and calculate the cefaclor content as follows:
    (a) Preparation of working standard solution. Dissolve and dilute an 
accurately weighed portion of the cefaclor working standard in 
sufficient 0.1M potassium phosphate buffer, pH 4.5 (as described in 
Sec. 436.101(a)(4) of this chapter) to obtain a concentration of 1 
milligram of cefaclor per milliliter.
    (b) Preparation of sample solution. Dissolve an accurately weighed 
portion of the sample in sufficient 0.1M potassium phosphate buffer, pH 
4.5 (as described in Sec. 436.101(a)(4) of this chapter) to obtain a 
concentration of 1 milligram of cefaclor per milliliter.
    (c) Calculations. Calculate the cefaclor content in micrograms per 
milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.151

where:

Au = Absorbance of sample solution;
Pa = Potency of working standard solution in micrograms per 
          milliliter;
As = Absorbance of working standard solution;
Wu = Milligrams of sample per milliliter of sample solution.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous suspension containing 25 milligrams per milliliter.
    (4) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation described in paragraph (b)(2) of that 
section.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[46 FR 3832, Jan. 16, 1981]



Sec. 442.6  Cefadroxil monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefadroxil monohydrate is 7-[D-2-amino-
2(p-hydroxy- phenyl)acetamido] - 3 - methyl - 8 - oxo- 5-thia-1-
azabicyclo[4.2.0] oct-2-ene-2-carboxylic acid monohydrate. It is so 
purified and dried that:
    (i) Its potency is not less than 900 micrograms and not more than 
1,050 micrograms of cefadroxil per milligram on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 4.2 percent and not more 
than 6.0 percent.
    (iv) Its pH in an aqueous solution containing 50 milligrams per 
milliliter is not less than 4.0 and not more than 6.0.
    (v) When calculated on an anhydrous basis, its absorptivity at 264 
nanometers is not less than 95 percent and not more than 104 percent of 
that of the cefadroxil standard similarly treated and corrected for 
potency.
    (vi) It passes the identity test.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, absorptivity, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the hydroxylamine 
colorimetric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to give a stock solution of 
convenient concentration. Further dilute an aliquot of the stock 
solution with solution 1 to the reference concentration of 20 micrograms 
of cefadroxil per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay for cefadroxil. Proceed as 
directed in Sec. 442.40(b)(1)(ii) of this chapter, except prepare the 
working standard and sample solutions and calculate the potency of the 
sample as follows:
    (a) Preparation of working standard solutions. Dissolve and dilute 
an accurately weighed portion of the cefadroxil working standard in 
sufficient distilled water to obtain a stock

[[Page 596]]

solution of convenient concentration. Further dilute an aliquot of this 
solution with distilled water to a concentration of 1 milligram of 
cefadroxil per milliliter.
    (b) Preparation of sample solutions. Dissolve an accurately weighed 
portion of the sample in sufficient distilled water to obtain a stock 
solution of convenient concentration. Further dilute an aliquot of this 
solution with distilled water to a concentration of 1 milligram of 
cefadroxil per milliliter (estimated).
    (c) Calculate the potency of the sample in micrograms per milligram 
as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.152

where:

Au=Absorbance of sample solution;
Pa=Potency of working standard solution in micrograms per 
          milliliter;
As=Absorbance of working standard solution;
Wu=Milligrams of sample per milliliter of sampe solution;
m=Percent moisture in sample.

    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 50 milligrams per milliliter.
    (5) Absorptivity. Determine the absorbance of the sample and 
standard solutions in the following manner: Dissolve accurately weighed 
portions of approximately 50 milligrams each of the sample and standard 
in 250 milliliters of distilled water. Transfer a 10-milliliter aliquot 
to a 100-milliliter volumetric flask and dilute to volume with distilled 
water. Using a suitable spectrophotometer and distilled water as the 
blank, determine the absorbance of each solution at 264 nanometers. 
Determine the percent absorptivity of the sample relative to the 
absorptivity of the standard using the following calculations:

Percent relative absorptivity=[Absorbance of sample  x  milligrams 
          standard  x  potency of standard in micrograms per milligram 
          x  10]/[Absorbance of standard  x  milligrams sample  x  (100-
          m)]

where:
m=Percent moisture in the samples.

    (6) Identity. Using the sample and working standard solutions 
prepared as described in paragraph (b)(5) of this section and a suitable 
spectrophotometer, record the ultraviolet spectrum from 220 to 340 
nanometers. The spectrum of the sample compares qualitatively with that 
of the cefadroxil working standard.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[43 FR 20977, May, 16, 1978; 43 FR 27180, June 23, 1978, as amended at 
50 FR 19919, May 13, 1985]



Sec. 442.7  Cefadroxil hemihydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefadroxil hemihydrate is 7-[D-2-amino-
2(p-hydroxyphenyl)acetamido]-3-methyl-8-oxo-5-thia-1-
azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid hemihydrate. It is so 
purified and dried that:
    (i) Its potency is not less than 900 micrograms and not more than 
1,050 micrograms of cefadroxil activity per milligram on an anhydrous 
basis.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 2.4 percent and not more 
than 4.5 percent.
    (iv) The pH of an aqueous solution containing 50 milligrams per 
milliliter is not less than 4.0 and not more than 6.0.
    (v) When calculated on an anhydrous basis, its absorptivity at 264 
nanometers is not less than 95 percent and not more than 104 percent of 
that of the cefadroxil standard similarly treated and corrected for 
potency.
    (vi) It passes the identity test.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cefadroxil potency, 
moisture, pH, absorptivity, identity, and crystallinity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and

[[Page 597]]

Research: 10 packages, each containing approximately 500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the hydroxylamine 
colorimetric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to give a stock solution of 
convenient concentration. Further dilute an aliquot of the stock 
solution with solution 1 to the reference concentration of 20 micrograms 
of cefadroxil per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay for cefadroxil. Proceed as 
directed in Sec. 442.40(b)(1)(ii), except prepare the working standard 
and sample solutions and calculate the potency of the sample as follows:
    (A) Preparation of working standard solutions. Dissolve and dilute 
an accurately weighed portion of the cefadroxil working standard in 
sufficient distilled water to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of this solution with distilled 
water to a concentration of 1 milligram of cefadroxil per milliliter.
    (B) Preparation of sample solutions. Dissolve an accurately weighed 
portion of the sample in sufficient distilled water to obtain a stock 
solution of convenient concentration. Further dilute an aliquot of this 
solution with distilled water to a concentration of 1 milligram of 
cefadroxil per milliliter (estimated).
    (C) Calculations. Calculate the potency of the sample in micrograms 
per milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.153

where:
AU=Absorbance of sample solution;
AS=Absorbance of working standard solution;
Pa=Potency of working standard solution in micrograms per 
          milliliter;
WU=Milligrams of sample per milliliter of sample solution; 
          and
m=Percent moisture content of the sample.

    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 50 milligrams per milliliter.
    (5) Absorptivity. Determine the absorbance of the sample and 
standard solutions in the following manner: Dissolve accurately weighed 
portions of approximately 50 milligrams each of the sample and standard 
in 250 milliliters of distilled water. Transfer a 10-milliliter aliquot 
to a 100-milliliter volumetric flask and dilute to volume with distilled 
water. Using a suitable spectrophotometer and distilled water as the 
blank, determine the absorbance of each solution at 264 nanometers. 
Determine the percent absorptivity of the sample relative to the 
absorptivity of the standard using the following calculations:
Percent relative absorptivity = [Absorbance of sample X milligrams 
    standard X potency of standard in micrograms per milligram X 10]/
    [Absorbance of standard X milligrams sample X (100-m)]

where:
m = Percent moisture in the samples.

    (6) Identity. Using the sample and working standard solutions 
prepared as described in paragraph (b)(5) of this section and a suitable 
spectrophotometer, record the ultraviolet spectrum from 220 to 340 
nanometers. The spectrum of the sample compares qualitatively with that 
of the cefadroxil working standard.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[59 FR 8857, Feb. 24, 1994]



Sec. 442.8a  Sterile cefamandole nafate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile cefamandole nafate is the sodium 
salt of 7-D-mandelamido-3-[[(1-methyl-1H- tetrazol-5-yl)thio]methyl]-8-
oxo-5-thia-1-azabicyclo[4.2.0]-oct-2-ene-2-carboxylate formate (ester). 
It is so purified and dried that:
    (i) Its potency is not less than 810 micrograms and not more than 
1,000

[[Page 598]]

micrograms of cefamandole per milligram on an anhydrous basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 2.0 percent.
    (vi) Its pH in an aqueous solution containing 100 milligrams per 
milliliter is not less than 3.5 and not more than 7.0.
    (vii) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, and identity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (b) For sterility testing: 20 packages, each containing equal 
portions of approximately 250 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the hydroxylamine 
colorimetric assay shall be conclusive.
    (i) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, except use the cefamandole 
working standard.
    (ii) Polarographic assay. Proceed as directed in Sec. 436.324 of 
this chapter.
    (iii) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), to obtain a concentration of 1 
milligram of cefamandole per milliliter (estimated). Hydrolyze this 
solution in a 37 deg. C constant temperature water bath for 60 minutes. 
Further dilute a portion of the hydrolyzed solution with 1 percent 
potassium phosphate buffer, pH 6.0 (solution 1), to the reference 
concentration of 2.0 micrograms of cefamandole per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cefamandole per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the mineral oil mull prepared as described in paragraph (b)(2) of 
that section.
[47 FR 32708, June 1, 1982, as amended at 50 FR 19919, May 13, 1985]



Sec. 442.9a  Sterile cefamandole sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile cefamandole sodium is 5-thia-1-
azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid, 7-
[(hydroxyphenylacetyl)amino]-3-[[(1-methyl-1H-tetrazol-5-
yl)thio]methyl]-8-oxo-, monosodium salt [6R-[6, 
7(R*)]]-. It is so purified and dried that:
    (i) Its cefamandole content is not less than 860 micrograms and not 
more than 1,000 micrograms of cefamandole per milligram on an anhydrous 
basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 3.0 percent.
    (vi) Its pH in an aqueous solution containing 100 milligrams per 
milliliter is not less than 3.5 and not more than 7.0.
    (vii) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cefamandole 
content, sterility, pyrogens, moisture, pH, and identity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.

[[Page 599]]

    (b) For sterility testing: 20 packages, each containing equal 
portions of approximately 250 milligrams.
    (b) Tests and methods of assay--(1) Cefamandole content. Proceed as 
directed in Sec. 436.324 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cefamandole per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the mineral oil mull prepared as described in paragraph (b)(2) of 
that section.
[47 FR 20756, May 14, 1982, as amended at 50 FR 19919, May 13, 1985]



Sec. 442.10  Cefazolin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefazolin is 3-[[(5-methyl-1, 3, 4-
thiadiazol-2-yl)-thio]methyl]-7-[2-(1H-tetrazol-1-yl) acetamido]-3-
cephem-4-carboxylic acid. It is so purified and dried that:
    (i) Its cefazolin content is not less than 950 micrograms and not 
more than 1,030 micrograms of cefazolin per milligram calculated on an 
anhydrous basis.
    (ii) Its moisture content is not more than 2 percent.
    (iii) Its heavy metals content is not more than 20 parts per 
million.
    (iv) It gives a positive identity test for cefazolin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cefazolin content, 
moisture, heavy metals, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: Nine packages, each containing approximately 
500 milligrams, and one package containing approximately 5 grams.
    (b) Tests and methods of assay--(1) Cefazolin content. Proceed as 
directed in Sec. 436.342 of this chapter.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
    (4) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the cefazolin working standard.
[48 FR 33479, July 22, 1983, as amended at 55 FR 11582, Mar. 29, 1990]



Sec. 442.11a  Sterile cefazolin sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile cefazolin sodium is the sodium 
salt of 3-[[(5-methyl - 1,3,4 - thiadiazol - 2 - yl) - thio]methyl] - 7 
- [2 - (1H - tetrazol-1-yl)acetamido] - 3 - cephem - 4 - carboxylic 
acid. It is so purified and dried that:
    (i) Its potency is not less than 850 micrograms and not more than 
1050 micrograms of cefazolin per milligram calculated on an anhydrous 
basis. If it is packaged for dispensing, its cefazolin content is 
satisfactory if it contains not less than 90 percent and not more than 
115 percent of the number of milligrams of cefazolin that it is 
represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 6 percent.
    (vi) Its pH in an aqueous solution containing 100 milligrams of 
cefazolin per milliliter is not less than 4.5 and not more than 6.0.
    (vii) The specific rotation in a 0.1M sodium bicarbonate solution 
containing 50 milligrams of cefazolin per milliliter at 25 deg. C. is 
-17 deg.plus-minus7 deg. calculated on an anhydrous basis.
    (viii) It gives a positive identity test for cefazolin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 600]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, specific rotation, and identity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 9 packages, each containing 
approximately 500 milligrams, and 1 package containing approximately 5 
grams.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers, except if each contains less than 1.0 gram, a minimum of 24 
immediate containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Dissolve an accurately weighed sample in sufficient 1.0 percent 
potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration; also if it is packaged for 
dispensing, reconstitute as directed in the labeling. Then using a 
suitable hypodermic needle and syringe, remove all of the withdrawable 
contents. Dilute with sufficient solution 1 to give a stock solution of 
convenient concentration.
    (ii) Assay procedure. Use either of the following methods; however, 
the results obtained from the microbiological agar diffusion assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 micrograms of 
cefazolin per milliliter (estimated).
    (b) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter, preparing the working standard solution as 
follows: Dissolve an accurately weighed portion of approximately 30 
milligrams of cefazolin working standard in 3 milliliters of 10 percent 
potassium phosphate buffer, pH 6.0 (solution 6), and further dilute with 
solution 1 to the final concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cefazolin per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams of cefazolin per 
milliliter.
    (7) Specific rotation. Proceed as directed in Sec. 436.210 of this 
chapter, using a solution containing 50 milligrams of cefazolin per 
milliliter in 0.1M sodium bicarbonate and a polarimeter tube 1.0 
decimeter in length. Calculate the specific rotation on an anhydrous 
basis.
    (8) Identity. Using a 0.002 percent solution of the sample in 0.1M 
sodium bicarbonate solution and a suitable spectrophotometer, record the 
ultraviolet spectrum from 220 to 340 nanometers. The spectrum compares 
qualitatively to that of the cefazolin working standard similarly 
tested.
[39 FR 19040, May 30, 1974, as amended at 42 FR 18059, Apr. 5, 1977; 50 
FR 19919, May 13, 1985]



Sec. 442.12  Cefoperazone sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefoperazone sodium is the sodium salt of 
(6R, 7R)-7-[(R)-2-(4-ethyl-2,3-dioxo-1-piperazinecarboxamido)-2-(p-
hydroxyphenyl)acetamido]-3-[[(1-methyl-1H-tetrazol-5-yl)thio]methyl]-8-
oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylate. It is a white to 
off-white crystalline powder or a lyophilized powder. It is so purified 
and dried that:
    (i) Its cefoperazone content is not less than 870 micrograms and not 
more than 1,015 micrograms of cefoperazone per milligram on an anhydrous 
basis.
    (ii) Its moisture content is not more than 5.0 percent, except if it 
is the

[[Page 601]]

lyophilized powder, its moisture content is not more than 2.0 percent.
    (iii) The pH of an aqueous solution containing 250 milligrams per 
milliliter is not less than 4.5 and not more than 6.5.
    (iv) It passes the identity test if the retention times of the 
sample and working standard agree within 3.0 percent.
    (v) It is crystalline, except if it is the lyophilized powder.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cefoperazone 
content, moisture, pH, identity, and crystallinity (if it is not the 
lyophilized powder).
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Cefoperazone content. Proceed as 
directed in Sec. 436.338 of this chapter.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 250 milligrams per milliliter.
    (4) Identity. From the high-performance liquid chromatograms of the 
sample and the cefoperazone working standard determined as directed in 
paragraph (b)(1) of this section, calculate the adjusted retention times 
of the cefoperazone in the sample and standard solutions as follows:

Adjusted retention time of cefoperazone=t-ta
where:
t=Retention time measured from point of injection into the chromatograph 
          until the maximum of the cefoperazone sample or working 
          standard peak appears on the chromatogram; and
ta=Retention time measured from point of injection into the 
          chromatograph until the maximum of nonretarded solute appears 
          in the chromatogram.


The sample and the cefoperazone working standard should have 
corresponding adjusted cefoperazone retention times within 
3.0 percent.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[51 FR 36688, Oct. 15, 1986, as amended at 55 FR 11583, Mar. 29, 1990]



Sec. 442.12a  Sterile cefoperazone sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile cefoperazone sodium is the sodium 
salt of (6R, 7R)-7-[(R)-2-(4-ethyl-2,3-dioxo-1-piperazinecarboxamido)-2-
(p- hydroxyphenyl)acetamido]-3-[[(1-methyl-1H-tetrazol-5-
yl)thio]methyl]-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylate. 
It is a white to off-white crystalline powder or it may be a lyophilized 
powder. It is so purified and dried that:
    (i) If the cefoperazone sodium is not packaged for dispensing, its 
cefoperazone content is not less than 870 micrograms and not more than 
1,015 micrograms of cefoperazone per milligram on an anhydrous basis. If 
the cefoperazone sodium is packaged for dispensing, its cefoperazone 
content is not less than 870 micrograms and not more than 1,015 
micrograms of cefoperazone per milligram on an anhydrous basis and also, 
each container contains not less than 90 percent and not more than 120 
percent of the number of milligrams of cefoperazone that it is 
represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 5.0 percent, except if it 
is the lyophilized powder, its moisture content is not more than 2.0 
percent.
    (v) Its pH in an aqueous solution containing 250 milligrams per 
milliliter is not less than 4.5 and not more than 6.5.
    (vi) It passes the identity test if the retention times of the 
sample and working standard agree within 3 percent.
    (vii) It is crystalline, except if it is the lyophilized powder, it 
is not crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 602]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cefoperazone 
content, sterility, pyrogens, moisture, pH, identity, and crystallinity 
(if it is not the lyophilized powder).
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) If the batch is packaged for repacking or for manufacturing use:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (2) For sterility testing: 20 packages, each containing equal 
portions of approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers of the batch.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Cefoperazone content. Proceed as 
directed in Sec. 436.338 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 10 milligrams of cefoperazone per 
milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 250 milligrams per milliliter.
    (6) Identity. From the high-pressure liquid chromatograms of the 
sample and the cefoperazone working standard determined as directed in 
paragraph (b)(1) of this section, calculate the adjusted retention times 
of the cefoperazone in the sample and standard solutions as follows:

Retention time of cefoperazone=ts-tu

where:
ts=Retention time of working standard measured from point of 
          injection into the chromatograph until the peak maximum 
          appears on the chromatogram; and
tu=Retention time of sample measured from point of injection 
          into the chromatograph until the peak maximum appears on the 
          chromatogram.

    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[48 FR 790, Jan. 7, 1983; 43 FR 7439, Feb. 22, 1983; 48 FR 28250, June 
21, 1983, as amended at 55 FR 11583, Mar. 29, 1990]



Sec. 442.13  Cefotaxime sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefotaxime sodium is the sodium salt of 
5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid, 3-
[(acetyloxy)methyl]-7-[[(2-amino-4-thiazolyl)  
(methoxyimino)acetyl]amino]-8-ox-o,[6R-[6 alpha, 7 beta(Z)]]-. It is so 
purified and dried that:
    (i) Its potency is not less than 855 micrograms and not more than 
1,002 micrograms of cefotaxime per milligram on an anhydrous basis.
    (ii) Its moisture content is not more than 6.0 percent.
    (iii) Its pH in an aqueous solution is not less than 4.5 and not 
more than 6.5.
    (iv) It gives a positive identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research; 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the hydroxylamine 
colorimetric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1.0 percent 
potassium phosphate buffer, pH 6.0 (solution 1), to obtain a stock 
solution of convenient concentration. Further dilute an aliquot of the 
stock solution

[[Page 603]]

with solution 1 to the reference concentration of 2.0 micrograms of 
cefotaxime per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii), except prepare the working standard and sample 
solutions and calculate the potency of the sample as follows:
    (a) Preparation of the working standard solution. Dissolve and 
dilute an accurately weighed portion of the cefotaxime working standard 
in sufficient distilled water to obtain a concentration of 1 milligram 
of cefotaxime per milliliter.
    (b) Preparation of sample solution. Dissolve and dilute an 
accurately weighed portion of the sample in sufficient distilled water 
to obtain a concentration of 1 milligram of cefotaxime per milliliter 
(estimated).
    (c) Calculation. Calculate the cefotaxime content in micrograms per 
milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.154

where:

Au=Absorbance of sample solution;
Pa=Potency of working standard solution in micrograms per 
          milliliter;
As=Absorbance of working standard solution; and
Wu=Milligrams of sample per milliliter of sample solution.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (4) Identity. Proceed as directed in Sec. 436.323 of this chapter, 
except prepare spotting solutions as follows: Prepare solutions of the 
sample and working standard, each containing 1 milligram of cefotaxime 
per milliliter in distilled water.
[50 FR 45109, Oct. 30, 1985, as amended at 55 FR 11583, Mar. 29, 1990]



Sec. 442.13a  Sterile cefotaxime sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefotaxime sodium is the sodium salt of 
5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid, 3-
[(acetyloxy)methyl]-7-[[(2-amino-4-thiazolyl) 
(methoxyimino)acetyl]amino]-8-oxo-,[6R-[6, 7(Z)]]-. It 
is so purified and dried that:
    (i) Its potency is not less than 855 micrograms and not more than 
1,002 micrograms of cefotaxime per milligram on an anhydrous basis. If 
it is packaged for dispensing, its content is satisfactory if it is not 
less than 90 percent and not more than 110 percent of the number of 
milligrams of cefotaxime that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 6.0 percent.
    (vi) Its pH in an aqueous solution is not less than 4.5 and not more 
than 6.5.
    (vii) It gives a positive identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, and identity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use as an 
ingredient in the manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 1 gram.
    (2) For sterility testing: 20 packages, each containing 
approximately 1 gram.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the hydroxylamine 
colorimetric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately

[[Page 604]]

weighed sample in sufficient 1.0 percent potassium phosphate buffer, pH 
6.0 (solution 1), to obtain a stock solution of convenient 
concentration; also, if it is packaged for dispensing, reconstitute as 
directed in the labeling. Then using a suitable hypodermic needle and 
syringe, remove all of the withdrawable contents if it is represented as 
a single-dose container; or, if the labeling specifies the amount of 
potency in a given volume of the resultant preparation, remove an 
accurately measured representative portion from each container. Dilute 
with solution 1 to obtain a stock solution of convenient concentration. 
Further dilute an aliquot of the stock solution with solution 1 to the 
reference concentration of 2.0 micrograms of cefotaxime per milliliter 
(estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, except prepare the working 
standard and sample solutions and calculate the potency of the sample as 
follows:
    (a) Preparation of the working standard solution. Dissolve and 
dilute an accurately weighed portion of the cefotaxime working standard 
in sufficient distilled water to obtain a concentration of 1 milligram 
of cefotaxime per milliliter.
    (b) Preparation of sample solution. Dissolve and dilute an 
accurately weighed portion of the sample in sufficient distilled water 
to obtain a concentration of 1 milligram of cefotaxime per milliliter 
(estimated).
    (c) Calculations--(1) Calculate the cefotaxime content in micrograms 
per milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.155

where:

Au = Absorbance of sample solution;
Pa = Potency of working standard solution in micrograms per 
          milliliter;
As = Absorbance of working standard solution;
Wu = Milligrams of sample per milliliter of sample solution.

    (2) Calculate the cefotaxime content of the single-dose vial as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.156

where:

Au = Absorbance of sample solution;
Pa = Potency of working standard solution in micrograms per 
          milliliter;
As = Absorbance of working standard solution;
d = Dilution factor of the sample.

    (3) Calculate the cefotaxime content of the multiple-dose vial as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.157

where:

Au = Absorbance of sample solution;
Pa = Potency of working standard solution in micrograms per 
          milliliter;
As = Absorbance of working standard solution;
d = Dilution factor of the sample;
n = Volume of sample solution assayed.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cefotaxime per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (7) Identity. Proceed as directed in Sec. 436.323 of this chapter, 
except prepare spotting solutions as follows: Prepare solutions of the 
sample and working standard, each containing 1 milligram of cefotaxime 
per milliliter in distilled water.
[46 FR 25606, May 8, 1981, as amended at 50 FR 19919, May 13, 1985]



Sec. 442.14  Cefoxitin sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefoxitin sodium is the sodium salt of 3-
(hydroxymethyl)-7-methoxy-8-oxo-7-[2-(2-thienyl)acetamido]-5-
thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylic

[[Page 605]]

acid carbamate (ester). It is so purified and dried that:
    (i) Its cefoxitin content is not less than 850 micrograms and not 
more than 1,000 micrograms of cefoxitin per milligram.
    (ii) Its moisture content is not more than 2.0 percent.
    (iii) Its pH in an aqueous solution containing 100 milligrams per 
milliliter is not less than 4.2 and not more than 7.0.
    (iv) It gives a positive identity test.
    (v) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cefoxitin content, 
moisture, pH, identity, and crystallinity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Cefoxitin content. Proceed as 
directed in Sec. 436.347 of this chapter, preparing the working standard 
and sample solutions and calculating the cefoxitin content as follows:
    (i) Working standard solution. Dissolve an accurately weighed 
portion of the cefoxitin working standard with water to obtain a 
solution containing 1 milligram of cefoxitin per milliliter.
    (ii) Sample solution. Dissolve an accurately weighed portion of the 
sample with water to obtain a solution containing 1 milligram of 
cefoxitin per milliliter (estimated).
    (iii) Calculations. Calculate the micrograms of cefoxitin per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.158

where:
Au=Area of the cefoxitin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefoxitin peak in the chromatogram of the 
          cefoxitin working standard;
Ps=Cefoxitin activity in the cefoxitin working standard 
          solution in micrograms per milliliter; and
Cu=Milligrams of sample per milliliter of sample solution 
          (estimated).

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the titration procedure described in paragraph (e)(1) of that 
section, except add about 25 milliliters of methanol in lieu of solvent 
A to a dry titrating vessel and proceed as directed in titration 
procedure 1.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (4) Identity. Proceed as directed in Sec. 436.326 of this chapter.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[49 FR 47827, Dec. 7, 1984, as amended at 55 FR 11583, Mar. 29, 1990]



Sec. 442.14a  Sterile cefoxitin sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefoxitin sodium is the sodium salt of 3-
(hydroxymethyl) - 7-methoxy - 8 - oxo - 7 - [2 - (2 - thienyl) 
acetamido] - 5 - thia - 1 - azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid 
carbamate (ester). It is so purified and dried that:
    (i) Its potency is not less than 850 micrograms and not more than 
1,000 micrograms of cefoxitin per milligram. If it is packaged for 
dispensing, its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
cefoxitin that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 2.0 percent.
    (vi) Its pH in an aqueous solution is not less than 4.2 and not more 
than 7.0.
    (vii) It gives a positive identity test.
    (viii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:

[[Page 606]]

    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, identity, and crystallinity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use as an 
ingredient in the manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 1 gram.
    (2) For sterility testing: 20 packages, each containing 
approximately 1 gram.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.347 of this chapter, preparing the working standard and sample 
solutions and calculating the cefoxitin content as follows:
    (i) Working standard solution. Dissolve an accurately weighed 
portion of the cefoxitin working standard with distilled water to obtain 
a solution containing 1 milligram of cefoxitin per milliliter.
    (ii) Sample solutions. Dissolve an accurately weighed portion of the 
sample with distilled water to obtain a solution containing 1 milligram 
of cefoxitin per milliliter (estimated); and also if it is packaged for 
dispensing, reconstitute as directed in the labeling. Then using a 
suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is represented as a single-dose container; or, if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute with distilled water to obtain a 
solution containing 1 milligram of cefoxitin per milliliter (estimated).
    (iii) Calculations--(a) Calculate the cefoxitin content in 
micrograms per milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.159

where:
Au=Area of the cefoxitin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefoxitin peak in the chromatogram of the 
          cefoxitin working standard;
Ps=Cefoxitin activity in the cefoxitin working standard 
          solution in micrograms per milliliter; and
Cu=Milligrams of sample per milliliter of sample solution 
          (estimated).

    (b) Calculate the cefoxitin content of the vial as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.160
    
where:
Au=Area of the cefoxitin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefoxitin peak in the chromatogram of the 
          cefoxitin working standard;
Ps=Cefoxitin activity in the cefoxitin working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cefoxitin per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the titration procedure described in paragraph (e)(1) of that 
section, except add about 25 milliliters of methanol in lieu of solvent 
A to a dry titrating vessel and proceed as directed in titration 
procedure 1.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (7) Identity. Proceed as directed in Sec. 436.326 of this chapter, 
preparing the sample as follows: Prepare a solution containing about 2.5 
milligrams of cefoxitin per milliliter in distilled water.

[[Page 607]]

    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[44 FR 10374, Feb. 20, 1979, as amended at 50 FR 19919, May 13, 1985; 51 
FR 27532, Aug. 1, 1986]



Sec. 442.15  Cefixime trihydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefixime trihydrate is the trihydrate 
form of [6R-[6 , 7B(Z)]]-7-[[(2-amino-4-thiazolyl) 
[(carboxymethoxy)imino]acetyl ]amino]-3-ethenyl-8-oxo-5-thia-
azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid. It is so purified and 
dried that:
    (i) Its potency is not less than 950 micrograms and not more than 
1,030 micrograms of cefixime activity per milligram, on an anhydrous 
basis.
    (ii) Its moisture content is not less than 9.0 percent and not more 
than 12.0 percent.
    (iii) The pH of an aqueous solution containing the equivalent of 0.7 
milligram per milliliter is not less than 2.6 and not more than 4.1.
    (iv) It is crystalline.
    (v) The specific rotation in a 2.0 percent sodium bicarbonate 
solution containing 10.0 milligrams of cefixime per milliliter at 25 
deg.C is between -75 deg. and -88 deg. calculated on an anhydrous basis.
    (vi) It gives a positive identity test for cefixime.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results for tests and assays on the batch for potency, moisture, 
pH, crystallinity, specific rotation, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams, and 1 package containing approximately 5 grams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using an ultraviolet detection system 
operating at a wavelength of 254 nanometers, and a column (typically 3 
centimeters  x  4.6 millimeters) packed with a 3-micron octadecyl 
hydrocarbon bonded silica or equivalent at ambient temperature. 
Reagents, working standard, test and sample solutions, system 
suitability requirements, and calculations are as follows:
    (i) Reagents--(A) Phosphoric acid solution. Add 10 milliliters of 
concentrated phosphoric acid to 90 milliliters of water.
    (B) Tetrabutylammonium hydroxide solution. Dilute 25 milliliters of 
0.4M tetrabutylammonium hydroxide solution to 1,000 milliliters with 
water. Adjust the pH to 7.0 with phosphoric acid solution.
    (C) Mobile phase. Add 775 milliliters of the tetrabutylammonium 
hydroxide solution to 225 milliliters of acetonitrile. Filter the mobile 
phase through a suitable glass filter or equivalent which is capable of 
removing particulate contamination greater than 0.5 micron in diameter. 
Degas the mobile phase just prior to its introduction into the 
chromatograph.
    (D) 0.1M Phosphate buffer, pH 7.0. Add 6.8 milliliters of 
concentrated phosphoric acid to 300 milliliters of water. Adjust the pH 
to 7.0 with 10N sodium hydroxide. Dilute to 1,000 milliliters with 
water.
    (ii) Preparation of working standard, test and sample solutions--(A) 
Working standard solution. Dissolve an accurately weighed portion of the 
cefixime standard with sufficient 0.1M phosphate buffer, pH 7.0, to 
obtain a solution of known concentration containing approximately 2 
milligrams of cefixime activity per milliliter. Further dilute 
quantitatively to a final concentration of 0.2 milligram of cefixime 
activity per milliliter in 0.1 M phosphate buffer, pH 7.0. Prepare the 
working standard solution just prior to its introduction into the 
chromatograph.
    (B) System suitability test solution. Dissolve an accurately weighed 
portion of cefixime working standard in distilled water to obtain a 
solution containing approximately 1.0 milligram of cefixime activity per 
milliliter. Heat this solution at 95  deg.C (in an oil bath) for 45 
minutes. This procedure allows the (E)-isomer of cefixime to be 
generated in situ. Prepare the test solution

[[Page 608]]

just prior to its introduction into the chromatograph.
    (C) Sample solution. Accurately weigh approximately 100 milligrams 
of the sample into a 50-milliliter volumetric flask. Dilute to volume 
with 0.1M phosphate buffer, pH 7.0, to obtain a stock solution 
containing approximately 2 milligrams of cefixime activity per 
milliliter. Mix well. Immediately prior to chromatography, further 
dilute 10 milliliters of stock solution to 100 milliliters with 0.1 M 
phosphate buffer, pH 7.0 to obtain a solution containing 0.2 milligram 
of cefixime activity per milliliter (estimated).
    (iii) System suitability requirements--(A) Asymmetry factor. 
Calculate the asymmetry factor (As), measure data point that 
is 10 percent of the cefixime peak height from the baseline, as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.161

where:
a=Horizontal distance from point of ascent to point of maximum peak 
          height; and
b=horizontal distance from the point of maximum peak height to point of 
          descent.

    The asymmetry factor (As) is satisfactory if it is not 
less than 0.85 and not more than 1.5.
    (B) Efficiency of the column. From the number of theoretical plates 
(n) calculated as described in Sec. 436.216(c)(2) of this chapter 
calculate the reduced plate height (hr) for the cefixime peak 
as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.162

where:
L=Length of the column in centimeters;
n=number of theoretical plates; and
(dp)=Average diameter of the particles in the column in 
          micrometers.


The absolute efficiency (hr) is satisfactory if it is not 
more than 15 for the cefixime peak.
    (C) Resolution. The resolution (R) between the peak for cefixime and 
the peak for the (E)-isomer of cefixime (generated in situ) is not less 
than 1.1.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR) in percent) of five replicate 
injections is satisfactory if not more than 2.0 percent
    (E) Capacity factor (k). Calculate the capacity factor (k) for 
cefixime as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.163

where:
tr=Retention time of solute; and
tm=Retention time of solvent or unretained substance, 
          calculated as follows:
          [GRAPHIC] [TIFF OMITTED] TR01JA93.164
          
where:
D=Column diameter in centimeters;
L=Column length in centimeters;
0.75=Average total column porosity; and
F=Flow rate in milliliters per minute.

    The capacity factor (k) for cefixime is satisfactory if it is not 
less than 5 and not more than 11.
    If the system suitability requirements have been met, then proceed 
as described in Sec. 436.216(b) of this chapter. Alternate 
chromatographic conditions are acceptable provided that the system 
suitability parameters are met. However, the sample preparation 
described in paragraph (b)(1)ii)(C) of this section should not be 
changed.
    (iv) Calculations. Calculate the micrograms of cefixime anhydrous 
free acid per milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.165

where:
Au=Area of the cefixime peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefixime peak in the chromatogram of the 
          cefixime working standard;
Ps=Cefixime activity in the cefixime working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of sample solution; 
          and
m=Percent moisture content of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.

[[Page 609]]

    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 0.7 milligram per milliliter.
    (4) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (5) Specific rotation. Dissolve and dilute an accurately weighed 
sample with sufficient 2 percent sodium bicarbonate to obtain a 
concentration of approximately 10 milligrams of cefixime per milliliter. 
Proceed as directed in Sec. 436.210 of this chapter, using a 1.0-
decimeter polarimeter tube. Calculate the specific rotation on the 
anhydrous basis.
    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a potassium bromide disc containing 0.5 percent of cefixime. 
Dissolve 5 to 6 milligrams of cefixime in 2 milliliters of methanol. 
Triturate to insure solution. Evaporate the solvent to dryness and using 
the dried sample, prepare the potassium bromide disc.
[53 FR 24257, June 28, 1988; 53 FR 26712, July 14, 1988; 54 FR 47205, 
Nov. 13, 1989]



Sec. 442.16  Ceftazidime pentahydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ceftazidime pentahydrate is pyridinium, 
1-[[7-[[(2-amino-4-thiazolyl) [1-carboxy-1-methylethoxy)imino]acetyl]-
amino]-2-carboxy-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-en-3-yl]methyl]-, 
hydroxide, inner salt, [6R-[6,7(Z)]]-, pentahydrate. 
It is so purified and dried that:
    (i) Its potency is not less than 950 micrograms and not more than 
1,020 micrograms of ceftazidime activity per milligram on an anhydrous 
basis.
    (ii) Its loss on drying is not less than 13.0 percent and not more 
than 15.0 percent.
    (iii) The pH of an aqueous solution containing 5 milligrams of 
ceftazidime per milliliter is not less than 3.0 and not more than 4.0.
    (iv) It is crystalline.
    (v) It gives a positive identity test for ceftazidime.
    (vi) Its high molecular weight polymer content is not more than 0.05 
percent.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, crystallinity, identity, and high molecular weight polymer 
content.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 442.16a(b)(1).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 5 milligrams of ceftazidime per 
milliliter.
    (4) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (5) Identity. The high performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the ceftazidime working standard.
    (6) High molecular weight polymer content. Proceed as directed in 
Sec. 442.16a(b)(8).
[54 FR 40652, Oct. 3, 1989]



Sec. 442.16a  Sterile ceftazidime pentahydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile ceftazidime pentahydrate is 
pyridinium, 1-[[7-[[(2-amino-4-thiazolyl)[(1-carboxy-1-
methylethoxy)imino]acetyl]amino]-2-carboxy-8-oxo-5-thia-1-
azabicyclo[4.2.0]oct-2-en-3-yl]methyl]-, hydroxide, inner salt, [6R-
[6, 7(Z)]]-, pentahydrate. It is so purified and dried 
that:
    (i) Its potency is not less than 950 micrograms and not more than 
1,020 micrograms of ceftazidime activity per milligram on an anhydrous 
basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its loss on drying is not less than 13.0 and not more than 15.0 
percent.

[[Page 610]]

    (v) Its pH in an aqueous solution containing 5 milligrams of 
ceftazidime per milliliter is not less than 3.0 and not more than 4.0.
    (vi) It is crystalline.
    (vii) It gives a positive identity test for ceftazidime.
    (viii) Its high molecular weight polymer content is not more than 
0.05 percent.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain.
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, pH, crystallinity, identity, and high 
molecular weight polymer content.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (b) For sterility testing: One package containing approximately 6 
grams of a composite sample.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.356 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 254 nanometers, a column 
packed with microparticulate (3 to 10 micrometers in diameter) reversed 
phase packing material such as hexyl, octyl, or octaldecyl hydrocarbon 
bonded silicas, a flow rate of 2.0 milliliters per minute, and a known 
injection volume of 20 microliters. Reagents, working standard and 
sample solutions, system suitability requirements, and calculations are 
as follows:
    (i) Reagents--(a) Phosphate buffer, pH 7.0. Dissolve 42.59 grams of 
sodium phosphate, dibasic anhydrous and 27.22 grams of potassium 
phosphate, monobasic, in water and dilute to 1,000 milliliters.
    (b) Mobile phase. Mix 40 milliliters of acetonitrile and 200 
milliliters of phosphate buffer, pH 7.0, and dilute to 2,000 milliliters 
with water. Filter the mobile phase through a suitable glass fiber 
filter or equivalent that is capable of removing particulate 
contamination to 1 micron in diameter. Degas the mobile phase just prior 
to its introduction into the chromatograph pumping system.
    (ii) Preparation of working standard and sample solutions--(a) 
Working standard solution. Accurately weigh ceftazidime working standard 
equivalent to approximately 100 milligrams of the ceftazidime activity 
into a 100-milliliter volumetric flask containing 10 milliliters of 
phosphate buffer, pH 7.0. Shake until dissolved. Dilute to volume with 
water to obtain a stock solution containing approximately 1,000 
micrograms of ceftazidime activity per milliliter. Mix well. Immediately 
prior to chromatography, further dilute 5 milliliters of stock solution 
to 50 milliliters with water to obtain a solution containing 100 
micrograms of ceftazidime activity per milliliter.
    (b) Sample solution. Accurately weigh approximately 115 milligrams 
of the sample into a 100-milliliter volumetric flask containing 10 
milliliters of phosphate buffer, pH 7.0. Shake until dissolved. Dilute 
to volume with water to obtain a stock solution containing approximately 
1,000 micrograms of ceftazidime per milliliter. Mix well. Immediately 
prior to chromatography, further dilute 5 milliliters of stock solution 
to 50 milliliters with water to obtain a solution containing 100 
micrograms of ceftazidime activity per milliliter (estimated).
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.5 at 5 
percent of peak height.
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,500 theoretical plates.
    (c) Resolution. The resolution (R) between the peak for ceftazidime 
and its nearest eluting impurity is satisfactory if it is not less than 
2.0.
    (d) Coefficient of variation. The coefficient of variation (SR 
in percent) of five replicate injections is satisfactory if it is not 
more than 1.0 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.356(b) of this chapter. Alternate chromatographic 
conditions are acceptable provided reproducibility

[[Page 611]]

and resolution are provided comparable to the system. However, the 
sample preparation described in paragraph (b)(1)(ii)(b) of this section 
should not be changed.
    (iv) Calculations. Calculate the micrograms of ceftazidime per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.166

where:
Au=Area of the ceftazidime peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the ceftazidime peak in the chromatogram of the 
          ceftazidime working standard;
Ps=Ceftazidime activity in the ceftazidime working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of sample solution; 
          and
m=Percent loss on drying content of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
dissolve the sample in approximately 200 milliliters of diluting fluid 
H.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(i) of this chapter, 
using a solution containing 80 milligrams of ceftazidime per milliliter.
    (4) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 5 milligrams of ceftazidime per 
milliliter.
    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (7) Identity. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the ceftazidime working standard.
    (8) High molecular weight polymer content. Proceed as directed in 
Sec. 436.360 of this chapter, using a constant temperature between 20 
and 25  deg.C, an ultraviolet detection system operating at a wavelength 
of 235 nanometers, a column packed with a hydrophilic gel for gel 
permeation chromatography (such as Fractogel TSK HW-40(F), Merck) or 
equivalent, a flow rate of 1.0 milliliter per minute, and a known 
injection volume of 100 microliters. Reagents, working standard and 
sample solutions, system suitability requirements, and calculations are 
as follows:
    (i) Reagents--(a) Mobile phase. Adjust a 0.1M solution of potassium 
phosphate, dibasic, to pH 7.0plus-minus0.1 with phosphoric 
acid.
    (b) Blue dextran system suitability test solution. Prepare a 
solution in mobile phase containing 100 micrograms per milliliter of 
blue dextran (with a mean molecular weight of approximately 2,000,000).
    (ii) Preparation of working standard and sample solutions--(a) 
Working standard solution. Accurately weigh high molecular weight 
polymer working standard equivalent to approximately 400 micrograms of 
high molecular weight polymer into a 100-milliliter volumetric flask and 
add 80 milliliters of mobile phase. Shake until dissolved and dilute to 
volume with mobile phase to obtain a solution containing approximately 4 
micrograms of high molecular weight polymer per milliliter. Store the 
solution at ambient temperature and inject into the chromatograph within 
one hour of preparation.
    (b) Sample solution. Accurately weigh approximately 400 milligrams 
of the sample into a 100-milliliter volumetric flask and add 80 
milliliters of mobile phase. Shake until dissolved, dilute to volume 
with mobile phase, and immediately inject the solution into the liquid 
chromatograph.
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.5 for blue 
dextran.
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,500 theoretical plates for blue 
dextran.
    (c) Coefficient of variation. The coefficient of variation (SR 
in percent) of five replicate injections of blue dextran is satisfactory 
if it is not more than 4 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.360(b) of this chapter.
    (iv) Calculations. Calculate the percent of high molecular weight 
polymer content as follows:

[[Page 612]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.167


where:
Hu=Height of the high molecular weight polymer peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
Hs=Mean height of the high molecular weight polymer peaks in 
          the chromatograms of the high molecular weight polymer working 
          standard;
Ps=High molecular weight polymer content of the high 
          molecular weight polymer working standard solution in 
          micrograms per milliliter; and
Cu=Milligrams of sample per milliliter of sample solution.
[50 FR 48399, Nov 25, 1985; 50 FR 53308, Dec. 31, 1985; 51 FR 2478, Jan. 
17, 1986, as amended at 55 FR 11583, Mar. 29, 1990]



Sec. 442.17  Ceftizoxime sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ceftizoxime sodium is the sodium salt of 
[6R-[6, 7(Z)]]-7-[[(2,3-dihydro-2-imino-4-thiazolyl) 
(methoxyimino)acetyl]amino]-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-
carboxylic acid. It is so purified and dried that:
    (i) Its ceftizoxime content is not less than 850 micrograms and not 
more than 995 micrograms of ceftizoxime per milligram on an anhydrous 
basis.
    (ii) Its moisture content is not more than 8.5 percent.
    (iii) Its pH in an aqueous solution containing 100 milligrams per 
milliliter is not less than 6.0 and not more than 8.0
    (iv) It gives a positive identity test.
    (v) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for ceftizoxime 
content, moisture, pH, identity, and crystallinity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams, and 1 package containing approximately 5 grams.
    (b) Tests and methods of assay--(1) Ceftizoxime content. Proceed as 
directed in Sec. 436.345 of this chapter, preparing the sample solution 
and calculating the ceftizoxime content as described in paragraphs 
(e)(1) and (g)(1), respectively, of that section.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (4) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section, compares 
qualitatively to that of the ceftizoxime working standard.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[49 FR 49285, Dec. 19, 1984, as amended at 55 FR 11583, Mar. 29, 1990]



Sec. 442.17a  Sterile ceftizoxime sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ceftizoxime sodium is the sodium salt of 
[6R-[6,7(Z)]]-7-[[(2,3-dihydro-2-imino-4-thiazolyl) 
(methoxyimino) acetyl]amino]-8-oxo-5-thia-1-azabicyclo [4.2.0]oct-2-ene-
2-carboxylic acid. It is so purified and dried that:
    (i) If the ceftizoxime is not packaged for dispensing, its 
ceftizoxime content is not less than 850 micrograms and not more than 
995 micrograms of ceftizoxime per milligram on an anhydrous basis. If 
the ceftizoxime is packaged for dispensing, its ceftizoxime content is 
not less than 850 micrograms and not more than 995 micrograms of 
ceftizoxime per milligram on an anhydrous basis and also, each container 
contains not less than 90 percent and not more than 115 percent of the 
number of milligrams of ceftizoxime that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 8.5 percent.
    (v) Its pH in an aqueous solution containing 100 milligrams per 
milliliter is not less than 6.0 and not more than 8.0.
    (vi) It gives a positive identity test.
    (vii) It is crystalline.

[[Page 613]]

    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for ceftizoxime 
content, sterility, pyrogens, moisture, pH, identity, and crystallinity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing at 
least 500 milligrams.
    (2) For sterility testing: 20 packages, each containing equal 
portions of approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers; or if each container contains less than 1 gram of 
ceftizoxime, a minimum of 20 immediate containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Ceftizoxime content. Proceed as 
directed in Sec. 436.345 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of ceftizoxime per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (6) Identity. From the high-pressure liquid chromatograms of the 
sample and the ceftizoxime working standard determined as directed in 
paragraph (b)(1) of this section, calculate the adjusted retention times 
of the ceftizoxime in the sample and standard solutions as follows:

Adjusted retention time of ceftizoxime=t-ta

where:
t=Retention time measured from point of injection into the chromatograph 
          until the maximum of the ceftizoxime sample or working 
          standard peak appears on the chromatogram; and
ta=Retention time measured from point of injection into the 
          chromatograph until the maximum of nonretarded solute appears 
          in the chromatogram.


The sample and the ceftizoxime working standard should have 
corresponding adjusted ceftizoxime retention times.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[48 FR 46271, Oct. 12, 1983; 48 FR 49656, Oct. 27, 1983, as amended at 
55 FR 11583, Mar. 29, 1990]



Sec. 442.18  Cefuroxime sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefuroxime sodium is the sodium salt of 
(6R,7R)-3-carbamoyloxy-methyl-7-[(2Z)-2-(2-furyl)-2-
methoxyiminoacetamido]cepha-3-em-4-carboxylic acid. It is so purified 
and dried that:
    (i) Its potency is not less than 855 micrograms and not more than 
1,000 micrograms of cefuroxime activity per milligram on an anhydrous 
basis.
    (ii) Its moisture content is not more than 3.5 percent.
    (iii) The pH of an aqueous solution containing 100 milligrams of 
cefuroxime per milliliter is not less than 6.0 and not more than 8.5.
    (iv) It gives a positive identity test for cefuroxime.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 1 
gram.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 442.343.
    (2) Moisture. Proceed as directed in Sec. 436.18a(b)(4) of this 
chapter.

[[Page 614]]

    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams of cefuroxime per 
milliliter.
    (4) Identity. Proceed as directed in Sec. 442.18a(b)(6).
[54 FR 40654, Oct. 3, 1989; 54 FR 50686, Dec. 8, 1989]



Sec. 442.18a  Sterile cefuroxime sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefuroxime sodium is the sodium salt of 
(6R, 7R )-3-carbamoyloxy-methyl-7-[(2Z )-2-(2-furyl)-2-
methoxyiminoacetamido] cepha-3-em-4-carboxylic acid. It is so purified 
and dried that:
    (i) If the cefuroxime is not packaged for dispensing, its cefuroxime 
content is not less than 855 micrograms and not more than 1,000 
micrograms of cefuroxime per milligram on an anhydrous basis. If the 
cefuroxime is packaged for dispensing, its cefuroxime content is not 
less than 855 micrograms and not more than 1,000 micrograms of 
cefuroxime per milligram on an anhydrous basis and also, each container 
contains not less than 90 percent and not more than 120 percent of the 
number of milligrams of cefuroxime that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 3.5 percent.
    (v) Its pH in an aqueous solution is not less than 6.0 and not more 
than 8.5.
    (vi) It gives a positive identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cefuroxime content, 
sterility, pyrogens, moisture, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) If the batch is packaged for repacking or for use as an 
ingredient in the manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 1 gram.
    (2) For sterility testing: 20 packages, each containing 
approximately 1 gram.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Cefuroxime content. Proceed as 
directed in Sec. 436.343 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cefuroxime per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the titration procedure described in paragraph (e)(1) of that 
section.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (6) Identity. From the high-pressure liquid chromatograms of the 
sample and the cefuroxime working standard determined as directed in 
paragraph (b)(1) of this section, calculate the adjusted retention times 
of the cefuroxime in the sample and standard solutions as follows:

Adjusted retention time of cefuroxime=t-ta

where:
t=Retention time measured from point of injection into the chromatograph 
          until the maximum of the cefuroxime sample or working standard 
          peak appears on the chromatogram; and
ta=Retention time measured from point of injection into the 
          chromatograph until the maximum of nonretarded solute appears 
          in the chromatogram.


The sample and the cefuroxime working standard should have corresponding 
adjusted cefuroxime retention times.
[48 FR 38461, Aug. 24, 1983, as amended at 55 FR 11583, Mar. 29, 1990]



Sec. 442.19  Cefuroxime axetil.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefuroxime axetil is an amorphous mixture 
of the diastereo-isomers of 5-thia-1-azabicyclo[4.2.0]oct-

[[Page 615]]

2-ene-2-carboxylic acid, 3-[[(aminocarbonyl)oxy]methyl]-7-[[2-
furanyl(methoxyimino)acetyl]amino]-8-oxo-, 1-(acetyloxy)ethyl ester, 
[6R-[6 alpha, 7 beta (Z)]]-. It is so purified and dried that:
    (i) Its potency is not less than 745 micrograms and not more than 
875 micrograms of cefuroxime per milligram on an anhydrous basis. The 
ratio of isomer A to total isomer content is not less than 0.48 and not 
more than 0.55.
    (ii) Its moisture content is not more than 1.5 percent.
    (iii) It is amorphous and not crystalline.
    (iv) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Request for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cefuroxime potency, 
isomer A ratio, moisture, crystallinity, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 278 nanometers, a 25-
centimeter by 4.6-millimeter column packed with methyl silane bonded 
silica 5 micrometers in particle size, a flow rate of 1 milliliter per 
minute, and a known injection volume of 10 microliters. Reagents, 
working standard and sample solutions, system suitability requirements, 
and calculations are as follows:
    (i) Reagents--(A) 0.2M Ammonium phosphate solution. Transfer 23.0 
grams of ammonium dihydrogen phosphate to a 1-liter volumetric flask. 
Dissolve and dilute to volume with distilled water. Mix well.
    (B) Mobile phase. Transfer 380 milliliters of methanol to a 1-liter 
volumetric flask and dilute to volume with 0.2M ammonium phosphate 
solution.
    (C) Internal standard solution. Prepare a solution containing 5.4 
milligrams of acetanilide per milliliter in methanol.
    (D) System suitability test solution. Mix 10.0 milliliters of a 
solution containing 1.2 milligrams of cefuroxime axetil working standard 
per milliliter in methanol with 5.0 milliliters of internal standard 
solution, 2.0 milliliters of a solution containing 0.3 milligram of an 
authentic sample of (RS)-1-acetoxyethyl (6R, 7R)-3-carbamoyloxymethyl-7-
[(2'Z)-2-(fur-2-yl)-2-methoxy-iminoacetamido]ceph-2-em-4-carboxylate 
(delta-2 isomers of cefuroxime axetil) per milliliter in methanol and 
1.8 milliliters of methanol. Dilute to 50 milliliters with 0.2M ammonium 
phosphate solution.
    (ii) Preparation of working standard and sample solutions--(A) 
Working standard solution. Dissolve approximately 30 milligrams of the 
cefuroxime axetil working standard, accurately weighed, in methanol and 
dilute to 25 milliliters with methanol. Immediately transfer 10.0 
milliliters of the working standard solution to a 50-milliliter 
volumetric flask. Add 5.0 milliliters of internal standard solution and 
3.8 milliliters of methanol, and dilute to volume with 0.2M ammonium 
phosphate soluton to obtain a solution containing 0.2 milligram of 
cefuroxime activity per milliliter. Store the solution under 
refrigeration no more than 8 hours.
    (B) Sample solution. Dissolve approximately 30 milligrams of the 
sample, accurately weighed, in methanol and dilute to 25 milliliters 
with methanol. Immediately transfer 10.0 milliliters of the sample 
solution to a 50-milliliter volumetric flask. Add 5.0 milliliters of 
internal standard solution and 3.8 milliliters of methanol, and dilute 
to volume with 0.2M ammonium phosphate solution to obtain a solution 
containing 0.2 milligram of cefuroxime activity per milliliter 
(estimated). Store the solution under refrigeration no more than 8 
hours.
    (iii) System suitability requirements--(A) Tailing factor. The 
tailing factor (T) is satisfactory for isomer A if it is not more than 
1.5 at 5 percent of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory

[[Page 616]]

for isomer A if it is greater than 3,000 theoretical plates.
    (C) Resolution. The resolution (R) between isomer A and isomer B of 
cefuroxime axetil is satisfactory if it is not less than 1.5 and the 
resolution (R) between isomer A and the delta-2 isomers of cefuroxime 
axetil is satisfactory if it is not less than 1.5.
    (D) Coefficent of variation. The coefficient of variation 
(SRin percent) of five replicate injections is satisfactory 
if it is not more than 2.0 percent. If the system suitability 
requirements have been met, then proceed as described in Sec. 436.216(b) 
of this chapter. Alternate chromatographic conditions are acceptable 
provided reproducibility and resolutiona recomparable to the system. 
However, the sample preparation described in paragraph (b)(1)(ii)(B) of 
this section should not be changed.
    (iv) Calculations--(A) Calculate the micrograms of cefuroxime per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.168

where:
Ru = Sum of the peak height of the cefuroxime axetil sample 
          isomer A and isomer B peaks/Peak height of the internal 
          standard;
Rs = Sum of the peak heights of the cefuroxime axetil working 
          standard isomer A and isomer B peaks/Peak height of the 
          internal standard;
Ps = Cefuroxime activity in the cefuroxime axetil working 
          standard solution in micrograms per milliliter;
Cu = Milligrams of sample per milliliter of sample solution; 
          and
m = Percent moisture content of the sample.

    (B) Calculate the ratio of isomer A to total isomer content as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.169

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the titration procedure described in paragraph (e)(1) of that 
section.
    (3) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter, except that the particles do not reveal the phenomena of 
birefringence and extinction positions on revolving the microscope 
stage.
    (4) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the mineral oil mull prepared as described in paragraph (b)(2) of 
that section.
[52 FR 42432, Nov. 5, 1987; 52 FR 43966, Nov. 17, 1987; 52 FR 45528, 
Nov. 30, 1987, as amended at 54 FR 47351, Nov. 14, 1989; 55 FR 11583, 
Mar. 29, 1990]



Sec. 442.20a  Sterile cefonicid sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile cefonicid sodium is a white to 
off-white lyophilized powder. It is so purified and dried that:
    (i) If the cefonicid sodium is not packaged for dispensing, its 
cefonicid content is not less than 832 micrograms and not more than 970 
micrograms of cefonicid per milligram on an anhydrous basis. If the 
cefonicid sodium is packaged for dispensing, its cefonicid content is 
not less than 832 micrograms and not more than 970 micrograms of 
cefonicid per milligram on an anhydrous basis and also, each container 
contains not less than 90 percent and not more than 120 percent of the 
number of milligrams of cefonicid that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 5.0 percent.
    (v) Its pH in an aqueous solution containing 50 milligrams per 
milliliter is not less than 3.5 and not more than 6.5.
    (vi) The specific rotation in a methanol solution containing 10 
milligrams of cefonicid sodium per milliliter at 25 deg. C is 
-42 deg.5 deg..
    (vii) It passes the identity test.

[[Page 617]]

    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cefonicid content, 
sterility, pyrogens, moisture, pH, specific rotation, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) If the batch is packaged for repacking or for use as an 
ingredient in the manufacture of another drug;
    (1) For all tests except sterility: 10 packages, each containing at 
least 500 milligrams.
    (2) For sterility testing: 20 packages, each containing equal 
portions of approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Cefonicid content. Proceed as 
directed in Sec. 436.350 of this chapter, using ambient temperature, an 
ultraviolet detection system operating at a wavelength of 254 
nanometers, and a column packed with octadecyl silane bonded silica 
ranging from 3 to 30 micrometers in particle size. Reagents, working 
standard and sample solutions, system suitability requirements, and 
calculations are as follows:
    (i) Reagents--(a) 0.2M Ammonium phosphate solution. Transfer 23.0 
grams of ammonium dihydrogen phosphate to a 1-liter volumetric flask. 
Dissolve and dilute to volume with distilled water. Mix well.
    (b) Mobile phase. Mix 0.2M ammonium phosphate solution : methyl 
alcohol : distilled water (1 : 2.5 : 16.5). Filter through a suitable 
filter capable of removing particulate matter to 0.5 micron in diameter. 
Degas the mobile phase just prior to its introduction into the 
chromatograph.
    (ii) Working standard and sample solutions--(a) Preparation of 
working standard solution. Prepare the working standard solution fresh 
before injection by dissolving an accurately weighed portion of the 
cefonicid working standard with sufficient mobile phase as described in 
paragraph (b)(1)(i)(b) of this section to obtain a solution containing 
approximately 20 micrograms of cefonicid per milliliter.
    (b) Preparation of sample solutions--(1) Product not packaged for 
dispensing (micrograms of cefonicid per milligram). Dissolve an 
accurately weighed portion of the sample with sufficient mobile phase as 
described in paragraph (b)(1)(i)(b) of this section to obtain a 
concentration of approximately 20 micrograms of cefonicid per 
milliliter.
    (2) Product packaged for dispensing. Determine both micrograms of 
cefonicid per milligram of the sample and milligrams of cefonicid per 
container. Use separate containers for preparation of each sample 
solution as described in paragraphs (b)(1)(ii)(b)(2) (i) and (ii) of 
this section.
    (i) Micrograms of cefonicid per milligram. Dissolve an accurately 
weighed portion of the sample with sufficient mobile phase as described 
in paragraph (b)(1)(i)(b) of this section to obtain a concentration of 
approximately 20 micrograms of cefonicid per milliliter.
    (ii) Milligrams of cefonicid per container. Reconstitute the sample 
as directed in the labeling. Then, using a suitable hypodermic needle 
and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency in a given volume of the resultant preparation, 
remove an accurately measured representative portion from each 
container. Further dilute an aliquot of the solution thus obtained with 
sufficient mobile phase to obtain a concentration of approximately 20 
micrograms of cefonicid per milliliter.
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.3 at 5 
percent of peak height.
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,500 theoretical plates.
    (c) Resolution factor. Prepare a resolution solution containing 
desacetyl cefonicid by heating a 200-microgram-

[[Page 618]]

per-milliliter solution of cefonicid working standard in mobile phase 
described in paragraph (b)(1)(i)(b) of this section, on a steam bath for 
30 minutes. Inject a known volume between 10 and 20 microliters of the 
desacetyl cefonicid containing solution in the same manner as described 
for the standard solution. The resolution factor (R) between cefonicid 
and desacetyl cefonicid is satisfactory if it is not less than 1.1.
    (d) Coefficient of variation. The coefficient of variation (SR 
in percent) of five replicate injections is satisfactory if it is not 
more than 2.0 percent.

If the system suitability parameters have been met, then proceed as 
described in Sec. 436.350(b) of this chapter.
    (iv) Calculations--(a) Calculate the micrograms of cefonicid per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.170

where:
Au=Area of the cefonicid peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefonicid peak in the chromatogram of the 
          cefonicid working standard;
Ps=Cefonicid activity in the cefonicid working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of sample solution; 
          and
m=Percent moisture content of the sample.

    (b) Calculate the cefonicid content of the container as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.171
    
where:
Au=Area of the cefonicid peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefonicid peak in the chromatogram of the 
          cefonicid working standard;
Ps=Cefonicid activity in the cefonicid working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cefonicid per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 50 milligrams per milliliter.
    (6) Specific rotation. Dissolve and dilute an accurately weighed 
sample with sufficient methanol to obtain a concentration of 
approximately 10 milligrams of cefonicid sodium per milliliter. Proceed 
as directed in Sec. 436.210 of this chapter, using a 1.0-decimeter 
polarimeter tube. Calculate the specific rotation on an anhydrous basis.
    (7) Identity. The high-performance liquid chromatogram of the 
sample, determined as directed in paragraph (b)(1) of this section, 
compares qualitatively to that of the cefonicid working standard.
[49 FR 34348, Aug. 30, 1984; 49 FR 44460, Nov. 7, 1984, as amended at 54 
FR 41824, Oct. 12, 1989; 55 FR 11583, Mar. 29, 1990]



Sec. 442.21  Cephaloglycin dihydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephaloglycin dihydrate is the dihydrate 
form of 7-(D--amino-phenylacetamido) cephalosporanic acid. It 
is a white to off-white powder. It is so purified and dried that:
    (i) Its potency is not less than 900 micrograms of cephaloglycin per 
milligram on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture is not less than 8.2 and not more than 12 
percent.
    (iv) Its pH in an aqueous suspension containing 50 milligrams per 
milliliter is not less than 3.0 and not more than 5.5.
    (v) Its cephaloglycin content is not less than 95 and not more than 
104 percent on an anhydrous basis.
    (vi) It gives a positive identity test for cephaloglycin dihydrate.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.

[[Page 619]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, cephaloglycin content, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 
sterile distilled water to give a stock solution of 100 micrograms of 
cephaloglycin per milliliter (estimated). Further dilute an aliquot of 
the stock solution with 0.1M potassium phosphate buffer, pH 4.5 
(solution 4), to the reference concentration of 10 micrograms of 
cephaloglycin per milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous suspension containing 50 milligrams per milliliter.
    (5) Cephaloglycin content. Proceed as directed in Sec. 436.213 of 
this chapter, using the titration procedure described in paragraph 
(e)(2) of that section. Calculate the cephaloglycin content as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.172

where:
A=Milliliters of perchloric acid reagent used in titrating the sample;
B=Milliliters of perchloric acid reagent used in titrating the blank;
m=Percent moisture content of the sample.

    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 0.5-percent potassium bromide disc prepared as described in 
paragraph (b)(1) of that section.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19040, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 442.22a  Sterile cefmenoxime hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefmenoxime hydrochloride is 5-thia-1-
azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid, 7-[[(2-amino-4-thiazolyl) 
(methoxyimino)acetyl]amino]-3-[[(1-methyl-1H-tetrazol-5-yl)thio]methyl]-
8-oxo-, hydrochloride (2:1), [6R-[6,7(Z)]]-. It is so 
purified and dried that:
    (i) Its cefmenoxime content is not less than 869 and not more than 
1,015 micrograms of cefmenoxime per milligram on an anhydrous basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 1.5 percent.
    (v) It passes the identity test.
    (vi) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cefmenoxime 
content, sterility, pyrogens, moisture, identity, and crystallinity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (B) For sterility testing: 1 package containing approximately 6 
grams of a composite sample.
    (b) Tests and methods of assay--(1) Cefmenoxime content. Proceed as 
directed in Sec. 436.363 of this chapter, using

[[Page 620]]

ambient temperature, an ultraviolet detection system operating at a 
wavelength of 254 nanometers, a column packed with microparticulate (3 
to 10 micrometers in diameter) reversed phase packing material such as 
octadecyl hydrocarbon bonded silicas, a flow rate not to exceed 2.0 
milliliters per minute, and a known injection volume between 10 and 20 
microliters. Reagents, working standard and sample solutions, system 
suitability requirements, and calculations are as follows:
    (i) Reagents--(A) 0.1M Phosphate buffer solution, pH 6.8. Dissolve 
6.4 grams of monobasic potassium phosphate and 18.9 grams of dibasic 
sodium phosphate in 750 milliliters of water. Adjust the pH to 6.8 with 
1N sodium hydroxide and dilute to 1,000 milliliters.
    (B) Internal standard solution. Dissolve and dilute 0.15 gram of 
phthalimide in methanol to 100 milliliters.
    (C) Mobile phase. Mix water:acetonitrile:glacial acetic acid 
(50:10:1). Filter through a suitable filter capable of removing 
particulate matter to 0.5 micron in diameter. Degas the mobile phase 
just prior to its introduction into the chromatograph.
    (ii) Preparation of working standard and sample solutions--(A) 
Working standard solution. Dissolve approximately 50 milligrams of the 
cefmenoxime working standard, accurately weighed, in 10 milliliters of 
0.1M phosphate buffer solution, pH 6.8 and dilute to 50 milliliters with 
mobile phase. Transfer 4.0 milliliters of this solution to a 50-
milliliter volumetric flask, add 20 milliliters of internal standard 
solution and dilute to volume with mobile phase to obtain a solution 
containing 80 micrograms of cefmenoxime per milliliter.
    (B) Sample solution. Dissolve approximately 50 milligrams of 
cefmenoxime sample, accurately weighed, in 10 milliliters of 0.1M 
phosphate buffer solution, pH 6.8. Dilute to 50 milliliters with mobile 
phase. Transfer 4.0 milliliters of this solution to a 50-milliliter 
volumetric flask, add 20 milliliters of internal standard solution and 
dilute to volume with mobile phase.
    (iii) System suitability requirements--(A) Tailing factor. The 
tailing factor (T) for the cefmenoxime peak is satisfactory if it is not 
more than 1.6 at 5 percent of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,200 theoretical plates for the 
cefmenoxime peak.
    (C) Resolution. The resolution (R) between the peak for cefmenoxime 
and phthalimide is satisfactory if it is not less than 2.3.
    (D) Coefficient of variation. The coefficiednt of variation (SR 
in percent) of 5 replicate injections is satisfactory if it is not more 
than 2.0 percent. If the system suitability requirements have been met, 
then proceed as described in Sec. 436.363(b) of this chapter.
    (iv) Calculations. Calculate the micrograms of cefmenoxime per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.173

where:
Ru=Area of cefmenoxime peak in the chromatogram of the 
          sample/Area of internal standard peak;
Rs=Area of the cefmenoxime peak in the chromatogram of the 
          cefmenoxime working standard/Area of internal standard peak;
Ps=Cefmenoxime activity in the cefmenoxime working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of sample solution; 
          and
m=Percent moisture content of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
in lieu of diluting fluid A use diluting fluid H.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(i) of this chapter, 
using a solution containing 60 milligrams per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the sample preparation described in paragraph (d)(4) of that 
section and the titration procedure described in paragraph (e)(3) of 
that section, except:
    (i) In lieu of 3 milliliters of anhydrous methanol solution, inject 
20 milliliters of a formamide:methanol solution (2:1) into the container 
and shake to dissolve the contents (prior to use in preparation of the 
formamide:methanol solution, dry 500

[[Page 621]]

grams of formamide over 20 grams of anhydrous sodium sulfate for 24 
hours);
    (ii) Rinse the syringe, needle, and immediate container with two 
separate 5-milliliter portions of anhydrous methanol, in lieu of one 3-
milliliter portion of anhydrous methanol; and
    (iii) In Sec. 436.201(e)(3) of this chapter, add a sufficient volume 
of the formamide:methanol solution (2:1) to cover the electrodes in the 
dry titrating vessel, in lieu of 20 milliliters of solvent A before 
starting the titration.
    (5) Identity. Using a 0.0025-percent solution of the sample in 0.1M 
phosphate buffer, pH 6.8 and a suitable spectrophotometer, record the 
ultraviolet absorption spectrum from 220 to 310 nanometers. The spectrum 
compares qualitatively to that of the cefmenoxime working standard 
similarly tested.
    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[53 FR 13402, Apr. 25, 1988; 53 FR 19368, May 27, 1988]



Sec. 442.23a  Sterile cephaloridine.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephaloridine is 7-[-(2-
thienyl)-acetamido]-3-(1-pyridyl-methyl)-3-cephem-4-carboxylic acid 
betaine. It is a white to off-white powder. It is so purified and dried 
that:
    (i) Its potency is not less than 900 micrograms of cephaloridine per 
milligram. If it is packaged for dispensing, its potency is satisfactory 
if it is not less than 90 percent and not more than 115 percent of the 
number of milligrams of cephaloridine that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its loss on drying is not more than 2.5 percent.
    (vi) Its pH in an aqueous solution is not less than 3.5 and not more 
than 6.
    (vii) The specific rotation in an aqueous solution containing 10 
milligrams of cephaloridine per milliliter at 25 deg. C. is 
+48 deg.plus-minus4 deg..
    (viii) It is crystalline.
    (ix) The ultraviolet absorption spectrum between the wavelengths of 
220 and 310 nanometers compares qualitatively to that of the 
cephaloridine working standard. The ratio of the absorbance of the 
maximum at the wavelength of 240 nanometers to that of the shoulder at 
255 nanometers is not less than 1.05 and not more than 1.17.
    (2) Labeling. It shall be labeled in accordance with the 
requirements prescribed by Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, pH, specific rotation, crystallinity, and 
identity.
    (ii) Samples of the batch:
    (a) If the batch is packaged for repacking or for use as an 
ingredient in the manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing at 
least 500 milligrams.
    (2) For sterility testing: 20 packages, each containing equal 
portions of approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 13 immediate 
containers of the batch.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Dissolve an accurately weighed sample in sufficient 1.0 percent 
potassium phosphate buffer, pH 6.0 (solution 1), for the microbiological 
agar diffusion assay, distilled water for the iodometric assay or 
hydroxylamine colorimetric assay, to give a stock solution of convenient 
concentration; also if it is packaged for dispensing, reconstitute as 
directed in the labeling. Then using a suitable hypodermic needle and 
syringe, remove all of the withdrawable contents if it is represented as 
a single-dose container; or, if the labeling specifies the amount of 
potency in a given volume of the resultant preparation, remove an 
accurately measured representative portion from each container. Dilute 
with either solution 1 or distilled water as specified above to give a 
stock solution of convenient concentration.

[[Page 622]]

    (ii) Assay procedures. Use any of the following methods; however, 
the results obtained from the microbiological agar diffusion assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 microgram of 
cephaloridine per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter. If it is packaged for dispensing, dilute an aliquot of the 
stock solution with distilled water to the prescribed concentration.

    Note: The 10 milliliters of 0.01N iodine must be added within 20 
seconds after the addition of the 2.0 milliliters of 1.2N HCl, and the 
assay should be completed within 1 hour after the sample and standard 
are first put into solution. The working standard should be dried as 
described in Sec. 436.200(a) of this chapter.

    (c) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cephaloridine per 
milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 250 milligrams of cephaloridine per 
milliliter. If it is packaged for dispensing, however, use the solution 
obtained after reconstituting the drug as directed in the labeling.
    (7) Specific rotation. Dilute an accurately weighed sample with 
sufficient distilled water to give a concentration of approximately 10 
milligrams of cephaloridine per milliliter. Proceed as directed in 
Sec. 436.210 of this chapter using a 2.0-decimeter polarimeter tube.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (9) Identity. Using a 0.0025-percent solution of the sample in water 
and a suitable spectrophotometer, record the ultraviolet absorption 
spectrum from 220 to 310 nanometers. The spectrum compares qualitatively 
to that of the cephaloridine working standard similarly tested.
[39 FR 19040, May 30, 1974, as amended at 43 FR 9800, Mar. 10, 1978; 50 
FR 19919, May 13, 1985]



Sec. 442.25a  Sterile cephalothin sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile cephalothin sodium is the sodium 
salt of the compound formed by reaction of thiophene-2-acetic acid with 
7-amino-cephalosporanic acid. The 7-amino-cephalosporanic acid is 
obtained from a kind of cephalosporin. It is so purified and dried that:
    (i) Its potency is not less than 850 micrograms of cephalothin per 
milligram on an anhydrous basis. If it is packaged for dispensing, its 
potency is satisfactory if it is not less than 90 percent and not more 
than 115 percent of the number of milligrams of cephalothin that it is 
represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its loss on drying is not more than 1.5 percent.
    (vi) Its pH in an aqueous solution is not less than 4.5 and not more 
than 7.0.
    (vii) The specific rotation in an aqueous solution containing 50 
milligrams of cephalothin sodium per milliliter at 25 deg. C is 
+129 deg.plus-minus5 deg..
    (viii) It gives a positive identity test.
    (ix) It is crystalline.
    (2) Packaging. In addition to the requirements of Sec. 432.1 of this 
chapter, if it is packaged for dispensing and is intended for both 
intravenous and intramuscular use, each vial shall contain the 
equivalent of 1 gram of cephalothin; except that if it is packaged for 
dispensing and is intended solely for intravenous use, each vial shall 
contain the equivalent of 4 grams of cephalothin.
    (3) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, if it is packaged for dispensing, each 
package shall bear on its label and labeling, the following statement: 
``After reconstitution, store in a

[[Page 623]]

refrigerator and use within 48 hours. If kept at room temperature, use 
within 6 hours.''
    (4) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of test and assay on the batch for potency, sterility, 
pyrogens, loss on drying, pH, specific rotation, crystallinity, and 
identity.
    (ii) Samples of the batch:
    (a) If the batch is packaged for repacking or for use as an 
ingredient in the manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
equal portions of approximately 500 milligrams.
    (2) For sterility testing: 20 packages, each containing equal 
portions of approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers of the batch.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Dissolve an accurately weighed sample in sufficient 1.0 percent 
potassium phosphate buffer, pH 6.0 (solution 1), for the microbiological 
agar diffusion assay, distilled water for the iodometric assay or 
hydroxylamine colorimetric assay, to give a stock solution of convenient 
concentration; also if it is packaged for dispensing, reconstitute as 
directed in the labeling. Then, using a suitable hypodermic needle and 
syringe, remove all of the withdrawable contents if it is represented as 
a single-dose container; or, if the labeling specifies the amount of 
potency in a given volume of the resultant preparation, remove an 
accurately measured representative portion from each container. Dilute 
with either solution 1 or distilled water as specified above to give a 
stock solution of convenient concentration.
    (ii) Assay procedures. Use any of the following methods; however, 
the results obtained from the microbiological agar diffusion assay shall 
be conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 microgram of 
cephalothin per milliliter (estimated).
    (b) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter. If it is packaged for dispensing, dilute an aliquot of the 
stock solution with distilled water to the prescribed concentration.

    Note: The 10 milliliters of 0.01N iodine must be added within 20 
seconds after the addition of the 2.0 milliliters of 1.2N HCl, and the 
assay should be completed within 1 hour after the sample and standard 
are first put into solution. The working standard should be dried as 
described in Sec. 436.200(a) of this chapter.

    (c) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cephalothin per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 250 milligrams per milliliter; however, 
if it is packaged for dispensing, use the solution obtained after 
reconstituting the drug as directed in the labeling.
    (7) Specific rotation. Dilute an accurately weighed sample with 
sufficient distilled water to give a concentration of approximately 50 
milligrams per milliliter. Proceed as directed in Sec. 436.210 of this 
chapter, using a 1.0-decimeter polarimeter tube and calculate the 
specific rotation on an anhydrous basis.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (9) Identity. Using a 0.0025-percent solution of the sample in water 
and a suitable spectrophotometer, record the ultraviolet absorption 
spectrum from 220 to 310 nanometers. The spectrum compares qualitatively 
to that of the

[[Page 624]]

cephalothin working standard similarly tested.
[39 FR 19040, May 30, 1974, as amended at 46 FR 46312, Sept. 18, 1981; 
48 FR 11427, Mar. 18, 1983; 50 FR 19919, May 13, 1985]



Sec. 442.27  Cephalexin monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephalexin monohydrate is the monohydrate 
form of 7-(D-alpha- amino-alpha- phenylacetamido)-3-methyl-3-cephem-4-
carboxylic acid. It is so purified and dried that:
    (i) Its potency is not less than 900 micrograms of cephalexin per 
milligram on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 4.0 nor more than 8.0 
percent.
    (iv) Its pH in an aqueous solution containing 50 milligrams per 
milliliter is not less than 3.0 nor more than 5.5.
    (v) When calculated on an anhydrous basis, its absorptivity at 262 
nanometers is not less than 95 percent and not more than 104 percent of 
that of the cephalexin standard similarly treated and corrected for 
potency.
    (vi) It gives a positive identity test.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, absorptivity, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to give a stock solution 
containing 1.0 milligram per milliliter (estimated). Further dilute an 
aliquot of the stock solution with solution 1 to the reference 
concentration of 20 micrograms of cephaxelin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter.

    Note: The 10 milliliters of 0.01N iodine must be added within 20 
seconds after the addition of the 2.0 milliliters of 1.2N hydrochloric 
acid, and the assay should be completed within 1 hour after the sample 
and standard are first put into solution.

    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous suspension containing 50 milligrams per milliliter.
    (5) Absorptivity. Determine the absorbance of the sample and 
standard solutions in the following manner: Dissolve accurately weighed 
portions of approximately 50 milligrams each of the sample and standard 
in 250 milliliters of distilled water. Transfer a 10-milliliter aliquot 
to a 100-milliliter volumetric flask and dilute to volume with distilled 
water. Using a suitable spectrophotometer and distilled water as the 
blank, determine the absorbance of each solution at 262 nanometers. 
Determine the percent absorptivity of the sample relative to the 
absorptivity of the standard using the following calculations:
[GRAPHIC] [TIFF OMITTED] TR01JA93.174

where:
 m=percent moisture in the sample.

    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 0.5

[[Page 625]]

 percent potassium bromide disc prepared as described in paragraph 
(b)(1) of that section.
    (7) Crystallinity. Proceed as directed in Sec. 436.203 of this 
chapter.
[39 FR 19040, May 30, 1974, as amended at 50 FR 19919, May 13, 1985; 52 
FR 35912, Sept. 24, 1987]



Sec. 442.28  Cephalexin hydrochloride monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephalexin hydrochloride monohydrate is 
the hydrochloride salt of 7-(D-alpha- amino-alpha- phenylacetamido)-3-
methyl-3-cephem-4-carboxylic acid monohydrate. It is so purified and 
dried that:
    (i) Its potency is not less than 800 micrograms and not more than 
880 micrograms of cephalexin per milligram on an ``as is'' basis.
    (ii) Its moisture content is not less than 3.0 nor more than 6.5 
percent.
    (iii) The pH of an aqueous solution containing 10 milligrams per 
milliliter is not less than 1.5 nor more than 3.0.
    (iv) It gives a positive identity test.
    (v) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cephalexin potency, 
moisture, pH, identity, and crystallinity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Cephalexin potency. Proceed as 
directed in Sec. 442.40(b)(1)(ii), except that ``cephalexin'' is 
substituted at each occurrence of ``cephradine''.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (4) Identity. Proceed as directed in Sec. 436.367 of this chapter.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[54 FR 48860, Nov. 28, 1989]



Sec. 442.29a  Sterile cephapirin sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile cephapirin sodium is the sodium 
salt of 7-[(4-pyridylthio) - acetamido]-cephalosporanic acid. 
It is a white to off-white powder. It is so purified and dried that:
    (i) Its potency is not less than 855 micrograms and not more than 
1,000 micrograms of cephapirin per milligram on an ``as is'' basis. If 
it is packaged for dispensing, its content is satisfactory if it 
contains not less than 90 percent and not more than 115 percent of the 
number of milligrams of cephapirin that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 2.0 percent.
    (vi) Its pH in an aqueous solution containing 10 milligrams of 
cephapirin per milliliter is not less than 6.5 and not more than 8.5.
    (vii) Its cephapirin content is not less than 92 percent and not 
more than 105 percent on an anhydrous basis.
    (viii) It gives a positive identity test for sodium cephapirin.
    (ix) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, cephapirin content, identity, and crystallinity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 9 packages, each containing 
approximately 500 milligrams, and 1 package containing approximately 5 
grams.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.

[[Page 626]]

    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 14 immediate 
containers, except if each contains less than 1 gram, a minimum of 19 
immediate containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to give a stock solution of 
convenient concentration; also, if it is packaged for dispensing, 
reconstitute as directed in the labeling. Then using a suitable 
hypodermic needle and syringe, remove all of the withdrawable contents 
if it is represented as a single-dose container; or, if the labeling 
specifies the amount of potency in a given volume of the resultant 
preparation, remove an accurately measured representative portion from 
each container. Dilute with solution 1 to give a stock solution of 
convenient concentration. Further dilute an aliquot of the stock 
solution with solution 1 to the reference concentration of 1.0 microgram 
of cephapirin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter. In addition if it is packaged for dispensing, reconstitute as 
directed in the labeling. Then using a suitable hypodermic needle and 
syringe, remove all of the withdrawable contents if it is represented as 
a single-dose container; or, if the labeling specifies the amount of 
potency in a given volume of the resultant preparation, remove an 
accurately measured representative portion from each container. Dilute 
with distilled water to the prescribed concentration.
    (iii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter. In addition, if it is packaged for 
dispensing, reconstitute as directed in the labeling. Then using a 
suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is represented as a single-dose container; or, if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute with distilled water to the 
prescribed concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 100 milligrams of cephapirin per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (7) Cephapirin content. Proceed as directed in Sec. 436.213 of this 
chapter, using the titration procedure described in paragraph (e)(2) of 
that section. Calculate the cephapirin content as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.175

where:
A=Milliliters of perchloric acid reagent used in titrating the sample.
B=Milliliters of perchloric acid reagent used in titrating the blank.
m=Percent moisture content of the sample.

    (8) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 1.0 percent potassium bromide disc prepared

[[Page 627]]

as directed in paragraph (b)(1) of that section.
    (9) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19040, May 30, 1974, as amended at 40 FR 23725, June 2, 1975; 50 
FR 19919, May 13, 1985]



Sec. 442.40  Cephradine.

    (a) Requirements of certification--(1) Standards of identity, 
strength, quality, and purity. Cephradine is (6R, 7R)-7-[(R)-2-amino-2-
(1,4-cyclohexadien-1-yl) acetamido]-3-methyl-8-oxo-5-thia-1-azabicyclo 
[4.2.0]oct-2-ene-2-carboxylic acid. It is so purified and dried that:
    (i) Its potency is not less than 900 micrograms and not more than 
1,050 micrograms of cephradine per milligram on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 6.0 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 3.5 and not more than 6.0.
    (v) Its cephalexin content is not more than 5 percent on an 
anhydrous basis.
    (vi) It passes the identity test.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, cephalexin content, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to give a stock solution 
containing 1.0 milligram of cephradine per milliliter (estimated). 
Further dilute an aliquot of the stock solution with solution 1 to the 
reference concentration of 10 micrograms of cephradine per milliliter 
(estimated).
    (ii) Hydroxylamine colorimetric assay for cephradine--(a) Typical 
equipment. Use automated equipment capable of performing the following 
functions: Introduction of sample into reaction vessels, addition of 
reagents to the samples to form reaction mixtures, incubation of the 
reaction mixtures, colorimetric determination of the reaction product at 
480 nanometers using a 1-centimeter tubular flow cuvette, and 
documentation of the results with a strip chart recorder. A suitable 
system is the Auto Analyzer II equipment consisting of a Solid or Liquid 
Sampler II, a twenty channel Pump III, a colorimeter equipped with a 1-
centimeter tubular flow cuvette and light filters producing incident 
light at 480 nanometers, and a strip chart recorder with scale expander.
    (b) Reagents--(1) Hydroxylamine hydrochloride solution. Dissolve 20 
grams of hydroxylamine hydrochloride and 5 milliliters of emulsifying 
stock solution (prepared to contain 100 milligrams of polyoxyethylene 
fatty alcohol ether, such as Brij-35 or equivalent, per 100 milliliters 
distilled water) in sufficient distilled water to make 1 liter.
    (2) Buffer. Dissolve 173 grams of sodium hydroxide and 20.6 grams of 
sodium acetate in sufficient distilled water to make 1 liter. Dilute 75 
milliliters of this solution with distilled water to 500 milliliters.
    (3) 3.3N Sulfuric acid. Dilute 91 milliliters of concentrated 
sulfuric acid to 1 liter with distilled water.
    (4) Ferric nitrate solution. Dissolve 300 grams of ferric nitrate 
nonahydrate (9H2O) in a mixture of 2.8 milliliters of 
concentrated sulfuric acid and sufficient distilled water to make 1 
liter.
    (c) Preparation of working standard solutions. Dissolve and dilute 
an accurately weighed portion of the cephradine working standard in 
sufficient distilled water to obtain a concentration of 1 milligram of 
cephradine per milliliter.
    (d) Preparation of sample solutions. Dissolve an accurately weighed 
portion of the sample in distilled water and

[[Page 628]]

further dilute to 1 milligram of cephradine per milliliter (estimated).
    (e) Procedure. Use the standard and sample solutions prepared as 
indicated in paragraph (b)(1)(ii) (c) and (d) of this section 
respectively. The arrangement of the apparatus and flow of samples and 
reagents are shown in the manifold diagram set forth in this paragraph 
(b)(1)(ii)(e). The sampler rate is usually 40 per hour, but may be 
varied.

[[Page 629]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.382

    (f) Calculate the potency of the sample in micrograms per milligram 
as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.176


[[Page 630]]


where:
Au=Absorbance of sample solution;
Ps=Potency of working standard solution in micrograms, per 
milliliter;
As=Absorbance of working standard solution;
Wu=Milligrams of sample per milliliter of sample solution;
m=Percent moisture in sample.

    (iii) High-pressure liquid chromatographic assay. Proceed as 
directed in Sec. 436.337 of this chapter, preparing the sample as 
described in paragraph (e)(3)(i) of that section.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Cephalexin content. Proceed as directed in Sec. 436.337 of this 
chapter.
    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 1 percent potassium bromide disc prepared as described in 
paragraph (b)(1) of that section.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[40 FR 26270, June 23, 1975, as amended at 45 FR 16474, Mar. 14, 1980; 
46 FR 25608, May 8, 1981; 48 FR 51293, Nov. 8, 1983; 49 FR 47485, Dec. 
5, 1984; 50 FR 19919, May 13, 1985]



Sec. 442.40a  Sterile cephradine.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephradine is 7-[D-2 - amino -2- (1,4 - 
cyclohexadien - 1 -yl) acetamido] - 3 - methyl - 8 - oxo - 5-thia - 1- 
azabicyclo[4.2.0]oct -2 - ene-2-carboxylic acid. It is so purified and 
dried that:
    (i) Its potency is not less than 900 and not more than 1,050 
micrograms of cephradine per milligram on the anhydrous basis. If it is 
packaged for dispensing, its cephradine content is satisfactory if it is 
not less than 90 percent and not more than 115 percent of the number of 
milligrams of cephradine that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 6.0 percent.
    (vi) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 3.5 and not more than 6.0.
    (vii) Its cephalexin content is not more than 5 percent on an 
anhydrous basis.
    (viii) It passes the identity test.
    (ix) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, cephalexin content, identity, and crystallinity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for manufacturing use:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (2) For sterility testing: 1 package containing approximately 6 
grams of a composite sample.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to give a stock solution 
containing 1.0 milligram of cephradine per milliliter (estimated); also, 
if it is packaged for dispensing, reconstitute the sample as directed in 
the labeling, except use distilled water in lieu of reconstituting 
fluid. Then using a suitable hypodermic needle and syringe, remove an 
accurately measured representative portion from each container. Dilute 
with solution 1 to give a

[[Page 631]]

stock solution of convenient concentration. Further dilute an aliquot of 
the stock solution with solution 1 to the reference concentration of 10 
micrograms of cephradine per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii). If packaged for dispensing, reconstitute the 
sample as directed in the labeling using distilled water instead of the 
reconstituting fluid. Further dilute an aliquot of this solution with 
distilled water to 1 milligram of cephradine per milliliter (estimated).
    (iii) High-pressure liquid chromatographic assay. Proceed as 
directed in Sec. 436.337 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(g) of this chapter, 
using a solution containing 80 milligrams of cephradine per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (7) Cephalexin content. Proceed as directed in Sec. 442.40(b)(5).
    (8) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 1 percent potassium bromide disc prepared as described in 
paragraph (b)(1) of that section.
    (9) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[40 FR 51626, Nov. 6, 1975, as amended at 43 FR 14646, Apr. 7, 1978; 49 
FR 47485, Dec. 5, 1984; 50 FR 19919, May 13, 1985]



Sec. 442.41  Cephradine dihydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephradine dihydrate is the dihydrate 
form of (6R,7R)-7-[(R)-2-amino-2-(1,4-cyclohexadien-1-yl)acetamido]-3-
methyl-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid. It 
is so purified and dried that:
    (i) Its potency is not less than 900 micrograms and not more than 
1,050 micrograms of cephradine per milligram on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 8.5 percent and not more 
than 10.5 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 3.5 and not more than 6.0.
    (v) Its cephalexin content is not more than 5 percent on an 
anhydrous basis.
    (vi) It passes the identity test.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, cephalexin content, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use any of the 
following methods; however, the results obtained from the hydroxylamine 
colorimetric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to obtain a stock solution of 
convenient concentration. Further dilute an aliquot of the stock 
solution with solution 1 to the reference concentration of 10.0 
micrograms of cephradine per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay for cephradine. Proceed as 
directed in Sec. 442.40(b)(1)(ii).
    (iii) High-pressure liquid chromatographic assay. Proceed as 
directed in Sec. 436.337 of this chapter, preparing the sample as 
described in paragraph (e)(3)(i) of that section.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of thichapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.

[[Page 632]]

    (5) Cephalexin content. Proceed as directed in Sec. 436.337 of this 
chapter.
    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 1 percent potassium bromide disc prepared as described in 
paragraph (b)(1) of that section.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[47 FR 11856, Mar. 19, 1982, as amended at 49 FR 47485, Dec. 5, 1984; 50 
FR 19919, May 13, 1985]



Sec. 442.50a  Sterile ceforanide.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ceforanide is 5-thia-1-
azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid, 7-[[[2-(amino-
methyl)phenyl]acetyl]amino]-3-[[[1-(carboxymethyl)-1H-tetrazol-5-yl]-
thio]methyl]-8-oxo-,(6R-trans)-. It is a white to off-white powder. It 
is so purified and dried that:
    (i) Its ceforanide content is not less than 900 micrograms and not 
more than 1,050 micrograms of ceforanide per milligram.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 5.0 percent.
    (v) Its pH in an aqueous suspension containing 50 milligrams per 
milliliter is not less than 2.5 and not more than 4.5.
    (vi) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for ceforanide content, 
sterility, pyrogens, moisture, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (b) For sterility testing: One package containing approximately 6 
grams of a composite sample.
    (b) Tests and methods of assay--(1) Ceforanide content. Proceed as 
directed in Sec. 436.348 of this chapter, preparing the sample and 
calculating the ceforanide content as follows:
    (i) Preparation of sample solution. Prepare a solution containing 
1.0 milligram per milliliter in mobile phase. Inject each sample within 
5 minutes after dissolution.
    (ii) Calculations. Calculate the micrograms of ceforanide per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.177

where:
Au = Area of the ceforanide peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As = Area of the ceforanide peak in the chromatogram of the 
          ceforanide working standard;
Ps = Ceforanide activity in the ceforanide working standard 
          solution in micrograms per milliliter; and
Cu = Milligrams of sample per milliliter of sample solution.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except:
    (i) In paragraph (e)(1)(i)(a) of that section, use diluting fluid G 
in lieu of diluting fluid A; and
    (ii) In lieu of three 100-milliliter quantities of diluting fluid A 
in paragraph (e)(2) of that section, filter three 100-milliliter 
quantities of diluting fluid D followed by a 100-milliliter quantity of 
diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
except suspend 1 gram of sterile ceforanide in 12.5 milliliters of 
pyrogen-free water (diluent 1). Add 320 milligrams of pyrogen-free L-
lysine base, shake to dissolve the mixture. If the mixture is not 
dissolved, add an amount of L-lysine necessary to obtain a solution. The 
test sample should contain not more than a total of 340 milligrams of L-
lysine. Dilute the resulting solution to 20 milliliters. Use a test dose 
of 1 milliliter of the 50 milligrams per milliliter test solution per 
kilogram of rabbit weight.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.

[[Page 633]]

    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous suspension containing 50 milligrams per milliliter.
    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation as described in paragraph (b)(2) of that 
section.
[49 FR 25847, June 25, 1984; 49 FR 40006, Oct. 12, 1984, as amended at 
55 FR 11583, Mar. 29, 1990]



Sec. 442.52  Cefotetan.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefotetan is (6R,7S)-4-[[2-carboxy-7-
methoxy-3-[[(1-methyl-1H-tetrazol-5-yl)thio]methyl]-8-oxo-5-thia-1-
azabicyclo[4.2.0]oct-2-ene-7-yl]-carbamoyl]-1,3-dithietane-
2,-malonamic acid. It is so purified and 
dried that:
    (i) Its potency is not less than 950 micrograms and not more than 
1,030 micrograms of cefotetan activity per milligram on the anhydrous 
basis.
    (ii) Its moisture content is not more than 2.5 percent.
    (iii) It gives a positive identity test for cefotetan.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, except use the resolution test solution to 
determine resolution in lieu of the working standard solution. Perform 
the assay at ambient temperature, using an ultraviolet detection system 
operating at a wavelength of 254 nanometers, a column packed with 
microparticulate (3 to 10 micrometers in diameter) reversed phase 
packing material such as octadecyl hydrocarbon bonded silicas, a flow 
rate not exceeding 2.0 milliliters per minute, and a known injection 
volume of between 10 and 20 microliters. Reagents, working standard 
solution, sample solution, resolution test solution, system suitability 
requirements, and calculations are as follows:
    (i) Reagents--(A) Diluting solution. Mix water:methanol:acetonitrile 
(90:5:5).
    (B) Mobile phase. Mix 0.1M phosphoric acid:glacial acetic 
acid:methanol:acetonitrile (1700:100:105:105). Filter through a suitable 
filter capable of removing particulate matter greater than 0.5 micron in 
diameter. Degas the mobile phase just prior to its introduction into the 
chromatograph.
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(A) Working standard solution. Accurately weigh approximately 
50 milligrams of the cefotetan working standard into a 250-milliliter 
volumetric flask containing 12.5 milliliters of methanol. Swirl the 
flask for several minutes, then add 12.5 milliliters of acetonitrile. 
Swirl the flask until the cefotetan is dissolved. Dilute to volume with 
water to obtain a solution containing approximately 200 micrograms of 
cefotetan per milliliter. Mix well. Protect the working standard 
solution from light.
    (B) Sample solution. Dissolve an accurately weighed portion of the 
sample with sufficient diluting solution described in paragraph 
(b)(1)(i)(A) of this section to obtain a concentration of approximately 
200 micrograms of cefotetan per milliliter.
    (C) Resolution test solution. Place 10 milliliters of the working 
standard solution in a stoppered flask containing a few milligrams of 
magnesium carbonate. Close the flask and sonicate for 10 minutes. If the 
solution is not slightly turbid, add more magnesium carbonate and repeat 
sonication. Filter the turbid solution through a 0.5-micron filter and 
use within 2 hours. As this solution stands, the tautomer concentration 
increases.
    (iii) System suitability requirements--(A) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.3 at 10 
percent of peak height in lieu of 5 percent of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,500 theoretical plates.

[[Page 634]]

    (C) Resolution. The resolution (R) between the peak for cefotetan 
and its tautomer is satisfactory if it is not less than 2.0.
    (D) Coefficient of variation. The coefficient of variation 
(Srin percent) of five replicate injections is satisfactory 
if it is not more than 2.0 percent. If the system suitability 
requirements have been met, then proceed as described in Sec. 436.216 
(b) of this chapter. Alternate chromatographic conditions are acceptable 
provided comparable system suitability requirements are met. However, 
the sample preparation described in paragraph (b)(1)(ii)(B) of this 
section should not be changed.
    (iv) Calculation. Calculate the micrograms of cefotetan per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.178

where:
AU = Area of the cefotetan peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
AS = Area of the cefotetan peak in the chromatogram of the 
          cefotetan working standard;
PS = Cefotetan activity in the cefotetan working standard 
          solution in micrograms per milliliter;
Vf = Volume of flask used to dilute standard; and
Vs = Volume of sample diluted.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Identity. Proceed as directed in Sec. 436.211 of this chapter 
using the potassium bromide discs prepared as described in 
Sec. 436.211(b)(1) of this chapter or the mineral oil mull prepared as 
described in Sec. 436.211(b)(2) of this chapter.
[59 FR 26940, May 25, 1994, as amended at 60 FR 33712, June 29, 1995]



Sec. 442.53a  Sterile cefotetan disodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile cefotetan disodium is a white to 
off-white lyophilized powder. It is so purified and dried that:
    (i) If the cefotetan disodium is not packaged for dispensing, its 
potency is not less than 830 micrograms and not more than 970 micrograms 
of cefotetan per milligram on the anhydrous basis. If the cefotetan 
disodium is packaged for dispensing, its potency is not less than 830 
micrograms and not more than 970 micrograms of cefotetan per milligram 
on the anhydrous basis and also, each container contains not less than 
90 percent and not more than 120 percent of the number of milligrams of 
cefotetan that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 1.5 percent.
    (v) Its pH in an aqueous solution containing 100 milligrams of 
cefotetan disodium per milliliter is not less than 4.0 and not more than 
6.5.
    (vi) It gives a positive identity test for cefotetan.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, except use the resolution test solution to 
determine resolution in lieu of the working standard solution. Perform 
the assay at ambient temperature, using an ultraviolet detection system 
operating at a wavelength of 254 nanometers, a column

[[Page 635]]

packed with microparticulate (3 to 10 micrometers in diameter) reversed 
phase packing material such as octadecyl hydrocarbon bonded silicas, a 
flow rate not exceeding 2.0 milliliters per minute, and a known 
injection volume of between 10 and 20 microliters. Reagents, working 
standard solution, sample solution, resolution test solution, system 
suitability requirements, and calculations are as follows:
    (i) Reagents--(a) Diluting solution. Mix water:methanol:acetonitrile 
(90:5:5).
    (b) Mobile phase. Mix 0.1M phosphoric acid:glacial acetic 
acid:methanol:acetonitrile (1700:100:105:105). Filter through a suitable 
filter capable of removing particulate matter greater than 0.5 micron in 
diameter. Degas the mobile phase just prior to its introduction into the 
chromatograph.
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(a) Working standard solution. Accurately weigh approximately 
50 milligrams of the cefotetan working standard into a 250-milliliter 
volumetric flask containing 12.5 milliliters of methanol. Swirl the 
flask for several minutes, then add 12.5 milliliters of acetonitrile. 
Swirl the flask until the cefotetan is dissolved. Dilute to volume with 
water to obtain a solution containing approximately 200 micrograms of 
cefotetan per milliliter. Mix well. Protect the working standard 
solution from light.
    (b) Sample solutions--(1) Product not packaged for dispensing 
(micrograms of cefotetan per milligram). Dissolve an accurately weighed 
portion of the sample with sufficient diluting solution described in 
paragraph (b)(1)(i)(a) of this section, to obtain a concentration of 
approximately 200 micrograms of cefotetan per milliliter.
    (2) Product packaged for dispensing. Determine both micrograms of 
cefotetan per milligram of the sample and milligrams of cefotetan per 
container. Use separate containers for preparation of each sample 
solution as described in paragraphs (b)(1)(ii)(b)(2) (i) and (ii) of 
this section.
    (i) Micrograms of cefotetan per milligram. Dissolve an accurately 
weighed portion of the sample with sufficient diluting solution 
described in paragraph (b)(1)(i)(a) of this section, to obtain a 
concentration of approximately 200 micrograms of cefotetan per 
milliliter.
    (ii) Milligrams of cefotetan per container. Reconstitute the sample 
as directed in the labeling. Then, using a suitable hypodermic needle 
and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency in a given volume of the resultant preparation, 
remove an accurately measured representative portion from each 
container. Further dilute an aliquot of the solution thus obtained with 
sufficient diluting solution described in paragraph (b)(1)(i)(a) of this 
section, to obtain a concentration of approximately 200 micrograms of 
cefotetan per milliliter.
    (c) Resolution test solution. Place 10 milliliters of the working 
standard solution in a stoppered flask containing a few milligrams of 
magnesium carbonate. Close the flask and sonicate for 10 minutes. If the 
solution is not slightly turbid, add more magnesium carbonate and repeat 
sonication. Filter the turbid solution through a 0.5-micron filter and 
use within 2 hours. As this solution stands, the tautomer concentration 
increases.
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.3 at 10 
percent of peak height in lieu of 5 percent of peak height.
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,500 theoretical plates.
    (c) Resolution. The resolution (R) between the peak for cefotetan 
and its tautomer is satisfactory if it is not less than 2.0.
    (d) Coefficient of variation. The coefficient of variation (SR 
in percent) of five replicate injections is satisfactory if it is not 
more than 2.0 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.216(b) of this chapter. Alternate chromatographic 
conditions are acceptable provided comparable system suitability 
requirements are met. However, the sample preparation described in 
paragraph (b)(1)(ii)(b) of this section should not be changed.

[[Page 636]]

    (iv) Calculations--(a) Calculate the micrograms of cefotetan per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.179

where:

    Au=Area of the cefotetan peak in the chromatogram of the 
sample (at a retention time equal to that observed for the standard);
    As=Area of the cefotetan peak in the chromatogram of the 
cefotetan working standard;
    Ps=Cefotetan activity in the cefotetan working standard 
solution in micrograms per milliliter;
    Cu=Milligrams of sample per milliliter of sample 
solution; and
    m=Percent moisture content of the sample.

    (b) Calculate the cefotetan content of the container as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.180
    
where:

    Au=Area of the cefotetan peak in the chromatogram of the 
sample (at a retention time equal to that observed for the standard);
    As=Area of the cefotetan peak in the chromatogram of the 
cefotetan working standard;
    Ps=Cefotetan activity in the cefotetan working standard 
solution in micrograms per milliliter; and
    d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cefotetan per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams of cefotetan disodium per 
milliliter.
    (6) Identity. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section, compares 
qualitatively to that of the cefotetan working standard.
[51 FR 20263, June 4, 1986, as amended at 52 FR 35912, Sept. 24, 1987; 
55 FR 11583, Mar. 29, 1990]



Sec. 442.54  Cefpodoxime proxetil.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefpodoxime proxetil is ()-1-
hydroxyethyl(+)-(6R,7R)-7-[2-(2-amino-4-thiazolyl)glyoxylamido]-3-
(methoxymethyl)-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-
carboxylate,72-(Z)-(O-methyloxime), isopropyl carbonate 
(ester). It is so purified and dried that:
    (i) Its potency is not less than 690 micrograms and not more than 
804 micrograms of cefpodoxime activity per milligram, on an anhydrous 
basis.
    (ii) The ratio of its R-epimer to total cefpodoxime is not less than 
0.5 and not more than 0.6.
    (iii) Its moisture content is not more than 3 percent.
    (iv) It gives a positive identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cefpodoxime 
potency, isomer ratio, moisture, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using a suitable thermostatted column 
heating mechanism to maintain a column temperature of 40  deg.C, an 
ultraviolet detection system operating at a wavelength of 254 
nanometers, a 15 centimeter X 4.6 millimeter (i.d.) column packed with 
microparticulate (5 micrometers in diameter) reversed phase packing 
material such as octadecyl silane bonded to silicas, a flow rate of 0.8 
milliliter per minute, and a known injection volume of 2 microliters. 
The retention time for

[[Page 637]]

the S-epimer is approximately 22 minutes and the retention time for R-
epimer is approximately 28 minutes. The internal standard 
(propylparaben) has a retention time of 34 minutes. Mobile phase, 
dilution solvent, resolution solution, internal standard solution, 
working standard and sample solutions, system suitability requirements, 
and calculations are as follows:
    (i) Mobile phase. The mobile phase consists of 420 milliliters of 
methanol, 580 milliliters of deionized water, and 230 milligrams of L-
histidine hydrochloride. The pH is adjusted to 2.50.1 using 
2N sulfuric acid. The mobile phase must be at room temperature for a 
correct pH measurement. The methanol concentration may be adjusted to 
achieve comparable retention times from column to column. Increasing 
methanol reduces retention times. Filter the mobile phase through a 
suitable filter capable of removing particulate matter 0.5 micron in 
diameter and degas it just before its introduction into the 
chromatograph.
    (ii) Dilution solvent. Prepare a solvent for dilution by thoroughly 
mixing 495 milliliters of deionized water, 495 milliliters of 
acetonitrile, and 10 milliliters of acetic acid in an appropriate 
container.
    (iii) Resolution solution. Prepare a 1 milligram per milliliter 
solution of any bulk containing ANTI-A in dilution solvent. Use this 
solution to determine the resolution between ANTI-A and the later-
eluting drug epimer (R-epimer). Alternately, the resolution factor can 
be determined between the R and S isomers.
    (iv) Internal standard solution. Prepare a solution of propylparaben 
in dilution solvent at a concentration of 10 milligrams per milliliter.
    (v) Preparation of working standard solutions. Accurately weigh 
approximately 42 milligrams of the cefpodoxime proxetil working 
reference standard add 3 milliliters of internal standard solution and 
25 milliliters of dilution solvent. The standard solution is stable for 
at least 48 hours. Refrigeration is not recommended.
    (vi) Sample solution. Accurately weigh approximately 42 milligrams 
of the sample, add 3 milliliters of internal standard and 25 milliliters 
of dilution solvent. The sample solution is stable for at least 48 
hours. Refrigeration is not recommended.
    (vii) System suitability requirements--(A) Asymmetry factor.The 
asymmetry factor (As) is satisfactory if it is not less than 
0.8 and not more than 1.1 for the R-epimer of cefpodoxime peak.
    (B) Efficiency of the column. The absolute efficiency 
(hr) is satisfactory if it is not more than 5 for the R-
epimer peak.
    (C) Resolution factor. The resolution factor (R) between the peak 
for ANTI-A and the peak for the R-epimer is satisfactory if it is not 
less than 1.3. Alternately, the resolution factor (R) between the peak 
for the R-epimer and the peak for the S-epimer of cefpodoxime is not 
less than 11.
    (D) Coefficient of variation (Relative standard deviation). The 
coefficient of variation (SRin percent of 5 replicate 
injections) is satisfactory if it is not more than 2 percent.
    (E) Capacity factor (k'). The capacity factor (k') for the R-epimer 
of cefpodoxime is satisfactory if it is not less than 10.4 and not more 
than 15.6.
    (F) If the system suitability parameters in this paragraph 
(b)(1)(iv) have been met, then proceed as described in Sec. 436.216(b) 
of this chapter.
    (viii) Calculations. Calculate the micrograms of cefpodoxime 
proxetil per milligram of sample on an anhydrous basis as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.383

where:
Ru = Ratio of cefpodoxime proxetil peaks area (sum of both 
          epimers) to the internal standard peak response in the sample 
          solution;
Rs = Ratio of cefpodoxime proxetil peaks

[[Page 638]]

          area (sum of both epimers) to the internal standard peak 
          response in the working standard solution;
Ps = Cefpodoxime proxetil activity of the working standard 
          solution in micrograms per milliliter;
Cu = Milligrams of sample per milliliter of sample solution; 
          and
m = Percent moisture content of the sample.

    (2) Isomer ratio. Using the procedure described in paragraph (b)(1) 
of this section, calculate the ratio of the R-epimer (Ab) to the sum of 
the S-epimer and R-epimer (Aa and Ab), by the equation

    Isomer Ratio = Ab/(Aa + Ab)

where:
    Aa = Area of the early eluting S-epimer peak; and
    Ab = Area of the late eluting R-epimer peak.

    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
except use 30 milliliters of solvent C instead of 20 milliliters of 
solvent A.
    (4) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the mineral oil mull prepared as described in paragraph (b)(2) of 
that section.
[60 FR 58231, Nov. 27, 1995]



Sec. 442.55  Ceftriaxone sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ceftriaxone sodium is the 5-thia-1-
azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid, 7-[[(2-amino-4-thiazolyl) 
(methoxyimino)acetyl]amino]-8-oxo-3-[[(1,2,5,6-tetrahydro-2-methyl-5,6-
dioxo-1,2,4-triazin-3-yl)thio]methyl]-,disodium salt, [6R-[6alpha, 
7beta(Z)]]-. It is so purified and dried that:
    (i) Its ceftriaxone potency is not less than 795 micrograms of 
ceftriaxone per milligram on an anhydrous free acid basis.
    (ii) Its moisture content is not less than 8 percent and not more 
than 11 percent.
    (iii) The pH of an aqueous solution containing the equivalent of 
100.0 milligrams per milliliter is not less than 6.0 and not more than 
8.0.
    (iv) It is crystalline.
    (v) It gives a positive identity test for ceftriaxone.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for ceftriaxone 
potency, moisture, pH, crystallinity, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Ceftriaxone potency. Proceed as 
directed in Sec. 442.55a(b)(1) of this chapter, except prepare the 
sample solution and calculate the micrograms of ceftriaxone free acid 
per milligram as follows:
    (i) Preparation of sample solution. Dissolve an accurately weighed 
portion of the sample with sufficient water to obtain a concentration of 
180 micrograms of ceftriaxone activity per milliliter. Prepare the 
sample solution just prior to its introduction into the chromatograph.
    (ii) Calculation. Calculate the micrograms of ceftriaxone anhydrous 
free acid per milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.181

where:
Au=Area of the ceftriaxone peak in the chromatogam of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the ceftriaxone peak in the chromatogram of the 
          ceftriaxone working standard;
Ps=Ceftriaxone activity in the ceftriaxone working standard 
          solution in micrograms of anhydrous free acid per milliliter; 
          and
Cu=Milligrams of sample per milliliter of sample solution.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (4) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.

[[Page 639]]

    (5) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a potassium bromide disc containing 1.3 milligrams of ceftriaxone 
sodium in 300 milligrams of potassium bromide, prepared as described in 
paragraph (b)(1) of that section.
[52 FR 44860, Nov. 23, 1987, as amended at 55 FR 11583, Mar. 29, 1990]



Sec. 442.55a  Sterile ceftriaxone sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ceftriaxone sodium is 5-thia-1-
azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid, 7-[[(2-amino-4-thiazolyl) 
(methoxyimino)acetyl]amino]-8-oxo-3-[[(1,2,5,6-tetrahydro-2-methyl-5,6-
dioxo-1,2,4-triazin-3-yl)thio]methyl]-, disodium salt, [6R-
[6,7(Z)]]-. It is so purified and dried that:
    (i) If the ceftriaxone sodium is not packaged for dispensing, its 
ceftriaxone potency is not less than 795 micrograms of ceftriaxone per 
milligram on an anhydrous free acid basis. If the ceftriaxone sodium is 
packaged for dispensing, its ceftriaxone potency is not less than 776 
micrograms of ceftriaxone per milligram on an anhydrous free acid basis 
and also, each container contains not less than 90 percent and not more 
than 115 percent of the number of milligrams of ceftriaxone that it is 
represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not less than 8 percent and not more 
than 11 percent.
    (v) Its pH in an aqueous solution containing the equivalent of 100.0 
milligrams per milliliter is not less than 6.0 and not more than 8.0.
    (vi) It is crystalline.
    (vii) It gives a positive identity test for ceftriaxone.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for ceftriaxone 
potency, and if packaged for dispensing, potency and container content, 
sterility, pyrogens, moisture, pH, crystallinity, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) If the batch is packaged for repacking or for manufacturing use:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (2) For sterility testing: 20 packages, each containing equal 
portions of approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Ceftriaxone potency and 
container content. Proceed as directed in Sec. 436.354 of this chapter, 
using ambient temperature, an ultraviolet detection system operating at 
a wavelength of 270 nonometers (or 254 nanometers fixed mercury source), 
and a column packed with a five-micron octadecyl reverse phase packing 
or equivalent; and also, using the following system suitability 
requirements, reagents, working standard, test and sample solutions, and 
calculations:
    (i) System suitability requirements--(a) Capacity factor. The 
capacity factor (k) for the ceftriaxone peak is satisfactory if it is 
not less than 2 and not more than 5.
    (b) Resolution. The resolution (R) between the peak for ceftriaxone 
E-isomer and ceftriaxone is satisfactory if it is not less than 3.0.
    (c) Asymmetry factor. The asymmetry factor (As) is 
satisfactory if it is not more than 1.6 at 10 percent of the peak 
height.
    (d) Efficiency of the column. The efficiency of the column 
(hr) is satisfactory if it is less than 20 (equivalent to a 
value of 1,500 or greater theoretical plates when using a 15-centimeter 
column with 5-micrometer-size particles).
    (e) Coefficient of variation. The coefficient of variation (SR 
in percent) of five replicate injections is satisfactory if it is less 
than 2.0 percent.

[[Page 640]]


If the system suitability parameters have been met, then proceed as 
described in Sec. 436.354(b) of this chapter.
    (ii) Reagents--(a) pH 7.0 phosphate buffer. Dissolve 13.6 grams of 
dibasic potassium phosphate and 4.0 grams of monobasic potassium 
phosphate in sufficient water to make 1,000 milliliters. Adjust to pH 
7.0 0.1 with 18N phosphoric acid or 10N potassium hydroxide.
    (b) pH 5.0 citrate buffer. Dissolve 25.8 grams of sodium citrate in 
500 milliliters of water. Adjust the pH to 5.0plus-minus0.1 
with 20 percent aqueous citric acid, and dilute to 1,000 milliliters 
with water.
    (c) Mobile phase. Dissolve 4.0 grams of tetraheptylammonium bromide 
with 500 milliliters of acetonitrile. Add 440 milliliters of water, 55 
milliliters of pH 7.0 phosphate buffer, and 5 milliliters of pH 5.0 
citrate buffer. Mix and dilute 800 milliliters of this solution with 200 
milliliters of distilled water. Filter the mobile phase through a 
suitable glass fiber filter or equivalent which is capable of removing 
particulate contamination greater than 0.5 micron in diameter. Degas the 
mobile phase just prior to its introduction into the chromatograph.
    (iii) Working standard and sample solutions--(a) Preparation of 
working standard solution. Dissolve an accurately weighed portion of the 
ceftriaxone working standard with sufficient water to obtain a solution 
containing approximately 180 micrograms of ceftriaxone activity per 
milliliter. Prepare the working standard solution just prior to its 
introduction into the chromatograph.
    (b) Preparation of test solution. Dissolve together accurately 
weighed portions of the ceftriaxone working standard and the ceftriaxone 
sodium E-isomer reference standard with sufficient water to obtain a 
solution containing approximately 160 micrograms of ceftriaxone activity 
per milliliter of each standard. Prepare the test solution just prior to 
its introduction into the chromatograph.
    (c) Preparation of sample solution. Prepare the sample solution just 
prior to its introduction into the chromatograph.
    (1) Product not packaged for dispensing (micrograms of ceftriaxone 
anhydrous free acid per milligram). Dissolve an accurately weighed 
portion of the sample with sufficient water to obtain a concentration of 
180 micrograms of ceftriaxone activity per milliliter.
    (2) Product packaged for dispensing. Determine both potency 
(micrograms of ceftriaxone anhydrous free acid per milligram of the 
sample) and container content (milligrams of anhydrous free acid 
ceftriaxone per container). Use separate containers for preparation of 
each sample solution as described in paragraph (b)(1)(iii)(b)(2) (i) and 
(ii) of this section.
    (i) Micrograms of ceftriaxone anhydrous free acid per milligram. 
Dissolve an accurately weighed portion of the sample with sufficient 
water to obtain a concentration of approximately 180 micrograms of 
ceftriaxone activity per milliliter.
    (ii) Milligrams of ceftriaxone per container. Reconstitute the 
sample as directed in the labeling. Then, using a suitable hypodermic 
needle and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the potency contained in a given volume of the resulting preparation, 
remove an accurately measured representative portion from each 
container. Dilute the aliquot of the solution thus obtained with 
sufficient water to obtain a concentration of approximately 180 
micrograms of ceftriaxone activity per milliliter.
    (iv) Calculations. (a) Calculate the micrograms of ceftriaxone 
anhydrous free acid per milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.182

where:
Au=Area of the ceftriaxone peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the ceftriaxone peak in the chromatogram of the 
          ceftriaxone working standard;
Ps=Ceftriaxone activity in the ceftriaxone working standard 
          solution in micrograms of anhydrous free acid per milliliter; 
          and
Cu=Milligrams of sample per milliliter of sample solution.


[[Page 641]]


    (b) Calculate the ceftriaxone anhydrous free acid content of the 
container as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.183

where:
Au=Area of the ceftriaxone peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the ceftriaxone peak in the chromatogram of the 
          ceftriaxone working standard;
Ps=Ceftriaxone activity in the ceftriaxone working standard 
          solution in micrograms of anhydrous free acid per milliliter; 
          and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(h) of this chapter, 
using a solution containing 20 milligrams of ceftriaxone per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a potassium bromide disc containing 1.3 milligrams of ceftriaxone 
sodium in 300 milligrams of potassium bromide, prepared as described in 
paragraph (b)(1) of that section.
[50 FR 9999, Mar. 13, 1985; 50 FR 11690, Mar. 25, 1985; 50 FR 18243, 
Apr. 30, 1985, as amended at 55 FR 11583, Mar. 29, 1990]



Sec. 442.58a  Sterile cefotiam dihydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefotiam dihydrochloride is 5-thia-1-
azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid, 7-[[(2-amino-4-
thiazolyl)acetyl]-amino-3[[[1-[2-(dimethylamino)ethyl]-1H-tetrazol-5-
yl]-thio]methyl]-8-oxo-, dihydrochloride, (6R-trans)-. It is so purified 
and dried that:
    (i) Its potency is not less than 790 and not more than 925 
micrograms of cefotiam per milligram on an anhydrous basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 7.0 percent.
    (v) It passes the identity test.
    (vi) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, identity, and crystallinity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (B) For sterility testing: One package containing approximately 6 
grams of a composite sample.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 254 nanometers, a column 
packed with microparticulate (3 to 10 micrometers in diameter) reversed 
phase packing material such as octadecyl hydrocarbon bonded silicas, a 
flow rate not to exceed 2.0 milliliters per minute, and a known 
injection volume of between 10 and 20 microliters. Mobile phase, working 
standard and sample solutions, resolution test solution, system 
suitability requirements, and calculations are as follows:
    (i) Mobile phase. Dissolve 13.1 grams of ammonium sulfate in 850 
milliliters of water. Adjust the pH to 6.5 with dilute aqueous ammonia. 
Add 150 milliliters of acetonitrile. Filter through a suitable filter 
capable of removing particulate matter to 0.5 micron in diameter. Degas 
the mobile phase just prior to its introduction into the chromatograph.
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(A)

[[Page 642]]

Working standard solution. Dissolve approximately 100 milligrams of the 
cefotiam working standard, accurately weighed, in water and dilute to 
100 milliliters. Further dilute with mobile phase to obtain a solution 
containing 50 micrograms of cefotiam activity per milliliter.
    (B) Sample solution. Dissolve approximately 100 milligrams of the 
sample, accurately weighed, in water and dilute to 100 milliliters. 
Further dilute with mobile phase to obtain a solution containing 50 
micrograms of cefotiam activity per milliliter (estimated).
    (C) Resolution test solution. Dissolve an accurately weighed portion 
of cefotiam working standard in water to obtain a solution containing 
approximately 1.0 milligram of cefotiam activity per milliliter. Heat 
this solution at 95  deg.C for 15 minutes. This procedure allows 
cefotiam lactone to be produced. Dilute 1.0 milliliter of this solution 
to 100 milliliters with mobile phase.
    (iii) System suitability requirements--(A) Tailing factor. The 
tailing factor (T) for the cefotiam peak is satisfactory if it is not 
more than 1.76 at 5 percent of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1985 theoretical plates for the 
cefotiam peak.
    (C) Resolution factor. The resolution factor (R) between the peak 
for cefotiam and the peak for cefotiam lactone (generated in situ) is 
satisfactory if it is not less than 4.0.
    (D) Coefficient of variation. The coefficient of variation (SR 
in percent) of 5 replicate injections is satisfactory if it is not more 
than 1.0 percent. If the system suitability parameters have been met, 
then proceed as described in Sec. 436.216(b) of this chapter.
    (iv) Calculations. Calculate the micrograms of cefotiam per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.184

where:
Au=Area of the cefotiam peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefotiam peak in the chromatogram of the 
          cefotiam working standard;
Ps=Cefotiam activity in the cefotiam working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of the sample per milliliter of sample 
          solution; and
m=Percent moisture content of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(g) of this chapter, 
using a solution containing 40 milligrams per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the sample preparation described in paragraph (d)(4) of that 
section and the titration procedure described in paragraph (e)(3) of 
that section, except:
    (i) In lieu of 3 milliliters of anhydrous methanol solution, inject 
20 milliliters of a formamide:methanol solution (2:1) into the container 
and shake to dissolve the contents (prior to use in preparation of the 
formamide:methanol solution, dry 500 grams of formamide over 20 grams of 
anhydrous sodium sulfate for 24 hours);
    (ii) Rinse the syringe, needle, and immediate container with two 
separate 5-milliliter portions of anhydrous methanol, in lieu of one 3-
milliliter portion of anhydrous methanol; and
    (iii) In paragraph (e)(3) of that section, add a sufficient volume 
of the formamide:methanol solution (2:1) to cover the electrodes in the 
dry titrating vessel, in lieu of 20 milliliters of solvent A before 
starting the titration.
    (5) Identity. Using a solution containing 20 micrograms per 
milliliter of water and a suitable spectrophotometer, record the 
ultraviolet absorption spectrum from 220 to 310 nanometers. The spectrum 
compares qualitatively to that of the cefotiam working standard 
similarly tested.
    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[54 FR 20785, May 15, 1989]



Sec. 442.60  Cefpiramide.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefpiramide is (6R, 7R)-7-

[[Page 643]]

[(R)-2-(4-hydroxy-6-methyl-nicotinamido)-2-(p-hydroxyphenyl)acetamido]-
3-[[(1-methyl-1H-tetrazol-5-yl)thio]methyl]-8-oxo-5-thia-1-
azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid. It is so purified and 
dried that:
    (i) Its potency is not less than 974 micrograms of cefpiramide 
activity per milligram on an anhydrous basis.
    (ii) Its moisture content is not more than 9.0 percent.
    (iii) Its pH in an aqueous suspension containing 5 milligrams per 
milliliter is not less than 3.0 and not more than 5.0.
    (iv) Its total related substances content by high performance liquid 
chromatography is not more than 2.0 percent. No individual impurity is 
more than 0.7 percent.
    (v) The specific rotation in dimethylformamide solution containing 
10 milligrams of cefpiramide per milliliter is -106  6 
deg.C calculated on an anhydrous basis.
    (vi) It passes the identity test.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, total related substances, specific rotation, identity, and 
crystallinity.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research: 10 packages each containing approximately 500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating to a wavelength of 254 nanometers, a 15- to 
30-centimeter X 4-millimeter (inside diameter) column packed with 
microparticulate (5 to 10 micrometers in diameter) reversed phase 
packing material such as octylsilane bonded to silica, a flow rate not 
to exceed 2.0 milliliters per minute, and a known injection volume of 
between 10 and 20 microliters. Reagents, working standard and sample 
solutions, resolution test solution, system suitability requirements, 
and calculations are as follows:
    (i) Reagents--(A) 0.01M phosphate buffer. Dissolve 1.36 grams of 
monobasic potassium phosphate in 900 milliliters of water. Adjust the pH 
to 6.8 with 1N sodium hydroxide and dilute to 1,000 milliliters with 
water.
    (B) Mobile phase. Mix 0.01M phosphate buffer: acetonitrile: 
tetrahydrofuran: methanol (880:40:40:40). Filter through a suitable 
filter capable of removing particulate matter to 0.5 micron in diameter. 
Degas the mobile phase just prior to its introduction into the 
chromatograph.
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(A) Working standard solution. Dissolve and dilute an 
accurately weighed portion of the cefpiramide working standard in 
sufficient mobile phase to obtain a solution containing 0.25 milligram 
of cefpiramide activity per milliliter.
    (B) Sample solution. Dissolve an accurately weighed portion of the 
sample in mobile phase and further dilute to 0.25 milligram of 
cefpiramide per milliliter (estimated).
    (C) Resolution test solution. Dissolve an accurately weighed portion 
of cefpiramide working standard in 0.01N sodium hydroxide to obtain a 
solution containing approximately 1.0 milligram of cefpiramide activity 
per milliliter. Heat this solution at 95  deg.C for 10 minutes. This 
procedure allows cefpiramide lactone to be produced. Dilute 1.0 
milliliter of this solution to 20 milliliters with mobile phase.
    (iii) System suitability requirements)--(A) Asymmetry factor. 
Calculate the asymmetry factor (As), measured at a point 5 
percent of the peak height from the baseline as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.185

where:
    a=Horizontal distance from point of ascent to point of maximum peak 
height; and
    b=Horizontal distance from the point of maximum peak height to point 
of descent.

The asymmetry factor (As) is satisfactory if it is not less 
than 0.95 and not more than 1.4.

    (B) Efficiency of the column. From the number of theoretical plates 
(n) calculated as described in Sec. 436.216(c)(2) of

[[Page 644]]

this chapter calculate the reduced plate height (hr) as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.186

where:
    L=Length of the column in centimeters;
    n=Number of theoretical plates; and
    dp=Average diameter of the particles in the analytical 
column packing in micrometers.

The absolute efficiency (hr) is satisfactory if it is not 
more than 12.5 for the cefpiramide peak.
    (C) Resolution factor. The resolution factor (R) between the peak 
for cefpiramide and the peak for cefpiramide lactone (generated in situ) 
is satisfactory if it is not less than 6.0.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (Sr in percent of 5 replicate 
injections is satisfactory if it is not more than 2.0 percent.
    (E) Capacity factor (k'). Calculate the capacity (k') for 
cefpiramide as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.187

where:
    tr=Retention time of cefpiramide in minutes; and
    to=Column dead time in minutes, which is estimated from 
the following equation:
[GRAPHIC] [TIFF OMITTED] TR01JA93.188

where:
    D=Column diameter in centimeters;
    L=Column length in centimeters;
    0.75=Average total column porosity; and
    F=Flow rate in milliliters per minute.


The capacity factor (k') for cefpiramide is satisfactory if it is not 
less than 2.0 and not more than 3.0. If the system suitability 
parameters have been met, then proceed as described in Sec. 436.216(b) 
of this chapter.
    (iv) Calculations. Calculate the micrograms of cefpiramide per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.189

where:
    Au=Area of the cefpiramide peak in the chromatogram of 
the sample (at a retention time equal to that observed for the 
standard);
    As=Area of the cefpiramide peak in the chromatogram of 
the cefpiramide working standard;
    Ps=Cefpiramide activity in the cefpiramide working 
standard solution in micrograms per milliliter;
    Cu=Milligrams of cefpiramide sample per milliliter of 
sample solution; and
    m=Percent moisture content of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous suspension containing 5 milligrams of cefpiramide per 
milliliter.
    (4) Total related substances. Proceed as directed in paragraph 
(b)(1) of this section except use the following reagents, standard and 
sample solutions, and calculations:
    (i) Reagents--(A) 0.03M phosphate buffer. Dissolve 4.08 grams of 
monobasic potasssium phosphate in 800 milliliters of water. Adjust the 
pH to 7.5 with 1N sodium hydroxide and dilute to 1,000 milliliters with 
water.
    (B) Mobile phase. Mix 0.03M phosphate buffer: methanol (750:250). 
Filter through a suitable filter capable of removing particulate matter 
to 0.5 micron in diameter. Degas the mobile phase just prior to its 
introduction into the chromatograph.
    (ii) Preparation of working standard and sample solutions.
    (A) Working standard solution. Transfer about 12.5 milligrams of 5-
mercapto-1-methyl-1H-tetrazole (MMT) and an amount of cefpiramide 
working standard equivalent to about 25 milligrams of cefpiramide 
activity, both accurately weighed, to a 100-milliliter volumetric flask. 
Dissolve and dilute to volume with 0.03M phosphate buffer. Further 
dilute 2.0 milliliters of this solution to 100 milliliters with mobile 
phase.
    (B) Sample solution. Transfer about 25 milligrams of the test 
material, accurately weighed, to a 50-milliliter volumetric flask. 
Dissolve and dilute to volume with mobile phase.
    (iii) Calculations. Calculate the percentages, individually, of MMT 
and any other compounds detected as follows:

[[Page 645]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.190


where:
    Au=Area of the tetrazole sample peak;
    As=Area of the tetrazole working standard peak;
    Cs=Concentration of the working standard in milligrams 
per milliliter;
    Ps=Potency of the working standard in micograms per 
milligram;
    Cu=Concentration of the sample solutions in milligrams 
per milliliter;
    Ru=Sum of peak areas of other compounds, excepting MMT 
and cefpiramide, detected in the sample chromatogram.
    Rs=Area of the cefpiramide working standard peak; and
    Lu=Area of the largest related peak, except MMT.
    T=Percent total related compounds=T1+T2.

    (5) Specific rotation. Dilute an accurately weighed sample with 
sufficient dimethylformamide to obtain a concentration of approximately 
10 milligrams of cefpiramide per milliliter. Proceed as directed in 
Sec. 436.210 of this chapter, using a 1-decimeter polarimeter tube. 
Calculate the specific rotation on the anhydrous basis.
    (6) Identify. Proceed as directed in Sec. 436.211 of this chapter 
using a 1-percent potassium bromide disc prepared as directed in 
Sec. 436.211(b)(1).
    (7) Chrystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[55 FR 14240, Apr. 17, 1990]



Sec. 442.69  Cefmetazole.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefmetazole is (6R,7S)-7-[2-
[(cyanomethyl)thio]acetamido]-7-methoxy-3-[[(1-methyl-1H-tetrazol-5-
yl)thio]methyl]-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylic 
acid. It is so purified and dried that:
    (i) Its potency is not less than 970 micrograms of cefmetazole 
activity per milligram.
    (ii) Its moisture content is not more than 0.5 percent.
    (iii) It gives a positive identity test for cefmetazole.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 442.70a(b)(1).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Identity. Proceed as directed in Sec. 436.211 of this chapter 
using a mineral oil mull prepared as described in paragraph (b)(2) of 
that section.
[59 FR 12546, Mar. 17, 1994]



Sec. 442.70a  Sterile cefmetazole sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile cefmetazole sodium is the sodium 
salt of (6R-cis)-7-[[[cyanomethyl)thio]acetyl]amino]-7-methoxy-3-[[(1-
methyl-1H-tetrazol-5-yl)thio]methyl]-8-oxo-5-thia-1-
azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid. It is a lyophilized 
powder. It is so purified and dried that:
    (i) If the cefmetazole sodium is not packaged for dispensing, its

[[Page 646]]

cefmetazole potency is not less than 860 micrograms and not more than 
1,003 micrograms of cefmetazole activity per milligram on an anhydrous 
basis. If the cefmetazole sodium is packaged for dispensing, its 
cefmetazole potency is not less than 860 micrograms and not more than 
1,003 micrograms of cefmetazole activity per milligram on an anhydrous 
basis and also, each container contains not less than 90 percent and not 
more than 120 percent of the number of milligrams of cefmetazole that it 
is represented to contain.
    (ii) It is sterile.
    (iii) It contains not more than 0.2 endotoxin units per milligram.
    (iv) Its moisture content is not more than 0.5 percent.
    (v) The pH of an aqueous solution containing 100 milligrams per 
milliliter is not less than 4.2 and not more than 6.2.
    (vi) It gives a positive identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cefmetazole potenty 
and content (if packaged for dispensing), sterility, bacterial 
endotoxins, moisture, pH, and identity.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research:
    (A) If the batch is packaged for repacking or for use as an 
ingredient in the manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (2) For sterility testing: 20 packages, each containing equal 
portions of approximately 300 milligrams.
    (B) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers of the batch.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 214 nanometers, a 25-
centimeter X 4.0- or 4.6-millimeter (inside diameter) column packed with 
microparticulate (5 micrometers in diameter) reversed phase packing 
material such as octadecyl silane bonded to silicas, a flow rate of not 
more than 2.0 milliliters per minute, and a known injection volume of 
between 10 and 20 microliters. Mobile phase, working standard and sample 
solutions, resolution test solution, system suitability requirements, 
and calculations are as follows:
    (i) Mobile phase. Transfer 5.75 grams of ammonium dihydrogen 
phosphate to a 1-liter container. Add 700 milliliters of deionized water 
and agitate to aid dissolution. Transfer 3.2 milliliters of 40 percent 
tetrabutylammonium hydroxide (TBAH) in distilled water to the solution 
and shake. Add 280 milliliters of methanol and a range 20 to 30 
milliliters of tetrahydrofuran and mix well. Adjust the pH to 
4.50.1 with phosphoric acid. The mobile phase is 0.05M 
ammonium dihydrogen phosphate: methanol: tetrahydrofuran (700:280:20-
30). It is 0.005M with respect to TBAH. Filter the mobile phase through 
a suitable filter capable of removing particulate matter to 0.5 micron 
in diameter and degas it just prior to its introduction into the 
chromatograph.
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(A) Working standard solution. Dissolve and dilute and 
accurately weighed portion of the cefmetazole working standard in 
sufficient mobile phase to obtain a solution containing 0.2 milligram of 
cefmetazole activity per milliliter. Analyze this solution within 10 
minutes.
    (B) Sample solutions--(1) Product not packaged for dispensing 
(micrograms of cefmetazole per milligram). Dissolve an accurately 
weighed sample with sufficient mobile phase to obtain a solution 
containing approximately 0.2 milligram of cefmetazole per milliliter 
(estimated). Analyze this solution within 10 minutes.
    (2) Product packaged for dispensing. Determine both micrograms of 
cefmetazole per milligram of sample and milligrams of cefmetazole per 
container. Use separate containers for

[[Page 647]]

preparation of each sample solution as described in paragraphs 
(b)(1)(ii)(B)(2)(i) and (ii) of this section.
    (i) Micrograms of cefmetazole per milligram. Dissolve an accurately 
weighed sample with sufficient mobile phase to obtain a solution 
containing approximately 0.2 milligram of cefmetazole per milliliter 
(estimated). Analyze this solution within 10 minutes.
    (ii) Milligrams of cefmetazole per container. Reconstitute the 
sample as directed in the labeling. Then, using a suitable hypodermic 
needle and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency in a given volume of the resultant preparation, 
remove an accurately measured representative portion from each 
container. Dilute the solution thus obtained with sufficient distilled 
water to obtain a solution containing 1,000 micrograms of cefmetazole 
activity per milliliter (estimated). Further dilute this solution with 
mobile phase to obtain a solution containing 0.2 milligram of 
cefmetazole activity per milliliter (estimated). Analyze this solution 
within 10 minutes.
    (C) Resolution test solution. Dissolve an accurately weighed portion 
of cefmetazole working standard in 0.01N sodium hydroxide to obtain a 
solution containing approximately 1.0 milligram of cefmetazole activity 
per milliliter. Heat this solution at 95  deg.C for 10 minutes. This 
procedure generates cefmetazole lactone. Dilute 1.0 milliliter of this 
solution to 20 milliliters with mobile phase.
    (iii) System suitability requirements--(A) Asymmetry factor. 
Calculate the asymmetry factor (A s), measured at a point 10 
percent of the peak height from the baseline as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.191

where:

a=Horizontal distance from point of ascent to point of maximum peak 
          height; and
b=Horizontal distance from point of maximum peak height to point of 
          descent.


The asymmetry factor (As) is satisfactory if it is not less 
than 0.94 and not more than 1.6.
    (B) Efficiency of the column. From the number of theoretical plates 
(n) calculated as described in Sec. 436.216(c)(2) of this chapter 
calculate the reduced plate height (hr) as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.192

where:

L = Length of the column in centimeters;
n = Number of theoretical plates; and
dp = Average diameter of the particles in the analytical 
          column packing in micrometers.


The absolute efficiency (hr) is satisfactory if it is not 
more than 20 for the cefmetazole peak.
    (C) Resolution factor. The resolution factor (R) between the peak 
for cefmetazole and the peak for cefmetazole lactone (generated in situ) 
is satisfactory if it is not less than 3.0.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent of 5 replicate 
injections) is satisfactory if it is not more than 2.0 percent.
    (E) Capacity factor (k'). Calculate the capacity factor (k') for 
cefmetazole as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.193

where:

tr=Retention time of cefmetazole in minutes; and
to=Column dead time in minutes, which is estimated from the 
          following equation:
          [GRAPHIC] [TIFF OMITTED] TR01JA93.194
          
where:

D=Column diameter in centimeters;
L=Column length in centimeters;
0.75=Average total column porosity; and
F=Flow rate in milliliters per minute.


The capacity factor (k') for cefmetazole is satisfactory if it is not 
less than 2.0 and not more than 8.0. If the system suitability 
parameters have been met, then proceed as described in Sec. 436.216(b) 
of this chapter.
    (iv) Calculations--(A) Cefmetazole potency (micrograms of 
cefmetazole per milligram). Calculate the micrograms of

[[Page 648]]

cefmetazole per milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.195

where:

Au=Area of the cefmetazole peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefmetazole peak in the chromatogram of the 
          cefmetazole working standard;
Ps=Cefmetazole activity in the cefmetazole working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of cefmetazole sample per milliliter of sample 
          solution; and
m=Percent moisture content of the sample.
(B) Cefmetazole content (milligrams of cefmetazole per container). 
          Calculate the cefmetazole content of the container as follows:
          [GRAPHIC] [TIFF OMITTED] TR01JA93.196
          
where:

Au=Area of the cefmetazole peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefmetazole peak in the chromatogram of the 
          cefmetazole working standard;
Ps=Cefmetazole activity in the cefmetazole working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in Sec. 436.20(e)(1).
    (3) Bacterial endotoxins. Proceed as directed in the United States 
Pharmacopeia bacterial endotoxins test.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter 
using a mineral oil mull prepared as described in Sec. 436.211(b)(2).
[55 FR 6634, Feb. 26, 1990]



Sec. 442.80  Cefprozil.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefprozil is an approximate 9:1 mixture 
of the Z (cis) and the E (trans) isomers, respectively, of (6R,7R)-7-
[(R)-2-amino-2-(p-hydroxyphenyl)acetamido]8-oxo-3-propenyl-5-thia-1-
azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid. It is so purified and 
dried that:
    (i) Its potency is not less than 900 micrograms nor more than 1,050 
micrograms of cefprozil activity per milligram, on an anhydrous basis.
    (ii) The ratio of its (E) isomer to total cefprozil is not less than 
0.06 nor more than 0.11.
    (iii) Its moisture content is not less than 3.5 percent nor more 
than 6.5 percent.
    (iv) The pH of an aqueous solution containing 5 milligrams per 
milliliter is not less than 3.5 nor more than 6.5.
    (v) It is crystalline.
    (vi) It gives positive identity tests.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cefprozil potency, 
E isomer to total cefprozil ratio, moisture, pH, crystallinity, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 280 nanometers, a 25 
centimeter  x  3.9 to 4.6 millimeter (id) column packed with 
microparticulate (5 to 10 micrometers in diameter) reversed phase 
packing material such as octadecyl silane bonded to silicas, a flow rate 
of 1.0 milliliter per minute, and a known injection volume of 10 
microliters. The retention time for cefprozil (Z) is between 4 and 6 
minutes and the retention time for cefprozil (E) is between 6 and 8 
minutes. Mobile phase, working standard

[[Page 649]]

and sample solutions, system suitability requirements, and calculations 
are as follows:
    (i) Mobile phase. Dissolve 20.7 grams of ammonium phosphate, 
monobasic in 1,800 milliliters of water and adjust the pH to 4.4 with 
phosphoric acid, if necessary. Add 200 milliliters of acetonitrile and 
mix. Filter the mobile phase through a suitable filter capable of 
removing particulate matter 0.5 micron in diameter and degas it just 
prior to its introduction into the chromatograph. The proportion of 
acetonitrile may be modified in the range of 6 to 14 percent to obtain 
the desired retention times. Increasing the amount of acetonitrile will 
decrease both the retention times and the separation between the 
isomers, whereas, decreasing the amount of acetonitrile will increase 
retention times and the separation between the isomers.
    (ii) Preparation of working standard solutions--(A) Cefprozil (Z) 
working standard solution. Accurately weigh approximately 12.5 
milligrams of the cefprozil (Z) working standard into a 50-milliliter 
volumetric flask. Dilute to volume with water and shake the flask 
vigorously until the solute dissolves completely. Use this solution 
within 6 hours.
    (B) Cefprozil (E) working standard solution. Accurately weigh 
approximately 12.5 milligrams of the cefprozil (E) working standard into 
a 50-milliliter volumetric flask. Dilute to volume with water and shake 
the flask vigorously until the solute dissolves completely. Pipet 5 
milliliters into a 50-milliliter volumetric flask, dilute to volume with 
water and mix thoroughly. Use this solution within 6 hours.
    (iii) Sample solution. Accurately weigh approximately 15 milligrams 
of sample into a 50-milliliter volumetric flask. Dilute to volume with 
water and shake the flask vigorously until the solute dissolves 
completely. Use this solution within 6 hours.
    (iv) System suitability requirements--(A) Asymmetry factor. The 
asymmetry factor (AS)is satisfactory if it is not less than 
0.9 and not more than 1.1 for the cefprozil (Z) response.
    (B) Efficiency of the column. The absolute efficiency 
(hr) is satisfactory if it is not more than 10 for the 
cefprozil (Z) response.
    (C) Resolution factor. The resolution factor (R) between the 
response for cefprozil (Z) and the response for cefprozil (E) is 
satisfactory if it is not less than 2.5.
    (D) Coefficient of variation (Relative standard deviation). The 
coefficient of variation (SRof 5 replicate injections of the 
cefprozil (Z) reference solution response) is satisfactory if it is not 
more than 2.0 percent.
    (E) Capacity factor (k'). The capacity factor (k') for cefprozil (Z) 
is satisfactory if it is not less than 0.7 and not more than 1.1. If the 
system suitability parameters have been met, then proceed as described 
in Sec. 436.216(b) of this chapter.
    (v) Calculations. Calculate the micrograms of cefprozil per 
milligram of sample on an anhydrous basis as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.047

where:
AU = Area of the cefprozil (Z) or cefprozil (E) response in 
          the chromatogram of the sample (at a retention time equal to 
          that observed for the standard);
AS = Area of the cefprozil (Z) or cefprozil (E) response in 
          the chromatogram of the cefprozil (Z) or the cefprozil (E) 
          working standard ;
PS = Cefprozil (Z) or cefprozil (E) activity in the cefprozil 
          (Z) or the cefprozil (E) working standard solution in 
          micrograms per milliliter;
CU = Milligrams of sample per milliliter of sample solution; 
          and
m = Percent moisture content of the sample.

    (2) Cefprozil (E)/cefprozil ratio. Using the procedure described in 
paragraph (b)(1) of this section calculate the cefprozil (E)/cefprozil 
ratio as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.048


[[Page 650]]


    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
carbon dioxide free aqueous solution containing 5 milligrams of 
cefprozil per milliliter.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (6) Identity--(i) Infrared. Proceed as directed in Sec. 436.211 of 
this chapter, using a 1.0 percent potassium bromide disc prepared as 
described in paragraph (b)(1) of that section.
    (ii) High performance liquid chromatography (HPLC). The HPLC 
retention times for the responses of the cefprozil isomers in the assay 
preparation of the sample must be within 2 percent of the HPLC retention 
times of the responses of the corresponding cefprozil working standards.
[58 FR 26660, May 4, 1993]



                      Subpart B--Oral Dosage Forms



Sec. 442.104  Cefaclor monohydrate oral dosage forms.



Sec. 442.104a  Cefaclor monohydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefaclor monohydrate capsules are 
composed of cefaclor monohydrate and one or more suitable and harmless 
lubricants and diluents enclosed in a gelatin capsule. Each capsule 
contains cefaclor monohydrate equivalent to either 250 milligrams or 500 
milligrams of cefaclor. Its potency is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the number of 
milligrams of cefaclor that it is represented to contain. Its moisture 
content is not more than 8.0 percent. The cefaclor monohydrate used 
conforms to the standards prescribed by Sec. 442.4(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cefaclor monohydrate used in making the batch for potency, 
moisture, pH, identity, and crystallinity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The cefaclor monohydrate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, except prepare the working 
standard and sample solutions and calculate the potency of the sample as 
follows:
    (i) Preparation of working standard solution. Dissolve and dilute an 
accurately weighed portion of the cefaclor working standard in 
sufficient 0.1M potassium phosphate buffer, pH 4.5 (as described in 
Sec. 436.101(a)(4) of this chapter) to obtain a concentration of 1 
milligram of cefaclor per milliliter.
    (ii) Preparation of sample solution. Place one capsule into a high-
speed glass blender jar containing sufficient 0.1M potassium phosphate 
buffer, pH 4.5 (as described in Sec. 436.101(a)(4) of this chapter) to 
obtain a concentration of 1 milligram of cefaclor per milliliter. Filter 
a portion to be used through a 10-micron filter.
    (iii) Calculations. Calculate the cefaclor content in milligrams per 
capsule as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.049

where:

Au = Absorbance of sample solution;
Pa = Potency of working standard in micrograms per 
          milliliter;
As = Absorbance of working standard solution;
d= Dilution factor of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[46 FR 3833, Jan. 16, 1981; 46 FR 21360, Apr. 10, 1981]



Sec. 442.104b  Cefaclor monohydrate for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefaclor monohydrate for oral suspension 
is cefaclor monohydrate with one or more suitable

[[Page 651]]

and harmless diluents, buffer substances, colorings and flavorings. When 
reconstituted as directed in the labeling, each milliliter contains 
cefaclor monohydrate equivalent to 25 milligrams, 37.5 milligrams, 50 
milligrams, or 75 milligrams of cefaclor. Its potency is satisfactory if 
it is not less than 90 percent and not more than 120 percent of the 
number of milligrams of cefaclor that it is represented to contain. Its 
moisture content is not more than 2.0 percent. When reconstituted as 
directed in the labeling, its pH is not less than 2.5 and not more than 
5.0. The cefaclor monohydrate used conforms to the standards prescribed 
by Sec. 442.4(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cefaclor monohydrate used in making the batch for potency, 
moisture, pH, identity, and crystallinity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The cefaclor monohydrate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, except prepare the working 
standard and sample solutions and calculate the potency of the sample as 
follows:
    (i) Preparation of working standard solution. Dissolve and dilute an 
accurately weighed portion of the cefaclor working standard in 
sufficient 0.1M potassium phosphate buffer, pH 4.5 (as described in 
Sec. 436.101(a)(4) of this chapter) to obtain a concentration of 1 
milligram of cefaclor per milliliter.
    (ii) Preparation of sample solution. Reconstitute the sample as 
directed in the labeling. Transfer a 5.0-milliliter portion into an 
appropriate-sized volumetric flask and dilute to volume with 0.1M 
potassium phosphate buffer, pH 4.5 (as described in Sec. 436.101(a)(4) 
of this chapter) to obtain a concentration of 1 milligram of cefaclor 
per milliliter.
    (iii) Calculations. Calculate the cefaclor content as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.050
    
where:

Au=Absorbance of sample solution;
Pa=Potency of working standard in micrograms per milliliter;
As=Absorbance of working standard solution;
d=Dilution factor of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
[46 FR 3833, Jan. 16, 1981; 46 FR 21360, Apr. 10, 1981, as amended at 47 
FR 22515, May 25, 1982; 54 FR 41824, Oct. 12, 1989]



Sec. 442.106  Cefadroxil monohydrate oral dosage forms.



Sec. 442.106a  Cefadroxil monohydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefadroxil monohydrate capsules are 
composed of cefadroxil monohydrate and one or more suitable and harmless 
lubricants and diluents enclosed in a gelatin capsule. Each capsule 
contains either 250 or 500 milligrams of cefadroxil. Its moisture 
content is not more than 7.0 percent. The cefadroxil monohydrate used 
conforms to the standards prescribed by Sec. 442.6(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cefadroxil monohydrate used in making the batch for potency, 
moisture, pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The cefadroxil monohydrate used in making the batch: 10 
packages, each containing approximately 500 milligrams.

[[Page 652]]

    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the hydroxylamine 
colorimetric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 1 percent potassium phosphate buffer, 
pH 6.0 (solution 1), to give a stock solution of convenient 
concentration. Blend for 3 to 5 minutes. Remove an aliquot and further 
dilute with solution 1 to the reference concentration of 20.0 micrograms 
of cefadroxil per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, preparing the sample as follows: 
Blend a representative number of capsules in a high-speed glass blender 
jar with sufficient distilled water to give a stock solution of 
convenient concentration. Further dilute an aliquot of this solution 
with distilled water to a concentration of 1 milligram of cefadroxil per 
milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[43 FR 20978, May 16, 1978; 43 FR 27180, June 23, 1978; 45 FR 16472, 
Mar. 14, 1980. Redesignated and amended at 46 FR 2992, Jan. 13, 1981; 50 
FR 19919, May 13, 1985]



Sec. 442.106b  Cefadroxil monohydrate tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefadroxil monohydrate tablets are 
composed of cefadroxil monohydrate and one or more suitable and harmless 
binders and lubricants, and with or without coloring and film-coating 
substances. Each tablet contains cefadroxil monohydrate equivalent to 
1,000 milligrams of cefadroxil. Its potency is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of cefadroxil that it is represented to contain. Its moisture 
content is not more than 8.0 percent. The tablets disintegrate within 15 
minutes. The cefadroxil monohydrate used conforms to the standards 
prescribed by Sec. 442.6(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cefadroxil monohydrate used in making the batch for potency, 
moisture, pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency, moisture, and disintegration time.
    (ii) Samples required:
    (a) The cefadroxil monohydrate used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the hydroxylamine 
colorimetric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to obtain a stock solution of convenient concentration. 
Blend for 3 to 5 minutes. Further dilute an aliquot of the stock 
solution with solution 1 to the reference concentration of 20 micrograms 
of cefadroxil per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, except prepare the working 
standard and sample solutions and calculate the cefadroxil content as 
follows:
    (a) Preparation of working standard solution. Dissolve and dilute an 
accurately weighed portion of the cefadroxil working standard in 
sufficient distilled water to a final concentration of 1 milligram of 
cefadroxil per milliliter.
    (b) Preparation of sample solution. Blend a representative number of 
tablets in a high-speed glass blender jar

[[Page 653]]

with sufficient distilled water to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of this solution with distilled 
water to a concentration of 1 milligram of cefadroxil per milliliter 
(estimated).
    (c) Calculations. Calculate the cefadroxil content as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.051
    
where:

    Au=Absorbance of sample solution;
    Ps=Potency of working standard in micrograms per 
milligram;
    d=Dilution factor for sample;
    As=Absorbance of working standard solution;
    n=Number of tablets in the sample assayed.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure described in paragraph (e)(1) of that 
section.
[46 FR 2992, Jan. 13, 1981; 46 FR 15880, Mar. 10, 1981, as amended at 50 
FR 19919, May 13, 1985; 54 FR 47352, Nov. 14, 1989]



Sec. 442.106c  Cefadroxil monohydrate for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefadroxil monohydrate for oral 
suspension is cefadroxil monohydrate with one or more suitable and 
harmless preservatives, suspending agents, surfactants, binders, and 
flavorings. When reconstituted as directed in the labeling, each 
milliliter contains cefadroxil monohydrate equivalent to either 25, 50, 
or 100 milligrams of cefadroxil. Its potency is satisfactory if it is 
not less than 90 percent and not more than 120 percent of the number of 
milligrams of cefadroxil that it is represented to contain. Its moisture 
content is not more than 2.0 percent. When reconstituted as directed in 
the labeling, its pH is not less than 4.5 and not more than 6.0. The 
cefadroxil monohydrate used conforms to the standards prescribed by 
Sec. 442.6(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cefadroxil monohydrate used in making the batch for potency, 
moisture, pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The cefadroxil monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the hydroxylamine 
colorimetric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute the sample as directed in the labeling. Transfer an 
accurately measured representative portion of the suspension into an 
appropriate-sized volumetric flask and dilute to volume with 1 percent 
potassium phosphate buffer, pH 6.0 (solution 1). Further dilute an 
aliquot of this solution with solution 1 to the reference concentration 
of 20.0 micrograms of cefadroxil per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, except prepare the working 
standard and sample solutions and calculate the cefadroxil content as 
follows:
    (a) Preparation of working standard solution. Dissolve and dilute an 
accurately weighed portion of the cefadroxil working standard in 
sufficient distilled water to a final concentration of 1 milligram of 
cefadroxil per milliliter.
    (b) Preparation of sample solution. Reconstitute the sample as 
directed in the labeling. Transfer an accurately measured representative 
portion to a volumetric flask and bring to volume with distilled water 
to give a stock solution of convenient concentration.

[[Page 654]]

Further dilute an aliquot of this solution with distilled water to a 
concentration of 1 milligram of cefadroxil per milliliter (estimated).
    (c) Calculations. Calculate the cefadroxil content as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.052
    
where:
Au = Absorbance of sample solution;
Ps = Potency of working standard in micrograms per milligram;
d=Dilution factor for sample;
As = Absorbance of working standard solution.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
[46 FR 16679, Mar. 13, 1981, as amended at 50 FR 19919, May 13, 1985]



Sec. 442.107  Cefadroxil hemihydrate oral dosage forms.



Sec. 442.107a  Cefadroxil hemihydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefadroxil hemihydrate capsules are 
composed of cefadroxil hemihydrate and one or more suitable and harmless 
lubricants and diluents enclosed in a gelatin capsule. Each capsule 
contains cefadroxil hemihydrate equivalent to 500 milligrams of 
cefadroxil. Its cefadroxil content is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the number of 
milligrams of cefadroxil that it is represented to contain. Its moisture 
content is not more than 7.0 percent. It passes the dissolution test. 
The cefadroxil hemihydrate used conforms to the standards prescribed in 
Sec. 442.7(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefadroxil hemihydrate used in making the batch for potency, 
moisture, pH, absorptivity, identity, and crystallinity.
    (B) The batch for content, moisture, and dissolution.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The cefadroxil hemihydrate used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (B) The batch: A minimum of 100 capsules.
    (b) Tests and methods of assay--(1) Cefadroxil content. Use either 
of the following methods; however, the results obtained from the 
hydroxylamine colorimetric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 1 percent potassium phosphate buffer, 
pH 6.0 (solution 1), to give a stock solution of convenient 
concentration. Blend for 3 to 5 minutes. Remove an aliquot and further 
dilute with solution 1 to the reference concentration of 20 micrograms 
of cefadroxil per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay for cefadroxil. Proceed as 
directed in Sec. 442.40(b)(1)(ii), except prepare the working standard 
and sample solutions and calculate the potency of the sample as follows:
    (A) Preparation of working standard solutions. Dissolve and dilute 
an accurately weighed portion of the cefadroxil working standard in 
sufficient distilled water to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of this solution with distilled 
water to a concentration of 1 milligram of cefadroxil per milliliter.
    (B) Preparation of sample solutions. Blend a representative number 
of capsules in a high-speed glass blender jar with sufficient distilled 
water to obtain a stock solution of convenient concentration. Further 
dilute an aliquot of this solution with distilled water to a 
concentration of 1 milligram of cefadroxil per milliliter (estimated).
    (C) Calculations. Calculate the cefadroxil content as follows:

[[Page 655]]

[GRAPHIC] [TIFF OMITTED] TC01AP94.053


where:
AU = Absorbance of sample solution;
AS = Absorbance of working standard solution;
PS = Potency of working standard solution in micrograms per 
          milliliter;
d = Dilution factor of the sample;
n = Number of capsules in the sample assayed.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q (the amount of cefadroxil dissolved) is 75 
percent within 45 minutes.
[59 FR 8857, Feb. 24, 1994]



Sec. 442.107b  Cefadroxil hemihydrate tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefadroxil hemihydrate tablets are 
composed of cefadroxil hemihydrate and one or more suitable and harmless 
binders and lubricants, with or without coloring and film-coating 
substances. Each tablet contains cefadroxil hemihydrate equivalent to 
1,000 milligrams of cefadroxil. Its cefadroxil content is satisfactory 
if it is not less than 90 percent and not more than 120 percent of the 
number of milligrams of cefadroxil that it is represented to contain. 
Its moisture content is not more than 8.0 percent. It passes the 
dissolution test. The cefadroxil hemihydrate used conforms to the 
standards prescribed in Sec. 442.7(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefadroxil hemihydrate used in making the batch for potency, 
moisture, pH, absorptivity, identity, and crystallinity.
    (B) The batch for content, moisture, and dissolution.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The cefadroxil hemihydrate used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (B) The batch: A minimum of 100 tablets.
    (b) Tests and methods of assay--(1) Cefadroxil content. Use either 
of the following methods; however, the results obtained from the 
hydroxylamine colorimetric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to give a stock solution of convenient concentration. 
Blend for 3 to 5 minutes. Remove an aliquot and further dilute with 
solution 1 to the reference concentration of 20 micrograms of cefadroxil 
per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay for cefadroxil. Proceed as 
directed in Sec. 442.40(b)(1)(ii), except prepare the working standard 
and sample solutions and calculate the potency of the sample as follows:
    (A) Preparation of working standard solutions. Dissolve and dilute 
an accurately weighed portion of the cefadroxil working standard in 
sufficient distilled water to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of this solution with distilled 
water to a concentration of 1 milligram of cefadroxil per milliliter.
    (B) Preparation of sample solutions. Blend a representative number 
of tablets in a high-speed glass blender jar with sufficient distilled 
water to obtain a stock solution of convenient concentration. Further 
dilute an aliquot of this solution with distilled water to a 
concentration of 1 milligram of cefadroxil per milliliter (estimated).
    (C) Calculations. Calculate the cefadroxil content as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.054
    
where:
AU = Absorbance of sample solution;
AS = Absorbance of working standard solution;

[[Page 656]]

PS = Potency of working standard solution in micrograms per 
          milliliter;
d = Dilution factor of the sample; and
n = Number of tablets in the sample assayed.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q (the amount of cefadroxil dissolved) is 75 
percent within 30 minutes.
[59 FR 8857, Feb. 24, 1994]



Sec. 442.115  Cefixime trihydrate oral dosage forms.



Sec. 442.115a  Cefixime trihydrate for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefixime trihydrate for oral suspension 
is cefixime trihydrate with one or more suitable and harmless 
preservatives, suspending agents, diluents, and flavorings. When 
reconstituted as directed in the labeling, each milliliter contains the 
equivalent of 20 milligrams of cefixime. Its cefixime trihydrate potency 
is satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of cefixime that it is represented 
to contain. Its moisture content is not more than 2.0 percent. When 
reconstituted as described in labeling, the pH of the suspension is not 
less than 2.5 and not more than 4.5. It passes the identity test for the 
presence of the cefixime moiety. The cefixime trihydrate used conforms 
to the standards prescribed by Sec. 442.15(a)(1) of this part.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 436.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefixime trihydrate used in making the batch, for potency, 
moisture, pH, crystallinity, specific rotation, and identity.
    (B) The batch, for content, moisture, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research.
    (A) The cefixime used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch: A minimum of 10 immediate containers.
    (b) Tests and methods of assay--(1) Content. Proceed as directed in 
Sec. 442.15(b)(1) of this part, preparing the sample solution and 
calculating the cefixime content as follows:
    (i) Preparation of the sample solution. Reconstitute as directed in 
the labeling. Transfer a 5.0-milliliter portion of the suspension into 
an appropriately sized volumetric flask and quantitatively dilute 
stepwise with 0.1M phosphate buffer, pH 7.0, to obtain a concentration 
of 0.2 milligram of cefixime activity per milliliter (estimated).
    (ii) Calculations. Calculate the cefixime content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.197
    
where:
Au = Area of the cefixime peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As = Area of the cefixime peak in the chromatogram of the 
          cefixime working standard.
Ps = Cefixime activity in the cefixime working standard 
          solution in micrograms per milliliter; and
d = Dilution factor of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
    (4) Identity. The high performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section, compares 
qualitatively to that of the cefixime working standard.
[53 FR 24259, June 28, 1988]



Sec. 442.115b  Cefixime trihydrate tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefixime trihydrate tablets are composed 
of cefixime trihydrate and one or more suitable and harmless diluents, 
binders, lubricants, colorings, and coating substances. Each tablet

[[Page 657]]

contains cefixime trihydrate equivalent to either 200 milligrams or 400 
milligrams of cefixime. Its cefixime trihydrate content is satisfactory 
if it is not less than 90 percent and not more than 110 percent of the 
number of milligrams of cefixime that it is represented to contain. Its 
moisture content is not more than 10.0 percent. It passes the 
dissolution test. It passes the identity test for the presence of the 
cefixime moiety. The cefixime used conforms to the standards prescribed 
by Sec. 442.15(a)(1) of this part.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefixime used in making the batch for potency, moisture, pH, 
crystallinity, specific rotation, and identity.
    (B) The batch, for content, moisture, dissolution, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research.
    (A) The cefixime used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch: A minimum of 10 immediate containers.
    (b) Tests and methods of assay--(1) Content. Proceed as directed in 
Sec. 442.15(b)(1) of this part, preparing the sample solution and 
calculating the cefixime content as follows:
    (i) Preparation of sample solution. Grind one or a known number of 
tablets using a mortar and pestle. Quantitatively transfer the ground 
tablet(s) into a suitable volumetric flask, sonicate and dilute with 
0.1M phosphate buffer, pH 7.0 to a concentration of 4 milligrams per 
milliliter. Centrifuge the sample at 3,000 revolutions per minute for 10 
minutes. Take an aliquot of the supernatant and qualitatively dilute to 
a concentration of 0.2 milligram of cefixime activity per milliliter in 
0.1M phosphate buffer, pH 7.0 (estimated).
    (ii) Calculations. Calculate the cefixime content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.198
    
where:
Au = Area of the cefixime peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As = Area of the cefixime peak in the chromatogram of the 
          cefixime working standard.
Ps = Cefixime activity in the cefixime working standard 
          solution in micrograms per milliliter;
d = Dilution factor of the sample; and
n = Number of tablets in the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Dissolution test. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q (the amount of cefixime dissolved) is 75 percent 
within 45 minutes.
    (4) Identity. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the cefixime working standard.
[53 FR 24259, June 28, 1988]



Sec. 442.119  Cefuroxime axetil oral dosage forms.



Sec. 442.119a  Cefuroxime axetil tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefuroxime axetil tablets are composed of 
cefuroxime axetil and one or more suitable and harmless diluents, 
binders, lubricants, and colorings. Each tablet contains 125 milligrams, 
250 milligrams, or 500 milligrams of cefuroxime activity. Its potency is 
satisfactory if it is not less than 90 percent and not more than 110 
percent of the number of milligrams of cefuroxime activity that it is 
represented to contain. Its moisture content is not more than 2.0 
percent at the time of certification and not more than 6.0 percent at 
the time of expiry. It passes the dissolution test. It passes the film-
coat rupture test. It passes the identity test. The cefuroxime axetil 
used conforms to the standards prescribed by Sec. 442.19(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 658]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefuroxime axetil used in making the batch for potency, 
isomer A ratio, moisture, crystallinity, and identity.
    (B) The batch for potency, moisture, dissolution, film-coat rupture, 
and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The cefuroxime axetil used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (B) The batch: A minimum of 100 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 442.19(b)(1). Working standard and sample solutions, system 
suitability requirements, and calculations are as follows:
    (i) Preparation of working standard and sample solutions--(A) 
Working standard solution. Dissolve approximately 30 milligrams of the 
cefuroxime axetil working standard, accurately weighed, in methanol and 
dilute to 25 milliliters. Transfer 10.0 milliliters of the working 
standard solution to a 50-milliliter volumetric flask. Add 5.0 
milliliters of internal standard solution, 3.8 milliliters of methanol, 
and dilute to volume with 0.2M ammonium phosphate solution to obtain a 
stock solution containing 0.24 milligram of cefuroxime axetil per 
milliliter. Store the stock solution under refrigeration no more than 8 
hours.
    (B) Sample solution. Grind a representative number of tablets in a 
mortar and pestle. Immediately swirl the ground tablets in a volumetric 
flask containing methanol and shake for 10 minutes to dissolve the 
ground cefuroxime axetil. Dilute with methanol to give a stock solution 
of convenient concentration. Filter the stock solution. Transfer 5.0 
milliliters of filtrate to a 50-milliliter volumetric flask. Add 5.0 
milliliters of internal standard solution and 8.8 milliliters of 
methanol. Dilute to volume with 0.2M ammonium phosphate solution. Store 
in a refrigerator and use within 8 hours.
    (ii) System suitability requirements--(A) Tailing factor. The 
tailing factor (T) is satisfactory for isomer A if it is not more than 
1.5 at 5 percent of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory for isomer A if it is greater than 3,000 theoretical 
plates.
    (C) Resolution. The resolution (R) between isomer A and isomer B of 
cefuroxime axetil is satisfactory if it is not less than 1.5 and the 
resolution (R) between isomer A and the delta-2 isomers of cefuroxime 
axetil is satisfactory if it is not less than 1.5.
    (D) Coefficient of variation. The coefficient of variation (SR 
in percent) of five replicate injections is not more than 2.0 percent. 
If the system suitability requirements have been met, then proceed as 
described in Sec. 436.216(b) of this chapter. Alternate chromatographic 
conditions are acceptable provided reproducibility and resolution are 
comparable to the system. However, the sample preparation described in 
paragraph (b)(1)(i)(B) of this section should not be changed.
    (iii) Calculations. Calculate the cefuroxime content as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.055
    
where:
Ru=Sum of the peak heights of the cefuroxime axetil sample 
          isomer A and isomer B peaks/Peak height of the internal 
          standard;
Rs=Sum of the peak heights of the cefuroxime axetil working 
          standard isomer A and isomer B peaks/Peak height of the 
          internal standard;
Ps=Potency of the cefuroxime axetil working standard in 
          milligrams of cefuroxime activity per milliliter;
d=Dilution factor of the sample; and
n=Number of tablets in the sample assayed.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the titration procedure described in paragraph (e)(1) of that 
section.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q (the amount of cefuroxime activity dissolved) is 
60 percent at 15 minutes and 75 percent at 45 minutes.

[[Page 659]]

    (4) Film-coat rupture test. Proceed as directed in Sec. 436.217 of 
this chapter.
    (5) Identity. The high-performance liquid chromatogram of the sample 
solution determined as directed in paragraph (b)(1) of this section 
compares qualitatively to that of the cefuroxime axetil working standard 
solution.
[52 FR 42433, Nov. 5, 1987; 52 FR 45528, Nov. 30, 1987, as amended at 54 
FR 47352, Nov. 14, 1989; 54 FR 50472, Dec. 6, 1989; 55 FR 11583, Mar. 
29, 1990. Redesignated at 60 FR 27222, May 23, 1995]



Sec. 442.119b  Cefuroxime axetil for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefuroxime axetil for oral suspension is 
cefuroxime axetil with one or more suitable and harmless diluents, 
suspending and sweetening agents, and flavorings. When reconstituted as 
directed in the labeling, it contains cefuroxime axetil equivalent to 25 
milligrams of cefuroxime per millimeter. Its potency is satisfactory if 
it is not less than 90 percent and not more than 115 percent of the 
number of milligrams of cefuroxime that it is represented to contain. It 
passes the dissolution test. Its moisture content is not more than 0.2 
percent. When reconstituted as directed in the labeling, its pH is not 
less than 3.5 and not more than 5.5. It passes the identity test. The 
cefuroxime axetil used conforms to the standards prescribed by 
Sec. 442.19(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefuroxime axetil used in making the batch for potency, 
isomer A ratio, moisture, crystallinity, and identity.
    (B) The batch for cefuroxime potency, dissolution, moisture, pH of 
constituted suspension, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The cefuroxime axetil used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (B) The batch: A minimum of 12 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 442.19(b)(1). Working standard and sample solutions and 
calculations are as follows:
    (i) Preparation of working standard solution. Dissolve approximately 
15 milligrams of the cefuroxime axetil working standard, accurately 
weighed, in 20.0 milliliters of methanol in a 50-milliliter volumetric 
flask. Dilute to volume with deionized water, and swirl to mix. Store 
for no more than 8 hours under refrigeration and protected from light.
    (ii) Preparation of sample solution. Reconstitute the sample as 
directed in the labeling. Transfer an accurately measured representative 
portion of the suspension equivalent to one dose into a 200-milliliter 
volumetric flask. Add 10 milliliters of methanol and disperse the 
sample. Dilute to volume with methanol. Dilute 20.0 milliliters of this 
solution to volume in a 50-milliliter volumetric flask with deionized 
water, swirl to mix, and allow to stand for 10 minutes. (Note: A white 
turbidity is formed.) Filter this solution via a suitable disposable 
filter unit, discarding the first 5 milliliters. Store for no more than 
8 hours under refrigeration and protect from light.
    (iii) Calculations. Calculate the milligrams of cefuroxime per dose 
(5 milliliters) as follows:
[GRAPHIC] [TIFF OMITTED] TR23MY95.001

where:
AU = Sum of the areas of the cefuroxime axetil sample isomer 
          A and isomer B peaks;
AS = Sum of the peak areas of the cefuroxime axetil working 
          standard isomer A and isomer B peaks;
PS = Cefuroxime activity in the cefuroxime axetil working 
          standard solution in micrograms per milliliter; and
d = Dilution factor of the sample.

    (2) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q (the amount of cefuroxime activity dissolved) is 
60 percent at 30 minutes.

[[Page 660]]

    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Reconstitute as directed in the labeling and proceed as 
directed in Sec. 436.202 of this chapter.
    (5) Identity. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the cefuroxime axetil working standard.
[60 FR 27222, May 23, 1995]



Sec. 442.121  Cephaloglycin dihydrate oral dosage forms.



Sec. 442.121a  Cephaloglycin dihydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephaloglycin dihydrate capsules are 
composed of cephaloglycin dihydrate and one or more suitable lubricants 
and diluents enclosed in a gelatin capsule. Each capsule contains 
cephaloglycin dihydrate equivalent to 250 milligrams of cephaloglycin. 
Its potency is satisfactory if it is not less than 90 percent and not 
more than 120 percent of the number of milligrams of cephaloglycin that 
it is represented to contain. Its moisture content is not more than 9 
percent. The cephaloglycin used conforms to the standards prescribed by 
Sec. 442.21(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The cephaloglycin dihydrate used in making the batch for 
potency, moisture, pH, cephaloglycin content, identity, and 
crystallinity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The cephaloglycin dihydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar with sufficient 0.1M potassium phosphate buffer, pH 4.5 
(solution 4), to give a stock solution of convenient concentration. 
Blend for 3 to 5 minutes. Remove an aliquot and further dilute with 
solution 4 to the reference concentration of 10 micrograms of 
cephaloglycin per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19040, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 442.121b  Cephaloglycin dihydrate for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephaloglycin dihydrate for oral 
suspension is cephaloglycin dihydrate with one or more suitable 
diluents, buffer substances, colorings, and flavorings. When 
reconstituted as directed in the labeling, each milliliter contains 
cephaloglycin dihydrate equivalent to 50 milligrams of cephaloglycin. 
Its potency is satisfactory if it is not less than 90 percent and not 
more than 120 percent of the number of milligrams of cephaloglycin that 
it is represented to contain. Its moisture content is not more than 2 
percent. When reconstituted as directed in the labeling, its pH is not 
less than 3.0 and not more than 5.0. It passes the identity test for the 
presence of the cephaloglycin moiety. The cephaloglycin dihydrate used 
conforms to the standards prescribed by Sec. 442.21(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cephaloglycin dihydrate used in making the batch for 
potency, moisture, pH, cephaloglycin content, identity, and 
crystallinity.
    (b) The batch for potency, moisture, pH, and identity.
    (ii) Samples required:
    (a) The cephaloglycin dihydrate used in making the batch: 10 
packages, each

[[Page 661]]

containing approximately 500 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Place an accurately measured 
representative portion of the suspension into an appropriate-sized 
volumetric flask and dilute to volume with 0.1M potassium phosphate 
buffer, pH 4.5 (solution 4). Further dilute an aliquot of the stock 
solution with solution 4 to the reference concentration of 10 micrograms 
of cephaloglycin per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
    (4) Identity. Dilute a representative portion of the sample with 
sufficient distilled water to give a concentration of 2.5 milligrams of 
cephaloglycin per milliliter (estimated). Shake vigorously on a 
mechanical shaker for 30 minutes. Filter through Whatman No. 1 filter 
paper, discarding the first few milliliters of filtrate. Further dilute 
an aliquot of the filtrate with sufficient distilled water to give a 
concentration of 0.05 milligram of cephaloglycin per milliliter 
(estimated). Using a suitable spectrophotometer, record the ultraviolet 
absorption spectrum of this solution from 230 to 320 nanometers. The 
spectrum compares qualitatively to that of the cephaloglycin working 
standard similarly treated.
[39 FR 19040, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 442.127  Cephalexin monohydrate oral dosage forms.



Sec. 442.127a  Cephalexin monohydrate tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephalexin monohydrate tablets are 
composed of cephalexin monohydrate and one or more suitable and harmless 
diluents, binders, lubricants, colorings, and coating substances. Each 
tablet contains cephalexin monohydrate equivalent to 250, 500, or 1,000 
milligrams of cephalexin. Its potency is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the number of 
milligrams of cephalexin that it is represented to contain. Its moisture 
content is not more than 9 percent. The tablets disintegrate within 30 
minutes. The cephalexin monohydrate used conforms to the standards 
prescribed by Sec. 442.27(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cephalexin monohydrate used in making the batch for potency, 
moisture, pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency, moisture, and disintegration time.
    (ii) Samples required:
    (a) The cephalexin monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to give a stock solution of convenient concentration. 
Blend for 3 to 5 minutes. Further dilute with solution 1 to the 
reference concentration of 20.0 micrograms of cephalexin per milliliter 
(estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, preparing the sample as follows: Blend a representative number 
of tablets in a high-speed glass blender with sufficient distilled water 
to give a stock solution of convenient concentration. Further

[[Page 662]]

dilute with distilled water to the prescribed concentration of 
cephalexin.

    Note: The 10.0 milliliters of 0.01N iodine must be added within 20 
seconds after the addition of the 2.0 milliliters of 1.2N hydrochloric 
acid, and the assay should be completed within 1 hour after the sample 
and standard are first put into solution.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure described in paragraph (e)(1) of that 
section.
[39 FR 19040, May 30, 1974, as amended at 40 FR 49083, Oct. 21, 1975; 50 
FR 19919, May 13, 1985; 52 FR 20710, June 3, 1987]



Sec. 442.127b  Cephalexin monohydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephalexin monohydrate capsules are 
composed of cephalexin monohydrate and one or more suitable and harmless 
lubricants and diluents enclosed in a gelatin capsule. Each capsule 
contains cephalexin monohydrate equivalent to either 125, 250, or 500 
milligrams of cephalexin. Its potency is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the number of 
milligrams of cephalexin that it is represented to contain. Its moisture 
content is not more than 10 percent. The cephalexin monohydrate used 
conforms to the standards prescribed by Sec. 442.27(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cephalexin monohydrate used in making the batch for potency, 
moisture, pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The cephalexin monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar with sufficient 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to give a stock solution of convenient concentration. 
Blend for 3 to 5 minutes. Remove an aliquot and further dilute with 
solution 1 to the reference concentration of 20.0 micrograms of 
cephalexin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, preparing the sample as follows: Blend a representative number 
of capsules in a high-speed glass blender with sufficient distilled 
water to give a stock solution of convenient concentration. Further 
dilute with distilled water to the prescribed concentration of 
cephalexin.

    Note: The 10.0 milliliters of 0.01N iodine must be added within 20 
seconds after the addition of the 2.0 milliliters of 1.2N hydrochloric 
acid, and the assay should be completed within 1 hour after the sample 
and standard are first put into solution.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19040, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 442.127c  Cephalexin monohydrate for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephalexin monohydrate for oral 
suspension is cephalexin monohydrate with one or more suitable and 
harmless diluents, buffer substances, colorings, and flavorings. When 
reconstituted as directed in the labeling, each milliliter contains 
cephalexin monohydrate equivalent to 25 milligrams, 50 milligrams, or 
100 milligrams of cephalexin. Its potency is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of cephalexin that it is represented to contain. Its moisture 
content is not

[[Page 663]]

more than 2 percent. When reconstituted as directed in the labeling, its 
pH is not less than 3.0 and not more than 6.0. The cephalexin used 
conforms to the standards prescribed by Sec. 442.27(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cephalexin used in making the batch for potency, moisture, 
pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The cephalexin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute the sample as directed in the labeling. Transfer an 
accurately measured representative portion of the suspension into an 
appropriate-sized volumetric flask and dilute to volume with 1-percent 
potassium phosphate buffer, pH 6.0 (solution 1). Further dilute an 
aliquot of this solution with solution 1 to the reference concentration 
of 20.0 micrograms of cephalexin per milliliter (estimated).
    (ii) Iodometric assay. Proceed as directed in Sec. 436.204 of this 
chapter, preparing the sample as follows: Reconstitute the sample as 
directed in the labeling. Transfer an accurately measured representative 
portion to a volumetric flask and bring to volume with distilled water. 
Further dilute an aliquot of this solution with distilled water to the 
prescribed concentration of cephalexin.

    Note: The 10 milliliters of 0.01N iodine must be added within 20 
seconds after the addition of the 2.0 milliliters of 1.2N hydrochloric 
acid, and the assay should be completed within 1 hour after the sample 
and standard are first put into solution.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
[39 FR 19040, May 30, 1974, as amended at 45 FR 16472, Mar. 14, 1980; 50 
FR 19919, May 13, 1985]



Sec. 442.128  Cephalexin hydrochloride monohydrate tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephalexin hydrochloride monohydrate 
tablets are composed of cephalexin hydrochloride monohydrate and one or 
more suitable and harmless lubricants, colorings and coating substances. 
Each tablet contains cephalexin hydrochloride monohydrate equivalent to 
250 milligrams, 333 milligrams or 500 milligrams of cephalexin. Its 
cephalexin content is satisfactory if it is not less than 90 percent and 
not more than 120 percent of the number of milligrams of cephalexin that 
it is represented to contain. Its moisture content is not more than 8.0 
percent. The tablets pass the dissolution test. It passes the identity 
test. The cephalexin hydrochloride monohydrate used conforms to the 
standards prescribed by Sec. 442.28(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cephalexin hydrochloride monohydrate used in making the 
batch for cephalexin potency, moisture, pH, identity, and crystallinity.
    (B) The batch for cephalexin content, moisture, dissolution, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research.
    (A) The cephalexin hydrochloride monohydrate used in making the 
batch: 10 packages, each containing approximately 500 milligrams.
    (B) The batch: A minimum of 36 tablets.

[[Page 664]]

    (b) Tests and methods of assay--(1) Cephalexin content. Proceed as 
directed in Sec. 442.140c(b)(1)(ii), except that ``cephalexin'' is 
substituted at each occurrence of ``cephradine''.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q (the amount of cephalexin dissolved) is not less 
than 75 percent at 45 minutes.
    (4) Identity. Proceed as directed in Sec. 436.367 of this chapter.
[54 FR 48860, Nov. 28, 1989]



Sec. 442.140a  Cephradine for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephradine for oral suspension is 
cephradine with one or more suitable and harmless diluents, buffer 
substances, colorings, and flavorings. When reconstituted as directed in 
the labeling, each milliliter contains 25 milligrams or 50 milligrams of 
cephradine. Its potency is satisfactory if it is not less than 90 
percent and not more than 125 percent of the number of milligrams of 
cephradine that it is represented to contain. Its moisture content is 
not more than 1.5 percent. When reconstituted as directed in the 
labeling, its pH is not less than 3.5 and not more than 6.0. The 
cephradine used conforms to the standards prescribed by 
Sec. 442.40(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cephradine used in making the batch for potency, moisture, 
pH, cephalexin content, identity, and crystallinity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The cephradine used in making the batch: 10 packages, each 
containing 500 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute the sample as directed in the labeling. Transfer an 
accurately measured representative portion of the suspension into an 
appropriate-sized volumetric flask and dilute to volume with 1 percent 
potassium phosphate buffer, pH 6.0 (solution 1), to obtain a stock 
solution of convenient concentration. Further dilute an aliquot of the 
stock solution with solution 1 to the reference concentration of 10.0 
micrograms of cephradine per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, preparing the sample as follows: 
Reconstitute the sample as directed in the labeling. Transfer an 
accurately measured representative portion to a volumetric flask and 
bring to volume with distilled water. Further dilute an aliquot of this 
solution with distilled water to 1 milligram of cephradine per 
milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
[40 FR 26272, June 23, 1975, as amended at 45 FR 16476, Mar. 14, 1980; 
50 FR 19919, May 13, 1985]



Sec. 442.140b  Cephradine capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephradine capsules are composed of 
cephradine and one or more suitable and harmless lubricants and diluents 
enclosed in a gelatin capsule. Each capsule contains 250 milligrams or 
500 milligrams of cephradine. Its potency is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of cephradine that it is represented to contain. Its loss on 
drying is not more than 7.0 percent. The cephradine used conforms to the 
standards prescribed by Sec. 442.40(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 665]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cephradine used in making the batch for potency, moisture, 
pH, cephalexin content, identity, and crystallinity.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:
    (a) The cephradine used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) the batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 1 percent potassium phosphate buffer, 
pH 6.0 (solution 1), to give a stock solution of convenient 
concentration. Blend for 3 to 5 minutes. Remove an aliquot and further 
dilute with solution 1 to the reference concentration of 10.0 micrograms 
of cephradine per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii) of this chapter, preparing the sample as follows: 
Blend a representative number of capsules in a high-speed glass blender 
jar with sufficient distilled water to give a stock solution of 
convenient concentration. Further dilute an aliquot of this solution 
with distilled water to 1 milligrams of cephradine per milliliter 
(estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[40 FR 26272, June 23, 1975, as amended at 50 FR 19919, May 13, 1985]



Sec. 442.140c  Cephradine tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephradine tablets are composed of 
cephradine and one or more suitable and harmless diluents, binders, 
lubricants, and colorings. Each tablet contains 1,000 milligrams of 
cephradine. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
cephradine that it is represented to contain. Its moisture content is 
not more than 6.0 percent. It disintegrates within 30 minutes. The 
cephradine used conforms to the standards prescribed by 
Sec. 442.40(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cephradine used in making the batch for potency, moisture, 
pH, cephalexin content, identity, and crystallinity.
    (b) The batch for potency, moisture, and disintegration time.
    (ii) Samples required:
    (a) The cephradine used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the hydroxylamine 
colorimetric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to give a stock solution of convenient concentration. 
Blend for 3 to 5 minutes. Remove an aliquot and further dilute with 
solution 1 to the reference concentration of 10.0 micrograms of 
cephradine per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii), except prepare the sample and calculate the 
cephradine content as follows:
    (a) Preparation of sample. Blend a representative number of tablets 
in a high-speed glass blender jar with sufficient distilled water to 
give a stock solution of convenient concentration.

[[Page 666]]

Further dilute an aliquot of this solution with distilled water to 1 
milligram of cephradine per milliliter (estimated).
    (b) Calculations. Calculate the cephradine content as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.056
    
where:

    Au=Absorbance of sample solution;
    Ps=Potency of working standard in micrograms per 
milligram;
    d=Dilution factor for sample;
    As=Absorbance of working standard solution;
    n=Number of tablets in the sample assayed.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure described in paragraph (e)(1) of that 
section.
[45 FR 22919, Apr. 4, 1980, as amended at 50 FR 19919, May 13, 1985]



Sec. 442.141  Cephradine dihydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephradine dihydrate capsules are 
composed of cephradine dihydrate and one or more suitable and harmless 
lubricants and diluents enclosed in a gelatin capsule. Each capsule 
contains 250 milligrams or 500 milligrams of cephradine. Its potency is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of cephradine that it is represented 
to contain. Its moisture content is not more than 11.0 percent. It 
passes the dissolution test if the quantity Q is 85 percent at 60 
minutes. The cephradine dihydrate used conforms to the standards 
prescribed by Sec. 442.41(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cephradine dihydrate used in making the batch for potency, 
moisture, pH, cephalexin content, identity, and crystallinity.
    (b) The batch for potency, moisture, and dissolution.
    (ii) Samples required:
    (a) The cephradine dihydrate used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch: A minimum of 100 capsules.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the hydroxylamine 
colorimetric assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 1 percent potassium phosphate buffer, 
pH 6.0 (solution 1), to obtain a stock solution of convenient 
concentration. Blend for 3 to 5 minutes. Further dilute and aliquot of 
the stock solution with solution 1 to the reference concentration of 
10.0 micrograms of cephradine per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii), except prepare the sample solution and calculate 
the cephradine content as follows:
    (a) Preparation of sample solution. Blend a representative number of 
capsules in a high-speed glass blender jar with sufficient distilled 
water to obtain a stock solution of convenient concentration. Further 
dilute an aliquot of this solution with distilled water to a 
concentration of 1 milligram of cephradine per milliliter (estimated).
    (b) Calculations. Calculate the cephradine content as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.057
    
where:
    Au=Absorbance of sample solution;
    Ps=Potency of working standard in micrograms per 
milligram;
    d=Dilution factor for sample;
    As=Absorbance of working standard solution;
    n=Number of capsules in the sample assayed.


[[Page 667]]


    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Dissolution. Proceed as directed in Sec. 436.541 of this 
chapter, except:
    (i) A distance of 2.5plus-minus0.2 centimeters should be 
maintained between the lower edge of the stirring blade and the lowest 
inner surface of the vessel during test rather than 
4.5plus-minus0.5 centimeters as specified in paragraph (a) of 
that section; and
    (ii) In lieu of paragraph (d) of that section, use the 
interpretation described in the United States Pharmacopeia XX 
dissolution test.
[47 FR 11857, Mar. 19, 1982, as amended at 50 FR 19919, May 13, 1985]



Sec. 442.154  Cefpodoxime proxetil oral dosage forms.



Sec. 442.154a  Cefpodoxime proxetil tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefpodoxime proxetil tablets are composed 
of cefpodoxime proxetil and one or more suitable and harmless diluents, 
binders, lubricants, colorings, and coating substances. Each tablet 
contains cefpodoxime proxetil equivalent to either 100 milligrams or 200 
milligrams of cefpodoxime. Its cefpodoxime proxetil content is 
satisfactory if it is not less than 90 percent and not more than 110 
percent of the number of milligrams of cefpodoxime that it is 
represented to contain. Its loss on drying is not more than 5 percent. 
It passes the dissolution test. It passes the identity test. The 
cefpodoxime proxetil used conforms to the standards prescribed by 
Sec. 442.54(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefpodoxime proxetil used in making the batch for potency, 
isomer ratio, moisture, and identity.
    (B) The batch for content, loss on drying, dissolution, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The cefpodoxime proxetil used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (B) The batch: A minimum of 100 tablets.
    (b) Tests and methods of assay--(1) Cefpodoxime content. Proceed as 
directed in Sec. 442.54(b)(1), preparing the sample solution and 
calculating the cefpodoxime content as follows:
    (i) Preparation of sample solution. Obtain the average tablet weight 
of at least 20 tablets. Grind the tablets using a mortar and pestle. 
Weigh approximately 660 milligrams into a suitable container. Add 30 
milliliters of internal standard solution. Shake for 30 minutes using a 
horizontal platform shaker or equivalent. Centrifuge for about 10 
minutes at 3,000 revolutions per minute until the particulate matter has 
settled. Withdraw a 1 milliliter aliquot of the supernatant and dilute 
with 9 milliliters of dilution solvent. The sample solutions are stable 
for at least 48 hours. Refrigeration is not recommended.
    (ii) Calculations. Calculate the cefpodoxime content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.384
    
where:
Rsam = Ratio of cefpodoxime proxetil peaks area (sum of both 
          epimers) to the internal standard peak area in the sample 
          preparation;
Rstd = Ratio of cefpodoxime proxetil peaks area (sum of both 
          epimers) to the internal standard peak area in the standard 
          preparation;
Wstd = Weight of cefpodoxime proxetil reference standard, in 
          milligrams;
Wsam = Weight of sample, in milligrams;

[[Page 668]]

F1 = Volume of internal standard used in the sample 
          preparation, in milliliters;
F2 = 0.766; The ratio of molecular weight for free-acid 
          cefpodoxime over the molecular weight of cefpodoxime proxetil 
          (427.46/557.61);
F3 = Volume of internal standard used in the standard 
          preparation, in milliliters;
F4 = Average tablet weight, i.e., weight of tablets used in 
          sample preparation divided by the number of tablets; and
P = Purity of the cefpodoxime proxetil reference standard, expressed as 
          a decimal.

    (2) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter, except dry the sample at a temperature of 80 deg. C and a 
pressure of 5 millimeters of mercury or less for 16 hours.
    (3) Dissolution test. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q (the amount of cefpodoxime activity dissolved) 
is 70 percent within 30 minutes.
    (4) Identity. Using the high-performance liquid chromatographic 
procedure described in paragraph (b)(1) of this section, the retention 
times for the peaks of the active ingredients must be within 2 percent 
of the retention times for the peaks of the corresponding reference 
standards.
[60 FR 58232, Nov. 27, 1995]



Sec. 442.154b  Cefpodoxime proxetil granules for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefpodoxime proxetil granules for oral 
suspension is cefpodoxime proxetil and one or more suitable and harmless 
preservatives, sweeteners, suspending agents, buffers, and flavorings. 
When constituted as directed in the labeling, each milliliter contains 
the equivalent of either 10 or 20 milligrams cefpodoxime activity. Its 
cefpodoxime proxetil content is satisfactory if it is not less than 90 
percent and not more than 110 percent of the number of milligrams of 
cefpodoxime that it is represented to contain. Its loss on drying is not 
more than 0.5 percent. When constituted as described in the labeling, 
the pH of the suspension is not less than 4 and not more than 5.5. It 
passes the identity test. The cefpodoxime proxetil used conforms to the 
standards prescribed by Sec. 442.54(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefpodoxime proxetil used in making the batch for potency, 
isomer ratio, moisture, and identity.
    (B) The batch for content, loss on drying, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The cefpodoxime proxetil used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (B) The batch: A minimum of 10 intermediate containers.
    (b) Tests and methods of assay--(1) Cefpodoxime content. Proceed as 
directed in Sec. 442.54(b)(1), preparing the sample solution and 
calculating the cefpodoxime content as follows:
    (i) Preparation of sample solution. Reconstitute as directed in the 
labeling. Immediately before sampling the suspension, shake vigorously 
for several seconds. Into a suitable container, accurately weigh out 6 
grams of the 50 milligrams per 5 milliliters suspension, or 3 grams of 
the 100 milligrams per 5 milliliters suspension. Add 5 milliliters of 
internal standard solution and 25 milliliters of dilution solvent. Shake 
for 30 minutes using a horizontal platform shaker or equivalent. 
Centrifuge for about 10 minutes at 3,000 revolutions per minute until 
the particulate matter has settled. Withdraw a 1 milliliter aliquot of 
the supernatant and dilute with 1 milliliter of dilution solvent. The 
sample solutions are stable for at least 48 hours. Refrigeration is not 
recommended.
    (ii) Calculations. Calculate the cefpodoxime content as follows:

[[Page 669]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.385


where:
Rsam = Ratio of cefpodoxime proxetil peaks area (sum of both 
          epimers) to the internal standard peak area in the sample 
          preparation;
Rstd = Ratio of cefpodoxime proxetil peaks area (sum of both 
          epimers) to the internal standard peak area in the standard 
          preparation;
Wstd = Weight of cefpodoxime proxetil reference standard, in 
          milligrams;
Wsam = Weight of sample, in grams;
F1 = Volume of internal standard used in the sample; 
          preparation, in milliliters;
F2 = 0.766; The ratio of molecular weight for free-acid 
          cefpodoxime over the molecular weight of cefpodoxime proxetil 
          (427.46/557.61);
F3 = Volume of internal standard used in the standard 
          preparation, in milliliters;
F4 = 0.2; Factor to convert to 5 milliliters;
F5 = Specific gravity of suspension for milligram per 5 
          milliliter calculated on the air-free basis (specific gravity 
          is determined on a sample of suspension that has been shaken 
          gently on a platform shaker under vacuum for 2 hours); and
P = Purity of the cefpodoxime proxetil reference standard, expressed as 
          a decimal.

    (2) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter, except dry the sample at a temperature of 80 deg. C and a 
pressure of 5 millimeters of mercury or less for 16 hours.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug constituted as directed in the labeling.
    (4) Identity. Using the high-performance liquid chromatographic 
procedure described in paragraph (b)(1) of this section, the retention 
times for the peaks of the active ingredients must be within 2 percent 
of the retention times for the peaks of the corresponding reference 
standards.
[60 FR 58233, Nov. 27, 1995]



Sec. 442.180  Cefprozil oral dosage forms.



Sec. 442.180a  Cefprozil tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefprozil tablets are composed of 
cefprozil and one or more suitable and harmless diluents, binders, 
lubricants, colorings, and coating substances. Each tablet contains 
cefprozil equivalent to either 250 milligrams or 500 milligrams of 
anhydrous cefprozil. The cefprozil content of the tablets is 
satisfactory if it is not less than 90 percent nor more than 120 percent 
of the number of milligrams of anhydrous cefprozil that it is 
represented to contain. The moisture content of the tablets is not more 
than 7 percent. The tablets pass the dissolution test. The tablets pass 
the identity tests. The cefprozil used conforms to the standards 
prescribed by Sec. 442.80(a)(1) of this part.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefprozil used in making the batch for potency, E-isomer 
ratio, moisture, pH, crystallinity, and identity.
    (B) The batch for content, moisture, dissolution, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The cefprozil used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch: A minimum of 100 tablets.
    (b) Tests and methods of assay--(1) Cefprozil content. Proceed as 
directed in Sec. 442.80(b)(1) of this part, preparing the sample 
solution and calculating the cefprozil content as follows:
    (i) Preparation of sample solution. Place one or a known number of 
intact tablets into a 250-milliliter volumetric flask containing about 
180 milliliters of distilled water. Allow the tablet(s) to disintegrate 
as aided by swirling and brief ultrasonication. Dilute the contents to 
volume with distilled water and mix thoroughly. Transfer an aliquot of 
this solution to a volumetric

[[Page 670]]

flask of suitable size to obtain a solution containing 0.3 milligram per 
milliliter of cefprozil (estimated) when diluted to volume with water. 
Filter through a 0.45 micron filter prior to injection into the 
chromatographic system.
    (ii) Calculations. Calculate the cefprozil content as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.058
    
where:
AU=Area of the cefprozil (Z) or cefprozil (E) response in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
AS=Area of the cefprozil (Z) or cefprozil (E) response in the 
          chromatogram of the cefprozil (Z) or the cefprozil (E) working 
          standard;
PS=Cefprozil (Z) or cefprozil (E) activity in the cefprozil 
          (Z) or the cefprozil (E) working standard solution in 
          micrograms per milliliter;
d = Dilution factor of the sample; and
n = Number of tablets taken in the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Dissolution test. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q (the amount of cefprozil activity dissolved) is 
75 percent at 45 minutes.
    (4) Identity--(i) High performance liquid chromatography. Using the 
high performance liquid chromatographic procedure described in paragraph 
(b)(1) of this section, the retention times for the responses of the 
active ingredients must be within 2 percent of the retention times for 
the responses of the corresponding reference standards.
    (ii) Thin layer chromatography. Proceed as directed in Sec. 436.368 
of this chapter.
[58 FR 26661, May 4, 1993]



Sec. 442.180b  Cefprozil for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefprozil for oral suspension is 
cefprozil with one or more suitable and harmless preservatives, 
sweeteners, suspending agents, buffers, and flavorings. The cefprozil 
content of the oral suspension is satisfactory if it is not less than 90 
percent nor more than 120 percent of the number of milligrams of 
anhydrous cefprozil that it is represented to contain. When constituted 
as directed in the labeling, each milliliter contains the equivalent of 
either 25 or 50 milligrams anhydrous cefprozil activity. Its moisture 
content is not more than 3 percent. When constituted as described in the 
labeling, the pH of the suspension is not less than 4.0 nor more than 
6.0. It passes the identity tests. The cefprozil used conforms to the 
standards prescribed by Sec. 442.80(a)(1) of this part.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefprozil used in making the batch for potency, E-isomer 
ratio, moisture, pH, crystallinity, and identity.
    (B) The batch for content, moisture, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The cefprozil used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch: A minimum of 10 intermediate containers.
    (b) Tests and methods of assay--(1) Cefprozil content. Proceed as 
directed in Sec. 442.80(b)(1), preparing the sample solution and 
calculating the cefprozil content as follows:
    (i) Preparation of sample solution. Constitute as directed in the 
labeling. Transfer a portion of the suspension containing 250 milligrams 
(estimated) of cefprozil into a 250-milliliter volumetric flask using a 
glass syringe and a 13-gauge needle. Dilute to volume with water, 
ultrasonicate briefly to dissolve and mix well. Transfer a 15-milliliter 
aliquot of this solution to a 50-milliliter volumetric flask and dilute 
to volume with water to obtain a solution containing 0.3 milligram per

[[Page 671]]

milliliter of cefprozil (estimated). Filter through a 0.45 micron filter 
prior to injection into the chromatographic system.
    (ii) Calculations. Calculate the cefprozil content as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.059
    
where:
AU=Area of the cefprozil (Z) or cefprozil (E) response in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
AS=Area of the cefprozil (Z) or cefprozil (E) response in the 
          chromatogram of the cefprozil (Z) or the cefprozil (E) working 
          standard;
PS=Cefprozil (Z) or cefprozil (E) activity in the cefprozil 
          (Z) or the cefprozil (E) working standard solution in 
          micrograms per milliliter;
d = Dilution factor of the sample; and
V = Volume of sample taken in milliliters.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug constituted as directed in the labeling.
    (4) Identity--(i) High Performance liquid chromatography. Using the 
high-performance liquid chromatographic procedure described in paragraph 
(b)(1) of this section, the retention times for the responses of the 
active ingredients must be within 2 percent of the retention times for 
the responses of the corresponding reference standards.
    (ii) Thin layer chromatography. Proceed as directed in Sec. 436.368 
of this chapter.
[58 FR 26661, May 4, 1993]



                   Subpart C--Injectable Dosage Forms



Sec. 442.208  Cefamandole nafate for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefamandole nafate for injection is a dry 
mixture of cefamandole nafate and one or more suitable and harmless 
buffering agents. The cefamandole nafate may be isolated in the 
manufacture of cefamandole nafate for injection. Its cefamandole content 
is satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of milligrams of cefamandole that it is 
represented to contain. It is sterile. It is nonpyrogenic. Its moisture 
content is not more than 3.0 percent. Its pH is not less than 6.0 and 
not more than 8.0. If isolated, the cefamandole nafate used conforms to 
the standards prescribed by Sec. 442.8a(a)(1). If the cefamandole nafate 
is not isolated, the potency of the dry mixture is not less than 810 
micrograms and not more than 1,000 micrograms of cefamandole per 
milligram on an anhydrous basis when corrected for sodium carbonate; and 
the dry mixture gives a positive identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) If isolated, the cefamandole nafate used in making the batch for 
cefamandole content, moisture, pH, and identity.
    (b) The batch for cefamandole content, sterility, pyrogens, 
moisture, and pH. In addition, if the cefamandole nafate is not 
isolated, results of tests and assays on the dry mixture for potency and 
identity.
    (ii) Samples required:
    (a) For all tests except sterility: A minimum of 10 immediate 
containers, unless the cefamandole nafate is not isolated, a minimum of 
15 immediate containers.
    (b) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Cefamandole content. Proceed as 
directed in Sec. 436.324 of this chapter, preparing the sample solution 
and calculating the cefamandole content as follows:
    (i) Sample preparation. Reconstitute the sample as directed in the 
labeling.

[[Page 672]]

If it is represented as a single dose container, remove all of the 
withdrawable contents with a suitable hypodermic needle and syringe. If 
the labeling specifies the amount of cefamandole content in a given 
volume of the resultant preparation, remove an accurately measured 
representative portion from each container. Further dilute an aliquot of 
this solution with distilled water to obtain a concentration of 2.0 
milligrams of cefamandole per milliliter (estimated). Transfer 5 
milliliters of this solution to a 50-milliliter volumetric flask, add 30 
milliliters of pH 2.3 buffer, dilute to volume with distilled water, and 
mix. In addition, if the cefamandole nafate is not isolated, prepare the 
sample solution as described in Sec. 436.324(d) of this chapter. 
Determine the sodium carbonate content as follows: Dissolve an 
accurately weighed portion of the dry mixture, approximately 1.0 gram, 
with approximately 100 milliliters of distilled water. Titrate with 0.2N 
hydrochloric acid. Determine the end-point potentiometrically to the 
first equivalent using a glass calomel combination electrode. Each 
milliliter of 0.2N hydrochloric acid is equivalent to 21.2 milligrams of 
sodium carbonate.
    (ii) Calculations--(a) Calculate the cefamandole content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.199
    
    where:
A=The peak height of the sample;
B=The peak height of the working standard; and
f=The dilution factor of the sample.

    (b) If the cefamandole nafate is not isolated in the manufacture of 
cefamandole nafate for injection, calculate the micrograms of 
cefamandole per milligram of sample as described in Sec. 436.324(f) of 
this chapter. The micrograms per milligram of cefamandole is corrected 
for sodium carbonate content and moisture content.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cefamandole per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter, except 
determine the pH 30 minutes after preparation of the sample solution.
    (7) Identity. Proceed as directed in Sec. 436.323 of this chapter.
[44 FR 20665, Apr. 6, 1979, as amended at 47 FR 42099, Sept. 24, 1982; 
50 FR 19919, May 13, 1985]



Sec. 442.209  Cefamandole sodium for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefamandole sodium for injection is a dry 
mixture of cefamandole sodium and one or more suitable and harmless 
buffering agents. Its cefamandole content is satisfactory if it is not 
less than 90 percent and not more than 115 percent of the number of 
milligrams of cefamandole that it is represented to contain. It is 
sterile. It is nonpyrogenic. Its moisture content is not more than 3.0 
percent. Its pH is not less than 6.0 and not more than 8.5. The 
cefamandole sodium used conforms to the standards prescribed by 
Sec. 442.9a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 673]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cefamandole sodium used in making the batch for cefamandole 
content, moisture, pH, and identity.
    (b) The batch for cefamandole content, sterility, pyrogens, 
moisture, and pH.
    (ii) Samples required:
    (a) The cefamandole sodium used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Cefamandole content. Proceed as 
directed in Sec. 436.324 of this chapter, preparing the sample solution 
and calculating the cefamandole content as follows:
    (i) Sample solution. Reconstitute the sample as directed in the 
labeling. If it is represented as a single-dose container, remove all 
the withdrawable contents with a suitable hypodermic needle and syringe. 
If the labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Further dilute an aliquot of this solution 
with distilled water to obtain a concentration of 2.0 milligrams of 
cefamandole per milliliter (estimated). Transfer 5 milliliters of this 
solution to a 50-milliliter volumetric flask, add 30 milliliters of pH 
2.3 buffer, dilute to volume with distilled water, and mix.
    (ii) Calculations. Calculate the cefamandole content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.200
    
where:
    A=The peak height of the sample;
    B=The peak height of the working standard;
    f=The dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cefamandole per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
[47 FR 20756, May 14, 1982, as amended at 50 FR 19919, May 13, 1985]



Sec. 442.211  Cefazolin sodium injectable dosage forms.



Sec. 442.211a  Sterile cefazolin sodium.

    The requirements for certification and the tests and methods of 
assay for sterile cefazolin sodium packaged for dispensing are described 
in Sec. 442.11a, except for the following additional requirements if it 
is packaged with lidocaine hydrochloride injection 0.5 percent U.S.P.:
    (a) The pH, when reconstituted and diluted to 100 milligrams per 
milliliter with lidocaine hydrochloride injection 0.5 percent U.S.P., is 
not less than 5.5 and not more than 7.0.
    (b) In addition to the information required by Sec. 442.11a 
(a)(3)(i), the following shall be submitted:

[[Page 674]]

    (1) The pH on the batch reconstituted with lidocaine hydrochloride 
injection 0.5 percent U.S.P.; and
    (2) Results of tests and assays on the lidocaine hydrochloride 
injection 0.5 percent to show conformance with U.S.P. requirements.
[42 FR 18059, Apr. 5, 1977. Redesignated at 48 FR 33479, July 22, 1983]



Sec. 442.211b  Cefazolin sodium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefazolin sodium injection is a frozen 
aqueous solution of cefazolin sodium in an isoosmotic diluent. Each 
milliliter contains cefazolin sodium equivalent to either 10 milligrams 
or 20 milligrams of cefazolin per milliliter. Its cefazolin content is 
satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of milligrams of cefazolin that it is represented 
to contain. It is sterile. It is nonpyrogenic. Its pH is not less than 
4.5 and not more than 7.0. It passes the identity test. The cefazolin 
used conforms to the standards prescribed by Sec. 442.10(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Requests of tests and assays on:
    (a) The cefazolin used in making the batch for cefazolin content, 
moisture, heavy metals, and identity.
    (b) The batch for cefazolin content, sterility, pyrogens, pH, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The cefazolin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Cefazolin content. Proceed as 
directed in Sec. 436.342 of this chapter, preparing the sample solution 
and calculating the cefazolin content as follows:
    (i) Preparation of sample solution. Using a suitable hypodermic 
needle and syringe, transfer an accurately measured representative 
portion from each container, equivalent to 40 milligrams of cefazolin, 
to a 100-milliliter volumetric flask. Dilute to volume with buffer 
solution, pH 7.0, and mix. Transfer 10.0 milliliters of this solution to 
a 200-milliliter volumetric flask, add 5.0 milliliters of internal 
standard solution, dilute to volume with buffer solution, pH 7.0, and 
mix
    (ii) Calculation. Calculate the milligrams of cefazolin per 
milliliter of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.201

where:
Ru=Area of the cefazolin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard) /Area of internal standard peak;
Rs=Area of cefazolin peak in the chromatogram of the 
          cefazolin working standard /Area of internal standard peak;
Ps=Cefazolin activity in the cefazolin working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
except inject a sufficient volume of the undiluted solution to deliver 
50 milligrams of cefazolin per kilogram.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (5) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the cefazolin working standard.
[48 FR 33479, July 22, 1983; 48 FR 40516, Sept. 8, 1983; 49 FR 48184, 
Dec. 11, 1984, as amended at 55 FR 11583, Mar. 29, 1990]

[[Page 675]]



Sec. 442.212  Cefoperazone injectable dosage forms.



Sec. 442.212a  Sterile cefoperazone sodium.

    The requirements for certification and the tests and methods of 
assay for sterile cefoperazone sodium packaged for dispensing are 
described in Sec. 442.12a.
[48 FR 790, Jan. 7, 1983. Redesignated at 51 FR 36688, Oct. 15, 1986]



Sec. 442.212b  Cefoperazone sodium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefoperazone sodium injection is a frozen 
aqueous iso-osmotic solution of cefoperazone sodium which may contain 
one or more suitable and harmless buffer substances in a diluent. Each 
milliliter contains cefoperazone sodium equivalent to either 20 
milligrams or 40 milligrams of cefoperazone per milliliter. Its 
cefoperazone content is satisfactory if it is not less than 90 percent 
and not more than 120 percent of the number of milligrams of 
cefoperazone that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its pH is not less than 4.5 and not more than 6.5. It 
passes the identity test. The cefoperazone sodium used conforms to the 
standards prescribed by Sec. 442.12(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cefoperazone sodium used in making the batch for potency, 
moisture, pH, and identity.
    (b) The batch for potency, sterility, pyrogens, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The cefoperazone sodium used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Potency. Proceed as directed in Sec. 436.338 of this chapter, 
preparing the sample solution and calculating the cefoperazone content 
as follows:
    (i) Sample solution. Using a suitable hypodermic needle and syringe, 
remove an accurately measured representative portion from each container 
and dilute with mobile phase to obtain a solution containing 160 
micrograms per milliliter (estimated).
    (ii) Calculations. Calculate the milligrams of cefoperazone per 
milliliter of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.202

where:
Au=Area of the cefoperazone peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefoperazone peak in the chromatogram of the 
          cefoperazone working standard;
Ps=Cefoperazone activity in the cefoperazone working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
except inject a sufficient volume of the undiluted solution to deliver 
10 milligrams of cefoperazone per kilogram.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (5) Identity. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the cefoperazone working standard.
[51 FR 36688, Oct. 15, 1986, as amended at 54 FR 47352, Nov. 14, 1989; 
55 FR 11583, Mar. 29, 1990]

[[Page 676]]



Sec. 442.213  Cefotaxime injectable dosage forms.



Sec. 442.213a  Sterile cefotaxime sodium.

    The requirements for certification and the tests and methods of 
assay for sterile cefotaxime sodium packaged for dispensing are 
described in Sec. 442.13a.
[46 FR 25607, May 8, 1981. Redesignated at 50 FR 45109, Oct. 30, 1985]



Sec. 442.213b  Cefotaxime sodium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefotaxime sodium injection is a frozen 
aqueous solution of cefotaxime sodium with one or more suitable and 
harmless buffer substances in an isosmotic diluent. Each milliliter 
contains cefotaxime sodium equivalent to either 20 milligrams or 40 
milligrams of cefotaxime per milliliter. Its cefotaxime content is 
satisfactory if it is not less than 90 percent and not more than 110 
percent of the number of milligrams of cefotaxime that it is represented 
to contain. It is sterile. It is nonpyrogenic. Its pH is not less than 
5.0 and not more than 7.5. It passes the identity test. The cefotaxime 
sodium used conforms to the standards prescribed by Sec. 442.13(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cefotaxime sodium used in making the batch for potency, 
moisture, pH, and identity.
    (b) The batch for potency, sterility, pyrogens, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The cefotaxime sodium used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Potency. Use either of the following methods; however, the 
results obtained from the hydroxylamine colorimetric assay shall be 
conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Using a suitable hypodermic needle and syringe, remove an accurately 
measured representative portion from each container and dilute with 
sufficient 1 percent potassium phosphate buffer, pH 6.0 (solution 1), to 
give a stock solution of convenient concentration. Further dilute an 
aliquot of the stock solution with solution 1 to the reference 
concentration of 2.0 micrograms of cefotaxime per milliliter 
(estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter, preparing the sample as follows: Using a 
suitable hypodermic needle and syringe, remove an accurately measured 
representative portion from each container and dilute with distilled 
water to give a stock solution of convenient concentration. Further 
dilute with distilled water to the prescribed concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
except inject a sufficient volume of the undiluted solution to deliver 
50 milligrams of cefotaxime per kilogram.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (5) Identity. Proceed as directed in Sec. 436.323 of this chapter, 
except prepare spotting solutions as follows: Prepare solutions of the 
sample and working standard, each containing 1 milligram of cefotaxime 
per milliliter in distilled water.
[50 FR 45109, Oct. 30, 1985; 50 FR 48078, Nov. 21, 1985, as amended at 
55 FR 11583, Mar. 29, 1990]

[[Page 677]]



Sec. 442.214  Cefoxitin injectable dosage forms.



Sec. 442.214a  Sterile cefoxitin sodium.

    The requirements for certification and the tests and methods of 
assay for sterile cefoxitin packaged for dispensing are described in 
Sec. 442.14a.
[44 FR 10376, Feb. 20, 1979. Redesignated at 49 FR 47827, Dec. 7, 1984]



Sec. 442.214b  Cefoxitin sodium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefoxitin sodium injection is a frozen 
aqueous solution of cefoxitin sodium with one or more suitable and 
harmless buffer substances in an isotonic diluent. Each milliliter 
contains cefoxitin sodium equivalent to either 20 or 40 milligrams of 
cefoxitin. Its cefoxitin content is satisfactory if it contains not less 
than 90 percent and not more than 120 percent of the number of 
milligrams of cefoxitin that it is represented to contain. It is 
sterile. It is nonpyrogenic. Its pH is not less than 4.5 and not more 
than 8.0. It passes the identity test. The cefoxitin sodium used 
conforms to the standards prescribed by Sec. 442.14(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cefoxitin sodium used in making the batch for cefoxitin 
content, moisture, pH, identity, and crystallinity.
    (b) The batch for cefoxitin content, sterility, pyrogens, pH, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The cefoxitin sodium used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Cefoxitin content. Proceed as directed in Sec. 436.347 of this 
chapter, preparing the working standard and sample solutions and 
calculating the cefoxitin content as follows:
    (i) Working standard solution. Dissolve an accurately weighed 
portion of the cefoxitin working standard with water to obtain a 
solution containing 200 micrograms of cefoxitin per milliliter.
    (ii) Sample solution. Using a suitable hypodermic needle and 
syringe, remove an accurately measured representative portion from each 
container and dilute with sufficient water to obtain a solution 
containing 200 micrograms of cefoxitin per milliliter (estimated).
    (iii) Calculations. Calculate the milligrams of cefoxitin per 
milliliter of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.203

where:
Au=Area of the cefoxitin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefoxitin peak in the chromatogram of the 
          cefoxitin working standard;
Ps=Cefoxitin activity in the cefoxitin working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
except inject a sufficient volume of the undiluted solution to deliver 
50 milligrams of cefoxitin per kilogram.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (5) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the cefoxitin working standard.
[49 FR 47827, Dec. 7, 1984, as amended at 55 FR 11583, Mar. 29, 1990]

[[Page 678]]



Sec. 442.216  Ceftazidime injectable dosage forms.



Sec. 442.216a  Ceftazidime pentahydrate for injection.

    (a) Requirements of certification--(1) Standards of identity, 
strength, quality, and purity. Ceftazidime pentahydrate for injection is 
a dry mixture of ceftazidime pentahydrate and sodium carbonate or L-
arginine. Its ceftazidime potency is satisfactory if each milligram of 
ceftazidime pentahydrate for injection contains not less than 900 
micrograms and not more than 1,050 micrograms of cefazidime activity 
when corrected for both loss on drying and its sodium carbonate or L-
arginine content, as appropriate for the formulation. Its ceftazidime 
content is satisfactory if it is not less than 90 percent and not more 
than 120 percent of the number of milligrams of ceftazidime that it is 
represented to contain. It is sterile. It is nonpyrogenic. Its loss on 
drying is not more than 12.5 percent if it contains L-arginine and not 
more than 13.5 percent if it contains sodium carbonate. The pH of its 
aqueous solution is not less than 5.0 and not more than 7.5. Its 
pyridine content, if it contains sodium carbonate, is not more than 0.4 
percent, except that for the issuance of a certificate for each batch of 
the sodium carbonate formulation, the pyridine content is not more than 
0.12 percent. Its pyridine content, if it contains L-arginine, is not 
more than 0.3 percent, except that for the issuance of a certificate, 
the pyridine content of the L- arginine formulation is not more than 
0.10 percent. The ceftazidime pentahydrate conforms to the standard 
prescribed by Sec. 442.16a(a)(1).
    (2) Labeling. In addition to the requirements of Sec. 432.5 of this 
chapter, each package of the L-arginine formulation shall bear on its 
outside wrapper or container and on the immediate container the 
statement ``For Patients 12 years and Older''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The ceftazidime pentahydrate used in making the batch for 
potency, loss on drying, pH, crystallinity, identity, and high molecular 
weight polymer content.
    (b) The batch for ceftazidime potency, ceftazidime content, 
sterility, pyrogens, loss on drying, pH, and pyridine content.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The ceftazidime pentahydrate used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Ceftazidime potency and content. 
Determine both micrograms of ceftazidime per milligram of sample and 
milligrams of ceftazidime per container. Proceed as directed in 
Sec. 442.16a(b)(1), preparing the sample solutions and calculating the 
potency and content as follows:
    (i) Preparation of sample solutions. Use separate containers for 
preparation of each sample solution as described in paragraphs (b)(1)(i) 
(a) and (b) of this section.
    (a) Ceftazidime potency (micrograms of ceftazidime per milligram). 
Accurately weigh and dissolve approximately 350 milligrams of 
ceftazidime sample in distilled water and dilute to volume in a 250-
milliliter volumetric flask to obtain a stock solution containing 
approximately 1,000 micrograms of ceftazidime per milliliter. Mix well. 
Immediately prior to chromatography, further dilute 5 milliliters of 
stock solution to 50 milliliters with water to obtain a solution 
containing 100 micrograms of ceftazidime activity per milliliter 
(estimated).
    (b) Ceftazidime content (milligrams of ceftazidime per vial). 
Reconstitute the sample as directed in the labeling. Then, using a 
suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is represented as a single-dose container; or, if the 
labeling specifies the amount of

[[Page 679]]

potency in a given volume of the resultant preparation, remove an 
accurately measured representative portion from each container. Further 
dilute an aliquot of this solution with distilled water to obtain a 
concentration of 1.0 milligram per milliliter (estimated). Immediately 
prior to chromatography, dilute 5.0 milliliters of the sample solution 
to 50 milliliters with water.
    (ii) Calculations--(a) Ceftazidime potency (micrograms per 
milligram). Calculate the micrograms of ceftazidime per milligram as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.204


where:
Au = Area of the ceftazidime peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As = Area of the ceftazidime peak in the chromatogram of the 
          ceftazidime working standard;
Ps = Ceftazidime activity in the ceftazidime working standard 
          solution in micrograms per milliliter;
Cu = Milligrams of sample per milliliter of sample solution;
m = Percent loss on drying (determined as directed in Sec. 436.200(h) of 
          this chapter if the formulation contains sodium carbonate and 
          determined as directed in Sec. 436.200(g) of this chapter if 
          the formulation contains L-arginine);
S = Percent sodium carbonate content of the sample (determined as 
directed in Sec. 436.357 of this chapter); and
A = Percent L-arginine content of the sample (determined as directed in 
Sec. 455.204 of this chapter, except use ceftazidime instead of 
aztreonam in the working standard solution and use water instead of 
mobile phase). Prepare the sample solution by diluting an accurately 
weighed portion of the contents of a vial with water to 0.2 milligram 
per milliliter (estimated). The resolution between the ceftazidime peak 
and the arginine peak is not less than 6.0, the asymmetry factor for the 
arginine peak is not more than 4.0).

    (b) Ceftazidime content (milligrams of ceftazidime per vial). 
Calculate the ceftazidime content of the vial as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.205

where:
Au=Area of the ceftazidime peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the ceftazidime peak in the chromatogram of the 
          ceftazidime working standard;
Ps=Ceftazidime activity in the ceftazidime working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 80 milligrams of ceftazidime per milliliter.
    (4) Loss on drying. Proceed as directed in Sec. 436.200(h) of this 
chapter if the formulation contains sodium carbonate and as directed in 
Sec. 436.200(g) of this chapter if the formulation contains L-arginine.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, 
preparing the sample solution as follows: reconstitute the sample in the 
sealed container to give an aqueous solution containing approximately 
100 milligrams per milliliter, relieving the pressure inside the 
container if necessary during the reconstitution.
    (6) Pyridine content. Proceed as directed in Sec. 436.358 of this 
chapter, using a temperature of 40  deg.C, an ultraviolet detection 
system operating at a wavelength of 254 nanometers, a column packed with 
microparticulate (5 micrometers in diameter) reversed phase packing 
material such as octadecyl hydrocarbon bonded silicas, a flow rate of 
1.6 milliliters per minute, and a known injection volume from 10 to 20 
microliters. Reagents, working standard and sample solutions, system 
suitability requirements, and calculations are as follows:
    (i) Reagents--(a) Phosphate buffer, pH 7.0. Dissolve 5.68 grams of 
sodium phosphate, dibasic, anhydrous and 3.63 grams of potassium 
phosphate, monobasic, in water and dilute to 1,000 milliliters.
    (b) Mobile phase. Mix 300 milliliters of acetonitrile and 100 
milliliters of 0.25M ammonium phosphate, monobasic, dilute to 1,000 
milliliters with water and add sufficient 10M ammonia solution to

[[Page 680]]

give a pH of 7.00.1. Filter the mobile phase through a 
suitable glass fiber filter or equivalent that is capable of removing 
particulate contamination to 1 micron in diameter. Degas the mobile 
phase just prior to its introduction into the chromatograph pumping 
system.
    (c) System suitability test solution. Prepare a solution in 
phosphate buffer, pH 7.0, containing 25 micrograms of pyridine and 25 
micrograms of an authentic sample of (6R, 7R)-7-[(Z)-2-(2-Aminothiazol-
4-yl)-2-(2-t- butoxy-carbonylprop-2-yloxyimino) acetamido]-3-(1-
pyridiniummethyl) ceph-3-em-4-carboxylate (t-butyl ceftazidime) per 
milliliter. Note, if no t-butyl ceftazidime is present in the sample 
solution, the working standard solution may be substituted for the 
system suitability test solution and the system suitability requirement 
for resolution for t-butyl ceftazidime is omitted.
    (ii) Preparation of working standard and sample solutions--(a) 
Working standard solution. Accurately weigh approximately 250 milligrams 
of pyridine into a 100-milliliter volumetric flask and dilute to volume 
with water to obtain a stock solution containing approximately 2,500 
micrograms of pyridine per milliliter. Mix well. Immediately prior to 
chromatography, further dilute 2.0 milliliters of stock solution to 200 
milliliters with phosphate buffer, pH 7.0, to obtain a solution 
containing 25 micrograms of pyridine per milliliter.
    (b) Sample solution. Accurately weigh approximately 660 milligrams 
of the sample into a 100-milliliter volumetric flask and add 50 
milliliters of phosphate buffer, pH 7.0. Shake until dissolved and 
dilute to volume with phosphate buffer, pH 7.0. Mix well. Store the 
solution at a temperature below 15  deg.C and inject into the 
chromatograph within 1 hour of preparation.
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 2.5 at 5 
percent of peak height.
    (b) Resolution. The resolution (R) between the peak for pyridine and 
the peak for t-butyl ceftazidime is satisfactory if it is not less than 
3.
    (c) Coefficient of variation. The coefficient of variation (SR 
in percent) of five replicate injections is satisfactory if it is not 
more than 3 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.358(b) of this chapter. Alternate chromatographic 
conditions are acceptable provided reproducibility and resolution are 
comparable to the system. However, the sample preparation described in 
paragraph (b)(6)(ii)(b) of this section should not be changed.
    (iv) Calculations. Calculate the pyridine content in percent of the 
sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.206

where:
Hu=Height of the pyridine peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
Hs=Height of the pyridine peak in the chromatogram of the 
          pyridine working standard;
Ps=Pyridine content of the pyridine working standard solution 
          in micrograms per milliliter; and
Cu=Milligrams of sample per milliliter of sample solution.
[50 FR 48400, Nov. 25, 1985; 50 FR 52917, Dec. 27, 1985; 50 FR 53308, 
Dec. 31, 1985; 51 FR 2478, Jan. 17, 1986. Redesignated at 54 FR 40652, 
Oct. 3, 1989, and amended at 55 FR 11583, Mar. 29, 1990; 56 FR 484, Jan. 
7, 1991; 59 FR 8398, Feb. 22, 1994]



Sec. 442.216b  Ceftazidime sodium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ceftazidime sodium injection is a frozen, 
aqueous, iso-osmotic solution of ceftazidime sodium which may contain 
one or more suitable and harmless buffer substances and a tonicity 
adjusting agent. Each milliliter contains ceftazidime sodium equivalent 
to 10, 20, or 40 milligrams of ceftazidime per milliliter. Its 
ceftazidime content is satisfactory if it is not less than 90 percent 
and not more than 120 percent of the number of milligrams of ceftazidime 
that it is represented to contain. It is sterile. It is nonpyrogenic. 
Its pH is not less than 5.0 and not more than 7.5 It passes the

[[Page 681]]

identity test. The ceftazidime pentahydrate conforms to the standards 
prescribed by Sec. 442.16(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The ceftazidime pentahydrate used in making the batch for 
potency, loss on drying, pH, crystallinity, identity, and high molecular 
weight polymer content.
    (B) The batch for ceftazidime content, sterility, pyrogens, pH, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research;
    (A) The ceftazidime pentahydrate used in making the batch: 10 
packages, each containing 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Ceftazidime content. Proceed as directed in Sec. 442.216(b)(1), 
except prepare the sample solution and calculate the ceftazidime content 
as follows:
    (i) Preparation of sample solution. Remove an accurately measured 
representative portion from each container immediately after thawing and 
reaching room temperature and dilute with mobile phase to obtain a 
solution containing 100 micrograms of ceftazidime per milliliter 
(estimated). Prepare the sample solution just prior to its introduction 
into the chromatograph.
    (ii) Calculation. Calculate the milligrams of ceftazidime per 
milliliter of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.207

where:
Au=Area of the ceftazidime peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the ceftazidime peak in the chromatogram of the 
          ceftazidime working standard;
Ps=Ceftazidime activity in the ceftazidime working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
except inject a sufficient volume of the diluted solution to deliver 80 
milligrams of ceftazidime per kilogram.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (5) Identify. The high performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the ceftazidime working standard.
[54 FR 40652, Oct. 3, 1989]



Sec. 442.217  Ceftizoxime injectable dosage forms.



Sec. 442.217a  Sterile ceftizoxime sodium.

    The requirements for certification and the tests and methods of 
assay for sterile ceftizoxime sodium packaged for dispensing are 
described in Sec. 442.17a.
[48 FR 46272, Oct. 12, 1983. Redesignated at 49 FR 49286, Dec. 19, 1984]



Sec. 442.217b  Ceftizoxime sodium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ceftizoxime sodium injection is a frozen 
aqueous solution of ceftizoxime sodium with one or more suitable and 
harmless buffer substances in an isoosmotic diluent. Each milliliter 
contains ceftizoxime sodium equivalent to either 20 milligrams or 40 
milligrams of ceftizoxime per milliliter. Ceftizoxime content is 
satisfactory if it is not less than 90 percent and not more than 115 
percent of the represented number of milligrams of ceftizoxime. It is 
sterile. It is nonpyrogenic. Its pH is not less than

[[Page 682]]

5.5 and not more than 8.0. It passes the identity test. The ceftizoxime 
sodium used conforms to the standards prescribed by Sec. 442.17(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The ceftizoxime sodium used in making the batch for ceftizoxime 
content, moisture, pH, identity, and crystallinity.
    (b) The batch for ceftizoxime content, sterility, pyrogens, pH, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research, of:
    (a) The ceftizoxime sodium used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assays. Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Ceftizoxime content. Proceed as directed in Sec.  436.345 of 
this chapter, except prepare the sample solution and calculate the 
ceftizoxime content as follows:
    (i) Sample solution. Using a suitable hypodermic needle and syringe, 
transfer an accurately measured representative portion from each 
container, equivalent to 40 milligrams of ceftizoxime, to a 100-
milliliter volumetric flask. Dilute to volume with pH 7.0 buffer 
solution and mix. Transfer 10.0 milliliters of this solution to a 200-
milliliter volumetric flask, add 5.0 milliliters of internal standard 
solution, dilute to volume with pH 7.0 buffer solution, and mix.
    (ii) Calculations. Calculate the milligrams of ceftizoxime per 
milliliter of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.208

where:

Ru=Area of the ceftizoxime peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard)/Area of the internal standard peak;
Rs=Area of the ceftizoxime peak in the chromatogram of the 
          ceftizoxime working standard/Area of the internal standard 
          peak;
Ps=Ceftizoxime activity in the ceftizoxime working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
except inject a sufficient volume of the undiluted solution to deliver 
50 milligrams of ceftizoxime per kilogram.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (5) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the ceftizoxime working standard.
[49 FR 49286, Dec. 19, 1984; 50 FR 253, Jan. 3, 1985, as amended at 55 
FR 11583, Mar. 29, 1990]



Sec. 442.218  Cefuroxime injectable dosage forms.



Sec. 442.218a  Sterile cefuroxime sodium.

    The requirements for certification and the tests and methods of 
assay for sterile cefuroxime sodium packaged for dispensing are 
described in Sec. 442.18a.
[48 FR 38461, Aug. 24, 1983. Redesignated at 54 FR 40654, Oct. 3, 1989]



Sec. 442.218b  Cefuroxime sodium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefuroxime sodium injection is a frozen, 
aqueous, iso-osmotic solution of cefuroxime sodium which may contain one 
or more suitable and harmless buffer substances and a tonicity adjusting 
agent. Each milliliter

[[Page 683]]

contains cefuroxime sodium equivalent to 15 or 30 milligrams of 
cefuroxime per milliliter. Its cefuroxime content is satisfactory if it 
is not less than 90 percent and not more than 120 percent of the number 
of milligrams of cefuroxime that it is represented to contain. It is 
sterile. It is nonpyrogenic. Its pH is not less than 5.0 and not more 
than 7.5. It passes the identity test. The cefuroxime sodium used 
conforms to the standards prescribed by Sec. 442.18(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefuroxime sodium used in making the batch for potency, 
moisture, pH, and identity.
    (B) The batch for cefuroxime content, sterility, pyrogens, pH, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The cefuroxime sodium used in making the batch: 10 packages, 
each containing 1 gram.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Cefuroxime content. Proceed as directed in Sec. 436.343 of this 
chapter, except prepare the sample solution and calculate the cefuroxime 
content as follows:
    (i) Preparation of sample solution. Remove an accurately measured 
representative portion from each container immediately after thawing and 
reaching room temperature and dilute with water to obtain a solution 
containing 50 micrograms of cefuroxime per milliliter (estimated). 
Prepare the sample solution just prior to its introduction in the 
chromatograph.
    (ii) Calculation. Calculate the milligrams of cefuroxime per 
milliliter of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.209

where:

Au=Area of the cefuroxime peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefuroxime peak in the chromatogram of the 
          cefuroxime working standard;
Ps=Cefuroxime activity in the cefuroxime working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) this chapter, 
except inject a sufficient volume of the undiluted solution to deliver 
50 milligrams of cefuroxime per kilogram.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (5) Identity. The high performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the cefuroxime working standard.
[54 FR 40654, Oct. 3, 1989]



Sec. 442.220  Sterile cefonicid sodium.

    The requirements for certification and the tests and methods of 
assay for sterile cefonicid sodium packaged for dispensing are described 
in Sec. 442.20a.
[49 FR 34349, Aug. 30, 1984]



Sec. 442.222  Cefmenoxime hydrochloride for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefmenoxime hydrochloride for injection 
is a dry mixture of cefmenoxime hydrochloride and sodium carbonate. Each 
milligram of cefmenoxime hydrochloride for injection contains not less 
than 869 and not more than 1,015 micrograms of cefmenoxime on an 
anhydrous and sodium carbonate-free basis. Its

[[Page 684]]

cefmenoxime content is satisfactory if it contains not less than 90 
percent and not more than 115 percent of the number of milligrams of 
cefmenoxime that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its loss on drying is not more than 1.5 percent. Its pH in 
an aqueous solution containing 100 milligrams per milliliter is not less 
than 6.4 and not more than 7.9. The cefmenoxime hydrochloride used 
conforms to the standards prescribed by Sec. 442.22a(a)(1) of this 
chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefmenoxime hydrochloride used in making the batch for 
cefmenoxime content, moisture, identity, and crystallinity.
    (B) The batch for cefmenoxime content, sterility, pyrogens, loss on 
drying, pH, and sodium carbonate content.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The cefmenoxime hydrochloride used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Cefmenoxime content. Proceed as 
directed in Sec. 436.363 of this chapter, using ambient temperature, an 
ultraviolet detection system operating at a wavelength of 254 
nanometers, a column packed with microparticulate (3 to 10 micrometers 
in diameter) reversed phase packing material such as octadecyl 
hydrocarbon bonded silicas, a flow rate not to exceed 2.0 milliliters 
per minute, and a known injection volume between 10 and 20 microliters. 
Reagents, working standard and sample solutions, system suitability 
requirements, and calculations are as follows:
    (i) Reagents--(A) 0.1M Phosphate buffer solution, pH 6.8. Dissolve 
6.4 grams of monobasic potassium phosphate and 18.9 grams of dibasic 
sodium phosphate in 750 milliliters of water. Adjust the pH to 6.8 with 
1N sodium hydroxide and dilute to 1,000 milliliters.
    (B) Internal standard solution. Dissolve and dilute 0.15 gram of 
phthalimide in methanol to 100 milliliters.
    (C) Mobile phase. Mix water:acetonitrile:glacial acetic acid 
(50:10:1). Filter through a suitable filter capable of removing 
particulate matter to 0.5 micron in diameter. Degas the mobile phase 
just prior to its introduction into the chromatograph.
    (ii) Preparation of working standard and sample solutions--(A) 
Working standard solution. Dissolve approximately 50 milligrams of the 
cefmenoxime working standard, accurately weighed, in 10 milliliters of 
0.1M phosphate buffer solution, pH 6.8 and dilute to 50 milliliters with 
mobile phase. Transfer 4.0 milliliters of this solution to a 50-
milliliter volumetric flask, add 20 milliliters of internal standard 
solution and dilute to volume with mobile phase to obtain a solution 
containing 80 micrograms of cefmenoxime per milliliter.
    (B) Sample solutions. Determine both micrograms of cefmenoxime per 
milligram of the sample and milligrams of cefmenoxime per container. Use 
separate containers for preparation of each sample solution as described 
in paragraphs (b)(1)(ii)(B) (1) and (2) of this section.
    (1) Micrograms of cefmenoxime per milligram. Dissolve the accurately 
weighed dry contents of a sample with sufficient distilled water to 
obtain a solution containing 1 milligram of cefmenoxime per milliliter 
(estimated). Transfer 4.0 milliliters of this solution to a 50-
milliliter volumetric flask, add 20 milliliters of internal standard 
solution and dilute to volume with mobile phase to obtain a solution 
containing 80 micrograms of cefmenoxime per milliliter (estimated).
    (2) Milligrams of cefmenoxime per container. Reconstitute the sample 
as directed in the labeling. Then, using a suitable hypodermic needle 
and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency in a given

[[Page 685]]

volume of the resultant preparation, remove an accurately measured 
representative portion from each container. Dilute the solution thus 
obtained with sufficient distilled water to obtain a solution containing 
1 milligram of cefmenoxime per milliliter (estimated). Transfer 4.0 
milliliters of this solution to a 50-milliliter volumetric flask, add 20 
milliliters of internal standard solution and dilute to volume with 
mobile phase to obtain a solution containing 80 micrograms of 
cefmenoxime per milliliter (estimated).
    (iii) System suitability requirements--(A) Tailing factor. The 
tailing factor (T) for the cefmenoxime peak is satisfactory if it is not 
more than 1.6 at 5 percent of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,200 theoretical plates for the 
cefmenoxime peak.
    (C) Resolution. The resolution (R) between the peak for cefmenoxime 
and phthalimide is satisfactory if it is not less than 2.3.
    (D) Coefficient of variation. The coefficient of variation (SR 
in percent) of 5 replicate injections is satisfactory if it is not more 
than 2.0 percent. If the system suitability requirements have been met, 
then proceed as described in Sec. 436.363(b) of this chapter.
    (iv) Calculations--(A) Micrograms per milligram. Calculate the 
micrograms of cefmenoxime per milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.210

where:

Ru=Area of the cefmenoxime peak in the chromatogram of the 
          sample/Area of internal standard peak;
Rs=Area of the cefmenoxime peak in the chromatogram of the 
          cefmenoxime working standard/Area of internal standard 
          peak;
Ps=Cefmenoxime activity in the cefmenoxime working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of sample solution;
d=Dilution factor of the sample;
L=Percent loss on drying (determined as directed in paragraph (b)(4) of 
          this section); and
S=Percent sodium carbonate (determined as directed in paragraph (b)(6) 
          of this section).

    (B) Milligrams of cefmenoxime per vial. Calculate the cefmenoxime 
content of the vial as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.211

where:

Ru=Area of the cefmenoxime peak in the chromatogram of the 
          sample/Area of internal standard peak;
Rs=Area of the cefmenoxime peak in the chromatogram of the 
          cefmenoxime working standard/Area of internal standard peak;
Ps=Cefmenoxime activity in the cefmenoxime working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 60 milligrams of cefmenoxime per milliliter.
    (4) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (6) Sodium carbonate content. Proceed as directed in Sec. 436.364 of 
this chapter.
[53 FR 13403, Apr. 25, 1988; 53 FR 19369, May 27, 1988]



Sec. 442.223  Sterile cephaloridine.

    The requirements for certification and the tests and methods of 
assay for sterile cephaloridine packaged for dispensing are described in 
Sec. 442.23a.
[39 FR 19040, May 30, 1974, as amended at 55 FR 11583, Mar. 29, 1990]



Sec. 442.225  Cephalothin sodium injectable dosage forms.



Sec. 442.225a  Sterile sodium cephalothin.

    The requirements for certification and the tests and methods of 
assay for sterile sodium cephalothin packaged for dispensing are 
described in Sec. 442.25a.
[39 FR 19040, May 30, 1974. Redesignated at 40 FR 11351, Mar. 11, 1975]

[[Page 686]]



Sec. 442.225b  Cephalothin sodium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephalothin sodium injection is a frozen 
aqueous solution of cephalothin sodium with one or more suitable and 
harmless buffer substances. It may contain sodium chloride or dextrose. 
Each milliliter contains cephalothin sodium equivalent to 20 milligrams, 
40 milligrams, or 100 milligrams of cephalothin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of milligrams of cephalothin that it is 
represented to contain. It is sterile. It is nonpyrogenic. Its pH is not 
less than 6.0 and not more than 8.5. The cephalothin sodium used 
conforms to the standards prescribed by Sec. 442.25a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cephalothin sodium used in making the batch for potency, 
loss on drying, pH, specific rotation, identity, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, and pH.
    (ii) Samples required:
    (a) The cephalothin sodium used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the ampoule contents as 
directed in the labeling. The sample solution used for testing must be 
at room temperature.
    (1) Potency. Use either of the following methods; however, the 
results obtained from the microbiological agar diffusion assay shall be 
conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Using a suitable hypodermic needle and syringe, remove an accurately 
measured representative portion from each container and dilute with 
sufficient 1 percent potassium phosphate buffer, pH 6.0 (solution 1), to 
give a stock solution of convenient concentration. Further dilute an 
aliquot of the stock solution with solution 1 to the reference 
concentration of 1.0 microgram of cephalothin per milliliter 
(estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter, preparing the sample as follows: Using a 
suitable hypodermic needle and syringe, remove an accurately measured 
representative portion from each container and dilute with distilled 
water to give a stock solution of convenient concentration. Further 
dilute with distilled water to the prescribed concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cephalothin per milliliter.
    (4) [Reserved]
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[40 FR 11351, Mar. 11, 1975, as amended at 49 FR 13493, Apr. 5, 1984; 50 
FR 19919, May 13, 1985]



Sec. 442.225c  Cephalothin sodium for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephalothin sodium for injection is a dry 
mixture of cephalothin sodium with one or more suitable and harmless 
buffer substances. The cephalothin sodium may be isolated in the 
manufacture of cephalothin sodium for injection. Its cephalothin content 
is satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of milligrams of cephalothin that it is 
represented to contain. It is sterile. It is nonpyrogenic. Its loss on 
drying is not more than 1.5 percent. When reconstituted as directed in 
the labeling, its pH is not less than 6.0 and not more than

[[Page 687]]

8.5. If isolated, the cephalothin sodium used conforms to the standards 
prescribed by Sec. 442.25a(a)(1). If the cephalothin sodium is not 
isolated: The potency of the dry mixture is not less than 850 micrograms 
of cephalothin per milligram on an anhydrous basis when corrected for 
sodium bicarbonate; the specific rotation of the dry mixture in an 
aqueous solution containing 50 milligrams of cephalothin per milliliter 
at 25 deg. C is +129 deg. plus-minus5 deg.; and the dry 
mixture gives a positive identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) If isolated, the cephalothin sodium used in making the batch for 
potency, loss on drying, pH, specific rotation, identity, and 
crystallinity.
    (b) The batch for potency, sterility, pyrogens, loss on drying, and 
pH. In addition, if the cephalothin sodium is not isolated, results of 
tests and assays on the dry mixture for potency, specific rotation, and 
identity.
    (ii) Samples required:
    (a) For all tests except sterility: A minimum of 10 immediate 
containers, unless the cephalothin sodium is not isolated, a minimum of 
15 immediate containers.
    (b) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Content; potency--(i) Sample 
preparation. Reconstitute as directed in the labeling. Then using a 
suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is represented as a single dose container; or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute with 1 percent potassium phosphate 
buffer, pH 6.0 (solution 1), for the microbiological agar diffusion 
assay or distilled water for the hydroxylamine colorimetric assay to 
obtain a stock solution of convenient concentration. In addition, if the 
cephalothin sodium is not isolated, dissolve an accurately weighed 
sample in sufficient 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), for the microbiological agar diffusion assay or distilled 
water for the hydroxylamine colorimetric assay to obtain a stock 
solution of convenient concentration. Correct the potency, micrograms of 
cephalothin per milligram, for sodium bicarbonate content determined as 
described in paragraph (b)(7) of this section.
    (ii) Assay procedures. Use either of the following methods; however, 
the results obtained from the hydroxylamine colorimetric assay shall be 
conclusive.
    (a) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, diluting an aliquot of the stock solution 
with solution 1 to the reference concentration of 1.0 microgram of 
cephalothin per milliliter (estimated).
    (b) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 436.205 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cephalothin per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
    (7) Specific rotation. Dissolve and dilute an accurately weighed 
portion of the dry mixture with sufficient distilled water to give a 
concentration of approximately 50 milligrams per milliliter. Proceed as 
directed in Sec. 436.210 of this chapter, using a 1.0-decimeter 
polarimeter tube. Calculate the specific rotation on an anhydrous basis 
and correct for sodium bicarbonate content. Determine the sodium 
bicarbonate content as follows: Dissolve an accurately weighed portion 
of the dry mixture, approximately 1.0 gram, with approximately 50 
milliliters of distilled water. Titrate with 0.1N sulfuric acid.

[[Page 688]]

Determine the end-point potentiometrically using a glass calomel 
combination electrode. Each milliliter of 0.1N sulfuric acid is 
equivalent to 8.401 milligrams of sodium bicarbonate.
    (8) Identity. Using a 0.0025-percent solution of the sample in water 
and a suitable spectrophotometer, record the ultraviolet absorption 
spectrum from 220 to 310 nanometers. The spectrum compares qualitatively 
to that of the working standard similarly tested.
[40 FR 5355, Feb. 5, 1975, as amended at 46 FR 38503, July 28, 1981; 48 
FR 51293, Nov. 8, 1983; 49 FR 5097, Feb. 10, 1984; 50 FR 19919, May 13, 
1985]



Sec. 442.229  Sterile cephapirin sodium.

    The requirements for certification and the tests and methods of 
assay for sterile cephapirin sodium packaged for dispensing are 
described in Sec. 442.29a.



Sec. 442.240  Cephradine injectable dosage forms.



Sec. 442.240a  Cephradine for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephradine for injection is a dry mixture 
of cephradine and one or more suitable and harmless solubilizing and 
buffering agents. Its potency is satisfactory if it contains not less 
than 90 percent and not more than 115 percent of the number of 
milligrams of cephradine that it is represented to contain. It is 
sterile. It is nonpyrogenic. Its loss on drying is not more than 5.0 
percent. Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 8.0 and not more than 9.6. The cephradine 
used conforms to the standards prescribed by Sec. 442.40a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The sterile cephradine used in making the batch for potency, 
moisture, pH, cephalexin content, identity, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, loss on drying, and 
pH.
    (ii) Samples required:
    (a) The cephradine used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological agar diffusion assay shall be conclusive.
    (i) Microbiological agar diffusion assay. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute the sample as directed in the labeling for intramuscular 
use. Using a suitable hypodermic needle and syringe, remove all of the 
withdrawable contents if it is represented as a single dose container; 
or if the labeling specifies the amount of potency in a given volume of 
the resultant preparation, remove an accurately measured representative 
portion from each container. Further dilute an aliquot of this solution 
with solution 1 to the reference concentration of 10.0 micrograms of 
cephradine per milliliter (estimated).
    (ii) Hydroxylamine colorimetric assay. Proceed as directed in 
Sec. 442.40(b)(1)(ii), preparing the sample as follows: Reconstitute the 
sample as directed in the labeling for intramuscular use. Using a 
suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is represented as a single dose container; or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Further dilute an aliquot of this solution 
with distilled water to 1 milligram of cephradine per milliliter 
(estimated)
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 80 milligrams of cephradine per milliliter.
    (4) [Reserved]

[[Page 689]]

    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
[40 FR 51626, Nov. 6, 1975. Redesignated at 43 FR 14646, Apr. 7, 1978; 
50 FR 19919, May 13, 1985]



Sec. 442.240b  Sterile cephradine.

    The requirements for certification and the tests and methods of 
assay for sterile cephradine packaged for dispensing are described in 
Sec. 442.40a.
[43 FR 14646, Apr. 7, 1978]



Sec. 442.250  Ceforanide for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ceforanide for injection is a dry mixture 
of ceforanide and L-lysine. Each milligram of ceforanide for injection 
contains not less than 900 micrograms and not more than 1,050 micrograms 
of ceforanide when corrected for L-lysine content. Its ceforanide 
content is satisfactory if it contains not less than 90 percent and not 
more than 115 percent of the number of milligrams of ceforanide that it 
is represented to contain. It is sterile. It is nonpyrogenic. Its 
moisture content is not more than 3.0 percent. When reconstituted as 
directed in the labeling, its pH is not less than 5.5 and not more than 
8.5. The ceforanide used conforms to the standards prescribed by 
Sec. 442.50a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The sterile ceforanide used in making the batch for ceforanide 
content, moisture, pH, and identity.
    (b) The batch for ceforanide content, sterility, pyrogens, moisture, 
and pH.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The ceforanide used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Ceforanide content. Determine 
both micrograms of ceforanide per milligram of sample and milligrams of 
ceforanide per container. Proceed as directed in Sec. 436.348 of this 
chapter, preparing the sample solution and calculating the ceforanide 
content as follows:
    (i) Preparation of sample solution. Use separate containers for 
preparation of each sample solution as described in paragraph (b)(1)(i) 
(a) and (b) of this section.
    (a) Micrograms of ceforanide per milligram. Prepare a solution 
containing 1.0 milligrams per milliliter in mobile phase. Inject each 
sample within 5 minutes after dissolution.
    (b) Milligrams of ceforanide per container. Reconstitute the sample 
with distilled water as directed in the labeling. Using a suitable 
hypodermic needle and syringe, remove all of the withdrawable contents 
if it is represented as a single-dose container; or, if the labeling 
specifies the amount of ceforanide content in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute with mobile phase to obtain a stock 
solution containing 10.0 milligrams per milliliter (estimated). 
Immediately dilute an aliquot of the stock solution with mobile phase to 
a concentration of 1.0 milligrams of ceforanide per milliliter 
(estimated). Inject within 5 minutes, after preparation.
    (ii) Calculations--(a) Micrograms of ceforanide per milligram. 
Calculate the micrograms of ceforanide per milligram of sample as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.212

where:
Au=Area of the ceforanide sample peak (at a retention time 
          equal to that observed for the standard);
As=Area of the ceforanide peak in the chromatogram of the 
          ceforanide working

[[Page 690]]

          standard;
Ps=Ceforanide activity in the ceforanide working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of sample solution; 
          and
L=Percent lysine content of the sample. (Determined as described in 
          Sec. 436.349 of this chapter.)

    (b) Milligrams of ceforanide per vial. Calculate the ceforanide 
content of the vial as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.213

where:
Au=Area of the ceforanide sample peak (at a retention time 
          equal to that observed for the standard);
As=Area of the ceforanide peak in the chromatogram of the 
          ceforanide working standard;
Ps=Ceforanide activity in the ceforanide working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
reconstitute the vials with approximately 3.0 milliliters of diluting 
fluid A per each gram of antibiotic activity. Transfer approximately 1 
milliliter from each of 20 vials into a sterile 500-milliliter 
Erlenmeyer flask containing 200 milliliters of diluting fluid A. Filter 
as described in paragraph (e)(1)(ii) of this section, except in lieu of 
filtering with three 100-milliliter quantities of diluting fluid A, 
rinse the filter membrane with three 100-milliliter portions of diluting 
fluid D followed by a final rinse with 100 milliliters of diluting fluid 
A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of ceforanide per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution obtained when the product is reconstituted as directed in 
the labeling.
[49 FR 25848, June 25, 1984; 49 FR 34347, Aug. 30, 1984; 49 FR 40006, 
Oct. 12, 1984, as amended at 55 FR 11583, Mar. 29, 1990]



Sec. 442.253  Cefotetan injectable dosage forms.



Sec. 442.253a  Sterile cefotetan disodium.

    The requirements for certification and the tests and methods of 
assay for sterile cefotetan disodium packaged for dispensing are 
described in Sec. 442.53a.
[51 FR 20264, June 4, 1986. Redesignated at 59 FR 26941, May 25, 1994]



Sec. 442.253b  Cefotetan sodium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefotetan sodium injection is a frozen, 
aqueous, iso-osmotic solution of cefotetan and sodium bicarbonate. It 
contains one or more suitable and harmless buffer substances and a 
tonicity adjusting agent. Each milliliter contains cefotetan disodium 
equivalent to 20 milligrams or 40 milligrams of cefotetan per 
milliliter. Its cefotetan content is satisfactory if it is not less than 
90 percent and not more than 120 percent of the number of milligrams of 
cefotetan that it is represented to contain. It is sterile. It contains 
not more than 0.17 endotoxin units per milligram of cefotetan. Its pH is 
not less than 4.0 and not more than 6.5. It passes the identity test. 
The cefotetan used conforms to the standards prescribed by 
Sec. 442.52(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefotetan used in making the batch for cefotetan potency, 
moisture, and identity.
    (B) The batch for cefotetan potency, sterility, bacterial 
endotoxins, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The cefotetan used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 12 immediate 
containers.

[[Page 691]]

    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Cefotetan potency. Proceed as directed in Sec. 442.52(b)(1), 
except prepare the sample solution and calculate the cefotetan content 
as follows:
    (i) Preparation of sample solution. Using a suitable hypodermic 
needle and syringe, remove an accurately measured portion from each 
container immediately after thawing and reaching room temperature and 
dilute with mobile phase to obtain a solution containing 200 micrograms 
of cefotetan per milliliter (estimated). Prepare the sample solution 
just prior to its introduction into the chromatograph.
    (ii) Calculation. Calculate the milligrams of cefotetan per 
milliliter of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.214

where:
AU=Area of the cefotetan peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
AS=Area of the cefotetan peak in the chromatogram of the 
          cefotetan working standard;
PS=Cefotetan activity in the cefotetan working standard 
          solution in micrograms per milliliter;
CU=Milligrams of sample per milliliter of sample solution; 
          and
m = Percent moisture content of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Bacterial endotoxins. Proceed as directed in the U.S. 
Pharmacopeia bacterial endotoxins test.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (5) Identity. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the cefotetan working standard.
[59 FR 26941, May 25, 1994]



Sec. 442.255  Ceftriaxone injectable dosage forms.



Sec. 442.255a  Sterile ceftriaxone sodium.

    The requirements for certification and the tests and methods of 
assay for sterile ceftriaxone sodium packaged for dispensing as 
described in Sec. 442.55a.
[50 FR 10001, Mar. 13, 1985. Redesignated at 52 FR 44860, Nov. 23, 1987]



Sec. 442.255b  Ceftriaxone sodium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ceftriaxone sodium injection is a frozen 
aqueous iso-osmotic solution of ceftriaxone sodium which may contain one 
or more suitable and harmless buffer substances. Each milliliter 
contains ceftriaxone sodium equivalent to 10, 20, or 40 milligrams of 
ceftriaxone per milliliter. Its ceftriaxone content is satisfactory if 
it is not less than 90 percent and not more than 115 percent of the 
number of milligrams of ceftriaxone that it is represented to contain. 
It is sterile. It is nonpyrogenic. Its pH is not less than 6.0 and not 
more than 8.0. It passes the identity test. The ceftriaxone sodium used 
conforms to the standards prescribed by Sec. 442.55(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The ceftriaxone sodium used in making the batch for potency, 
moisture, pH, crystallinity, and identity.
    (B) The batch for content, sterility, pyrogens, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The ceftriaxone sodium used in making the batch: 10 packages, 
each containing 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.

[[Page 692]]

    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Ceftriaxone content. Proceed as directed in Sec. 442.55a(b)(1) 
of this chapter, except prepare the sample solution and calculate the 
ceftriaxone content as follows:
    (i) Preparation of sample solution. Using a suitable hypodermic 
needle and syringe, remove an accurately measured representative portion 
from each container immediately after thawing and reaching room 
temperature and dilute with mobile phase to obtain a solution containing 
180 micrograms of ceftriaxone per milliliter (estimated). Prepare the 
sample solution just prior to its introduction into the chromatograph.
    (ii) Calculation. Calculate the milligrams of ceftriaxone anhydrous 
free acid per milliliter of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.215

where:
Au=Area of the ceftriaxone peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the ceftriaxone peak in the chromatogram of the 
          ceftriaxone working standard;
Ps=Ceftriaxone activity in the ceftriaxone working standard 
          solution in micrograms of anhydrous free acid per milliliter; 
          and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
except inject a sufficient volume of the undiluted solution to deliver 
40 milligrams of ceftriaxone per kilogram.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (5) Identify. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the ceftriaxone working standard.
[52 FR 44860, Nov. 23, 1987, as amended at 55 FR 11583, Mar. 29, 1990]



Sec. 442.258  Cefotiam dihydrochloride for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefotiam dehydrochloride for injection is 
a dry mixture of cefotiam dihydrochloride and sodium carbonate. Its 
cefotiam potency is satisfactory if each milligram of cefotiam 
dihydrochloride for injection contains not less than 790 micrograms and 
not more than 925 micrograms of cefotiam on an anhydrous basis, when 
corrected for sodium carbonate content. Its cefotiam content is 
satisfactory if it contains not less than 90 percent and not more than 
120 percent of the number of milligrams of cefotiam that it is 
represented to contain. It is sterile. It is nonpyrogenic. Its loss on 
drying is not more than 6.0 percent. The pH of an aqueous solution 
containing 100 milligrams per milliliter is not less than 5.7 and not 
more than 7.2. The cefotiam dihydrochloride used conforms to the 
standards prescribed by Sec. 442.58a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefotiam dihydrochloride used in making the batch for 
potency, moisture, identity, and crystallinity.
    (B) The batch for cefotiam potency, cefotiam content, sterility, 
pyrogens, loss on drying, pH, and sodium carbonate content.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The cefotiam dihydrochloride used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.

[[Page 693]]

    (b) Tests and methods of assay--(1) Cefotiam potency and content. 
Determine both micrograms of cefotiam per milligram of sample and 
milligrams of cefotiam per container. Proceed as directed in 
Sec. 442.58a(b)(1), preparing the sample solutions and calculating the 
potency and content as follows:
    (i) Preparation of sample solutions. Use separate containers for 
preparation of each sample solution as described in paragraphs (b)(1)(i) 
(A) and (B) of this section.
    (A) Cefotiam potency (micrograms of cefotiam per milligram). 
Dissolve an accurately weighed sample with sufficient distilled water to 
obtain a solution containing approximately 1,000 micrograms of cefotiam 
per milliliter. Further dilute this solution with mobile phase to obtain 
a solution containing 50 micrograms of cefotiam activity per milliliter 
(estimated).
    (B) Cefotiam content (milligrams of cefotiam per vial). Reconstitute 
the sample as directed in the labeling. Then, using a suitable 
hypodermic needle and syringe, remove all of the withdrawable contents 
if it is represented as a single-dose container; or, if the labeling 
specifies the amount of potency in a given volume of the resultant 
preparation, remove an accurately measured representative portion from 
each container. Dilute the solution thus obtained with sufficient 
distilled water to obtain a solution containing 1,000 micrograms of 
cefotiam activity per milliliter (estimated). Further dilute this 
solution with mobile phase to obtain a solution containing 50 micrograms 
of cefotiam activity per milliliter (estimated).
    (ii) Calculations--(A) Cefotiam potency (micrograms per milligram). 
Calculate the micrograms of cefotiam per milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.216

where:
Au=Area of the cefotiam peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefotiam peak in the chromatogram of the 
          cefotiam working standard;
Ps=Cefotiam activity in the cefotiam working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of the sample per milliliter of sample 
          solution;
L=Percent loss on drying (determined as directed in paragraph (b)(4) of 
          this section); and
S=Percent sodium carbonate (determined as directed in paragraph (b)(6) 
          of this section).

    (B) Cefotiam content (milligrams of cefotiam per vial). Calculate 
the cefotiam content of the vial as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.217

where:
Au=Area of the cefotiam peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cefotiam peak in the chromatogram of the 
          cefotiam working standard;
Ps=Cefotiam activity in the cefotiam working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(g) of this chapter, 
using a solution containing 40 milligrams of cefotiam per milliliter.
    (4) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (6) Sodium carbonate content. Proceed as directed in Sec. 436.357 of 
this chapter.
[54 FR 20786, May 15, 1989]



Sec. 442.260  Cefpiramide sodium for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefpiramide sodium for injection is a dry 
mixture of cefpiramide and sodium benzoate. It contains other buffers 
and preservatives. Its cefpiramide potency is satisfactory if each 
milligram of cefpiramide sodium for injection contains not less than 754 
micrograms and not more than 924

[[Page 694]]

micrograms of cefpiramide on an anhydrous basis. Its cefpiramide content 
is satisfactory if it contains not less than 90 percent and not more 
than 120 percent of the number of milligrams of cefpiramide that it is 
represented to contain. It is sterile. It is nonpyrogenic. Its moisture 
content is not more than 3.0 percent. Its pH in an aqueous solution 
containing 100 milligrams per milliliter is not less than 6.0 and not 
more than 8.0. It passes the identity test. The cefpiramide used 
conforms to the standards prescribed by Sec. 442.60(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefpiramide used in making the batch for potency, moisture, 
pH, total related substances, specific rotation, identity, and 
crystallinity.
    (B) The batch for cefpiramide potency, cefpiramide content, 
sterility, pyrogens, moisture, pH, and identity.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research:
    (A) The cefpiramide used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Cefpiramide potency and content. 
Determine both micrograms of cefpiramide per milligram of sample and 
milligrams of cefpiramide per container. Proceed as directed in 
Sec. 442.60(b)(1), preparing the sample solutions and calculating the 
potency and content as follows:
    (i) Preparation of sample solutions. Use separate containers for 
preparation of each sample solution as described in paragraphs 
(b)(1)(i)(A) and (b)(1)(i)(B) of this section.
    (A) Cefpiramide potency (micrograms of cefpiramide per milligram). 
Dissolve an accurately weighed sample with sufficient mobile phase to 
obtain a solution containing approximately 0.25 milligram of cefpiramide 
per milliliter (estimated).
    (B) Cefpiramide content (milligrams of cefpiramide per vial). 
Reconstitute the sample as directed in the labeling. Then, using a 
suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is represented as a single-dose container; or, if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute the solution thus obtained with 
sufficient distilled water to obtain a solution containing 1.0 milligram 
of cefpiramide activity per milliliter (estimated). Further dilute this 
solution with mobile phase to obtain a solution containing 0.25 
milligram of cefpiramide activity per milliliter (estimated).
    (ii) Calculations--(A) Cefpiramide potency (micrograms per 
milligram). Calculate the micrograms of cefpiramide per milligram as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.218

where:
    Au=Area of the cefpiramide peak in the chromatogram of 
the sample (at a retention time equal to that observed for the 
standard);
    As=Area of the cefpiramide peak in the chromatogram of 
the cefpiramide working standard;
    Ps=Cefpiramide activity in the cefpiramide working 
standard solution in micrograms per milliliter;
    Cu=Milligrams of the sample per milliliter of sample 
solution;
    m =Percent moisture content of the sample.

    (B) Cefpiramide content (milligrams of cefpiramide per vial). 
Calculate the cefpiramide content of the vial as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.219

where:
    Au=Area of the cefpiramide peak in the chromatogram of 
the sample (a ta retention time equal to that observed for the 
standards);

[[Page 695]]

    As=Area of the cefpiramide peak in the chromatogram of 
the cefpiramide working standard;
    Ps=Cefpiramide activity in the cefpiramide working 
standard solution in micrograms per milliliter; and
    d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in Sec. 436.20(e)(1).
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of cefpiramide per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (6) Identify. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the cefpiramide working standard.
[55 FR 14242, Apr. 17, 1990]



Sec. 442.270  Cefmetazole injectable dosage forms.



Sec. 442.270a  Sterile cefmetazole sodium.

    The requirements for certification and the tests and methods of 
assay for sterile cefmetazole sodium packaged for dispensing are 
described in Sec. 442.70a.
[55 FR 6636, Feb. 26, 1990]



Sec. 442.270b  Cefmetazole sodium injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cefmetazole sodium injection is a frozen, 
aqueous, iso-osmotic solution of cefmetazole and sodium citrate. It 
contains one or more suitable and harmless buffer substances and a 
tonicity adjusting agent. Each milliliter contains cefmetazole sodium 
equivalent to 20 milligrams or 40 milligrams of cefmetazole per 
milliliter. Its cefmetazole content is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the number of 
milligrams of cefmetazole that it is represented to contain. It is 
sterile. It contains not more than 0.2 endotoxin units per milligram. 
Its pH is not less than 4.2 and not more than 6.2. It passes the 
identity test. The cefmetazole used conforms to the standards prescribed 
by Sec. 442.69(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cefmetazole used in making the batch for potency, moisture, 
and identity.
    (B) The batch for potency, sterility, bacterial endotoxins, pH, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The cefmetazole used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 12 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Cefmetazole potency. Proceed as directed in Sec. 442.70a(b)(1), 
except prepare the sample solution and calculate the cefmetazole content 
as follows:
    (i) Preparation of sample solution. Using a suitable hypodermic 
needle and syringe, remove an accurately measured portion from each 
container immediately after thawing and reaching room temperature and 
dilute with mobile phase to obtain a solution containing 500 micrograms 
of cefmetazole per milliliter (estimated). Prepare the sample solution 
just prior to its introduction into the chromatograph.
    (ii) Calculation. Calculate the milligrams of cefmetazole per 
milliliter of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.220

where:

AU=Area of the cefmetazole peak in the chromatogram of the - 
          sample (at a retention time equal to that observed for the 
          standard);

[[Page 696]]

AS=Area of the cefmetazole peak in the chromatogram of the 
          cefmetazole working standard;
PS=Cefmetazole activity in the cefmetazole working standard 
          solution in micrograms per milliliter; and
d = Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Bacterial endotoxins. Proceed as directed in the United States 
Pharmacopeia bacterial endotoxins test.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (5) Identity. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the cefmetazole working standard.
[59 FR 12546, Mar. 17, 1994]



PART 443--CARBACEPHEM ANTIBIOTIC DRUGS--Table of Contents




                          Subpart A--Bulk Drugs

Sec.
443.20  Loracarbef.

                      Subpart B--Oral Dosage Forms

443.120  Loracarbef oral dosage forms.
443.120a  Loracarbef capsules.
443.120b  Loracarbef for oral suspension.

    Authority:  21 U.S.C. 357.

    Source:  58 FR 26667, May 4, 1993, unless otherwise noted.



                          Subpart A--Bulk Drugs



Sec. 443.20  Loracarbef.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Loracarbef is the monohydrate form of 
(6R,7S)-7-[(R)-2-amino-2-phenylacetamido]-3-chloro-8-oxo-1-
azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid. It is so purified and 
dried that:
    (i) Its potency is not less than 960 micrograms and not more than 
1,020 micrograms of loracarbef activity per milligram, on an anhydrous 
basis.
    (ii) Its moisture content is not less than 3.5 percent and not more 
than 6.0 percent.
    (iii) The pH of an aqueous slurry containing 100 milligrams per 
milliliter is not less than 3.5 and not more than 5.5.
    (iv) Its specific rotation in an 0.1 N HCl solution containing 10 
milligrams of loracarbef per milliliter at 25 deg. C is 
+27 deg.to+33 deg. on an anhydrous basis.
    (v) It is crystalline.
    (vi) It gives a positive identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for loracarbef potency, 
moisture, pH, specific rotation, crystallinity, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 265 nanometers, a 25-
centimeter by 4.6-millimeter (inside diameter) column packed with 
microparticulate (5 micrometers in diameter) reversed phase packing 
material such as octadecyl silane bonded to silicas, a flow rate of 1.5 
milliliters per minute, and a known injection volume between 10 and 20 
microliters. The retention time for loracarbef is between 10 and 13 
minutes. Mobile phase, working standard, sample and resolution test 
solutions, system suitability requirements, and calculations are as 
follows:
    (i) Mobile phase. Dissolve 2.0 grams of pentanesulfonic acid sodium 
salt monohydrate in 1,560 milliliters of water. Add 20 milliliters of 
triethylamine. Adjust the pH to 2.5 with phosphoric acid. Add 440 
milliliters of methanol and mix. Filter the mobile phase through a 
suitable filter capable of removing particulate matter 0.5 micron in 
diameter and degas it just prior to its introduction into the 
chromatograph.

[[Page 697]]

    (ii) Preparation of working standard, sample, and resolution test 
solutions--(A) Working standard solution. Accurately weigh approximately 
10 milligrams of the loracarbef working reference standard into a 50-
milliliter volumetric flask. Dissolve and dilute to volume with mobile 
phase. Brief sonication may be required to obtain complete dissolution 
of the material.
    (B) Sample solution. Accurately weigh approximately 10 milligrams of 
sample into a 50-milliliter volumetric flask. Dissolve and dilute to 
volume with mobile phase. Brief sonication may be required to obtain 
complete dissolution of the material.
    (C) Resolution test solution. Prepare a resolution test solution 
containing approximately 0.2 milligram per milliliter each of loracarbef 
and loracarbef L-isomer in the mobile phase.
    (iii) System suitability requirements--(A) Asymmetry factor. The 
asymmetry factor (AS) at 5 percent peak height is 
satisfactory if it is not less than 0.8 and not more than 1.3 for the 
loracarbef peak.
    (B) Efficiency of the column. The absolute efficiency 
(hr) is satisfactory if it is not more than 20 for the 
loracarbef peak.
    (C) Resolution factor. The resolution factor (R) between the peak 
for loracarbef and the peak for the resolution standard loracarbef L-
isomer in the resolution test solution is satisfactory if it is not less 
than 6.0.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent of 5 replicate 
injections) is satisfactory if it is not more than 2.0 percent.
     (E) Capacity factor (k'). The capacity factor (k') for loracarbef 
is satisfactory if it is not less than 5 and not more than 8.

If the system suitability parameters have been met, then proceed as 
described in Sec. 436.216(b) of this chapter.
    (iv) Calculations. Calculate the micrograms of loracarbef per 
milligram of sample on an anhydrous basis as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.221

where:
AU=Area of the loracarbef peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
AS=Area of the loracarbef peak in the chromatogram of the 
          loracarbef working standard;
PS=Loracarbef activity in the loracarbef working standard 
          solution in micrograms per milliliter;
CU=Milligrams of sample per milliliter of sample solution; 
          and
m = Percent moisture content of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous suspension containing 100 milligrams per milliliter.
    (4) Specific rotation. Dissolve and dilute an accurately weighed 
sample with sufficient 0.1 N HCl to obtain a concentration of 
approximately 10 milligrams of loracarbef activity per milliliter. 
Proceed as directed in Sec. 436.210 of this chapter, using a 1.0-
decimeter polarimeter tube. Calculate the specific rotation on the 
anhydrous basis.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 1.0 percent potassium bromide disc prepared as described in 
Sec. 436.211(b)(1).



                      Subpart B--Oral Dosage Forms



Sec. 443.120  Loracarbef oral dosage forms.



Sec. 443.120a  Loracarbef capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Loracarbef capsules are composed of 
loracarbef and one or more suitable and harmless lubricants and diluents 
enclosed in a gelatin capsule. Each capsule contains loracarbef 
equivalent to either 200 milligrams or 400 milligrams of loracarbef. Its 
loracarbef content is satisfactory if it is not less than 90 percent and 
not more than 110 percent of the number of milligrams of loracarbef that 
it is represented to contain. Its moisture content is not more than 8.5 
percent. It passes the dissolution test. It passes the identity test. 
The loracarbef used

[[Page 698]]

conforms to the standards prescribed by Sec. 443.20(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The loracarbef used in making the batch for potency, moisture, 
pH, specific rotation, crystallinity, and identity.
    (B) The batch for content, moisture, dissolution, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The loracarbef used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch: A minimum of 100 capsules.
    (b) Tests and methods of assay--(1) Loracarbef content. Proceed as 
directed in Sec. 443.20(b)(1), preparing the sample solution and 
calculating the loracarbef content as follows:
    (i) Preparation of sample solution. Place one intact capsule in a 
200-milliliter volumetric flask containing 150 milliliters of distilled 
water. Shake the mixture vigorously to aid disruption of the capsule. 
Sonicate the mixture briefly (5 minutes). Dilute the contents to volume 
with distilled water. Mix well and immediately transfer a suitable 
aliquot to a volumetric flask of appropriate size to obtain a solution 
containing 0.2 milligram per milliliter (estimated) of loracarbef when 
diluted to volume with mobile phase (described in Sec. 443.20(b)(1)(i)). 
Filter this solution through a 0.45-micron membrane filter before 
injecting it into the chromatograph.
    (ii) Calculations. Calculate the loracarbef content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.222
    
where:
AU=Area of the loracarbef peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
AS=Area of the loracarbef peak in the chromatogram of the 
          loracarbef working standard;
PS=Loracarbef activity in the loracarbef working standard 
          solution in micrograms per milliliter; and
d = Dilution factor of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Dissolution test. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q, the amount of loracarbef activity dissolved, is 
75 percent within 30 minutes.
    (4) Identity. The retention time of the loracarbef response in the 
high-performance liquid chromatographic procedure described in paragraph 
(b)(1) of this section as applied to the sample solution compares 
qualitatively to that of the loracarbef reference standard.



Sec. 443.120b  Loracarbef for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Loracarbef for oral suspension is 
loracarbef with one or more suitable and harmless preservatives, 
sweeteners, suspending agents, colorings, antifoaming agents, and 
flavorings. When constituted as directed in the labeling, each 
milliliter contains the equivalent of either 20 or 40 milligrams 
loracarbef activity. Its loracarbef content is satisfactory if it is not 
less than 90 percent and not more than 115 percent of the number of 
milligrams of loracarbef that it is represented to contain. Its moisture 
content is not more than 2.0 percent. When constituted as described in 
the labeling, the pH of the suspension is not less than 3.5 and not more 
than 6.0. It passes the identity test. The loracarbef used conforms to 
the standards prescribed by Sec. 443.20(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The loracarbef used in making the batch for potency, moisture, 
pH, specific rotation, crystallinity, and identity.
    (B) The batch for content, moisture, pH, and identity.

[[Page 699]]

    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The loracarbef used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch: A minimum of 10 immediate containers.
    (b) Tests and methods of assay--(1) Loracarbef content. Proceed as 
directed in Sec. 443.20(b)(1), preparing the sample solution and 
calculating the loracarbef content as follows:
    (i) Preparation of sample solution. Constitute as directed in the 
labeling. Transfer a 5.0-milliliter portion of the suspension into an 
appropriately sized volumetric flask and quantitatively dilute stepwise 
with mobile phase (described in Sec. 443.20(b)(1)(i)) to obtain a 
concentration of 0.2 milligram of loracarbef activity per milliliter 
(estimated).
    (ii) Calculations. Calculate the loracarbef content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.223
    
where:
AU=Area of the loracarbef peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
AS=Area of the loracarbef peak in the chromatogram of the 
          loracarbef working standard;
PS=Loracarbef activity in the loracarbef working standard 
          solution in micrograms per milliliter; and
d = Dilution factor of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug constituted as directed in the labeling.
    (4) Identity. The retention time of the loracarbef response in the 
high-performance liquid chromatographic procedure described in paragraph 
(b)(1) of this section as applied to the sample solution compares 
qualitatively to that of the loracarbef reference standard.



PART 444--OLIGOSACCHARIDE ANTIBIOTIC DRUGS--Table of Contents




                          Subpart A--Bulk Drugs

Sec.
444.6  Amikacin.
444.7  Amikacin sulfate.
444.10a  Dihydrostreptomycin sulfate, crystalline dihydrostreptomycin 
          sulfate, dihydrostreptomycin hydrochloride.
444.20  Gentamicin sulfate.
444.20a  Sterile gentamicin sulfate.
444.30  Kanamycin sulfate.
444.30a  Sterile kanamycin sulfate.
444.42  Neomycin sulfate.
444.42a  Sterile neomycin sulfate.
444.46  Netilmicin sulfate.
444.50  Paromomycin sulfate.
444.62  Sisomicin sulfate.
444.70a  Sterile streptomycin sulfate.
444.80  Tobramycin.
444.81a  Sterile tobramycin sulfate.

                      Subpart B--Oral Dosage Forms

444.130  Kanamycin sulfate capsules.
444.142  Neomycin sulfate oral dosage forms.
444.142a  Neomycin sulfate tablets.
444.142b  Neomycin sulfate oral solution.
444.150  Paromomycin sulfate oral dosage forms.
444.150a  Paromomycin sulfate capsules.
444.150b  Paromomycin sulfate sirup.

                   Subpart C--Injectable Dosage Forms

444.206  Amikacin sulfate injection.
444.220  Gentamicin sulfate injection.
444.230  Kanamycin sulfate injection.
444.246  Netilmicin sulfate injection.
444.262  Sisomicin sulfate injection.
444.270  Streptomycin sulfate injectable dosage forms.
444.270a  Sterile streptomycin sulfate.
444.270b  Streptomycin sulfate injection.
444.280  Tobramycin sulfate injection.
444.281  Sterile tobramycin sulfate.

                   Subpart D--Ophthalmic Dosage Forms

444.320  Gentamicin sulfate ophthalmic dosage forms.
444.320a  Gentamicin sulfate ophthalmic solution.
444.320b  Gentamicin sulfate ophthalmic ointment.
444.320c  Gentamicin sulfate-prednisolone acetate ophthalmic suspension.
444.320d  Gentamicin sulfate-prednisolone acetate ophthalmic ointment.
444.342  Neomycin sulfate ophthalmic dosage forms.
444.342a  Neomycin sulfate- ------------ ophthalmic suspension; neomycin 
          sulfate- ------------ ophthalmic solution (the blanks being 
          filled in with the established name(s) of the other active 
          ingredient(s) present in accordance with paragraph (a)(1) of 
          this section).
444.342b  Neomycin sulfate-polymyxin B sulfate-gramicidin ophthalmic 
          solution.
444.342c  Neomycin sulfate- gramicidin

[[Page 700]]

          ------------ ophthalmic solution; neomycin sulfate-gramicidin 
          ------------ ophthalmic suspension (the blanks being filled in 
          with the established name(s) of the other ingredient(s) 
          present in accordance with paragraph (a)(1) of this section).
444.342d  Neomycin sulfate-polymyxin B sulfate ------------ ophthalmic 
          suspension (the blank being filled in with the established 
          name(s) of the other active ingredient(s) present in 
          accordance with paragraph (a)(1) of this section).
444.342e  Neomycin sulfate ointment; neomycin sulfate- ------------ 
          ointment (the blank being filled in with the established 
          name(s) of certain other active ingredient(s)).
444.342f  Neomycin sulfate-gramicidin topical ointment; neomycin 
          sulfate-gramicidin-triamcinolone acetonide ointment; neomycin 
          sulfate-gramicidin-fludrocortisone acetate ointment.
444.342g  Neomycin sulfate-hydrocortisone acetate ophthalmic suspension; 
          neomycin sulfate-prednisolone acetate ophthalmic suspension.
444.342h  Neomycin sulfate-polymyxin B sulfate ophthalmic ointment.
444.342i  Neomycin sulfate-polymyxin B sulfate ophthalmic solution.
444.342j  Neomycin sulfate-polymyxin B sulfate-dexamethasone ophthalmic 
          suspension.
444.342k  Neomycin sulfate-polymyxin B sulfate-dexamethasone ophthalmic 
          ointment.
444.380  Tobramycin ophthalmic dosage forms.
444.380a  Tobramycin ophthalmic solution.
444.380b  Tobramycin ophthalmic ointment.
444.380c  Tobramycin-dexamethasone ophthalmic suspension.
444.380d  Tobramycin-dexamethasone ophthalmic ointment.
444.380e  Tobramycin-fluorometholone acetate ophthalmic suspension.

                      Subpart E--Otic Dosage Forms

444.442  Neomycin sulfate otic dosage forms.
444.442a--444.442c  [Reserved]
444.442d  Neomycin sulfate ointment; neomycin sulfate- ------------ 
          ointment (the blank being filled in with the established 
          name(s) of certain other active ingredient(s)).
444.442e  [Reserved]
444.442f  Neomycin sulfate-hydrocortisone-acetic acid otic suspension.
444.442g  Neomycin sulfate-polymyxin B sulfate-hydrocortisone otic 
          suspension.
444.442h  Neomycin sulfate-polymyxin B sulfate-hydrocortisone otic 
          solution.

                  Subpart F--Dermatologic Dosage Forms

444.520  Gentamicin sulfate dermatologic dosage forms.
444.520a  Gentamicin sulfate ointment.
444.520b  Gentamicin sulfate cream.
444.540  Neomycin palmitate dermatologic dosage forms.
444.542  Neomycin sulfate dermatologic dosage forms.
444.542a  Neomycin sulfate ointment; neomycin sulfate- ------------ 
          ointment (the blank being filled in with the established 
          name(s) of the other active ingredient(s) present in 
          accordance with paragraph (a)(1) of this section).
444.542b  Neomycin sulfate cream; neomycin sulfate______ cream (the 
          blank being filled in with the established name(s) of the 
          other active ingredient(s) present in accordance with 
          paragraph (a)(1) of this section).
444.542c  Neomycin sulfate- ------------ lotion (the blank being filled 
          in with the established name(s) of the other active 
          ingredient(s) present in accordance with paragraph (a)(1) of 
          this section).
444.542d  [Reserved]
444.542e  Neomycin sulfate-polymyxin B sulfate ointment.
444.542f  Neomycin sulfate-gramicidin topical ointment; neomycin 
          sulfate-gramicidin-triamcinolone acetonide ointment; neomycin 
          sulfate-gramicidin-fludrocortisone acetate ointment.
444.542g  Neomycin sulfate-gramicidin-triamcinolone acetonide cream.
444.542h  Neomycin sulfate-gramicidin-tri-amcinolone acetonide lotion; 
          neomycin sulfate-gramicidin-fludrocortisone acetate lotion.
444.542i  [Reserved]
444.542j  Neomycin sulfate-polymyxin B sulfate-gramicidin-benzocaine 
          ointment.
444.542k  Neomycin sulfate-polymyxin B sulfate-hydrocortisone acetate 
          cream.
444.542l  Neomycin sulfate-polymyxin B sulfate cream.

                        Subparts G-I  [Reserved]

                  Subpart J--Certain Other Dosage Forms

444.942  Neomycin sulfate in certain other dosage forms.
444.942a  Neomycin sulfate for compounding oral products.
444.942b  Sterile neomycin sulfate and polymyxin B sulfate solution.

    Authority:  21 U.S.C. 357.

    Source:  39 FR 19046, May 30, 1974, unless otherwise noted.

[[Page 701]]



                          Subpart A--Bulk Drugs



Sec. 444.6  Amikacin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amikacin is A-3-amino-3-deoxy-A-D-
glucopyranosyl (1-6) - A - [6 - amino - 6 - deoxy - A - D - 
glucopyranosyl (1-4)] - N 1 - [(s) - 4 - amino - 2 - hydroxy 
- 1 - oxobutyl] - 2 - deoxy - D - streptamine. It is so purified and 
dried that:
    (i) Its potency is not less than 900 micrograms per milligram on an 
anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 8.5 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 9.5 and not more than 11.5.
    (v) It gives a positive identity test for amikacin.
    (vi) Its residue on ignition is not more than 1.0 percent.
    (vii) Its specific rotation is not less than +97 deg. and not more 
than +105 deg..
    (viii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, safety, 
moisture, pH, identity, residue on ignition, specific rotation, and 
crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient sterile distilled 
water to obtain a stock solution of convenient concentration. Further 
dilute an aliquot of the stock solution with distilled water to the 
reference concentration of 10.0 micrograms of amikacin per milliliter 
(estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Identity. Proceed as directed in Sec. 436.318 of this chapter.
    (6) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (7) Specific rotation. Proceed as directed in Sec. 436.210 of this 
chapter, using an aqueous solution containing 20 milligrams of amikacin 
per milliliter and a 1.0-decimeter polarimeter tube. Calculate the 
specific rotation on an anhydrous basis.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[41 FR 49483, Nov. 9, 1976, as amended at 44 FR 10379, Feb. 20, 1979; 50 
FR 19919, May 13, 1985]



Sec. 444.7  Amikacin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amikacin sulfate is the sulfate salt of 
D-streptamine, 0-3-amino-3-deoxy--D-glucopyranosyl(1-6)-0-[6-
amino-6-deoxy--D-glucopyranosyl(1-4)]-N\1\-(4-amino-2-hydroxy-
1-oxobutyl)-2-deoxy-,(S)-. It is so purified and dried that:
    (i) Its potency is not less than 674 micrograms and not more than 
786 micrograms per milligram on an anhydrous basis if the molar ratio of 
amikacin to sulfuric acid (H2SO4) is 1:2 and is 
not less than 691 micrograms and not more than 806 micrograms per 
milligram on an anhydrous basis if the molar ratio of amikacin to 
H2SO4 is 1:1.8.
    (ii) Its loss on drying is not more than 13.0 percent.
    (iii) The pH of an aqueous solution containing 10 milligrams of 
amikacin sulfate per milliliter is not less than 2.0 and not more than 
4.0 if the molar ratio of amikacin to H2SO4 is 1:2 
and not less than 6.0 and not more than 7.3 if the molar ratio of 
amikacin to H2SO4 is 1:1.8.
    (iv) It gives a positive identify test for amikacin.
    (v) Its residue on ignition is not more than 1.0 percent.
    (vi) Its specific rotation is not less than +76 deg. and not more 
than +84 deg. on the anhydrous basis.
    (vii) It is crystalline.

[[Page 702]]

    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, identity, residue on ignition, specific rotation, and 
crystallinity.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research: 10 packages, each containing approximately 500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using a 25-centimeter by 4.6-millimeter 
column packed with irregular 5-micron octadecyl hydrocarbon bonded 
silica, thermostatted at 30  deg.C, an ultraviolet detection system 
operating at a wavelength of 340 nanometers, a flow rate not exceeding 
2.0 milliliters per minute, a chart speed of 1.0 centimeter per minute 
(the chart speed is increased to 5.0 centimeters per minute to obtain 
chromatograms used for performance parameter determinations), and a 
known injection volume between 15.0 and 30.0 microliters. Retention 
times of amikacin and kanamycin are about 10 and 15 minutes, 
respectively. Reagents, working standard solution, sample solution, 
resolution test solution, system suitability requirements, and 
calculations are as follows:
    (i) Reagents--(A) 1.0 percent 2,4,6-trinitrobenzenesulphonic acid 
solution. Dissolve 1.0 gram of 2,4,6-trinitrobenzenesulphonic acid in 
100 milliliters of distilled water.
    (B) 0.02M potassium dihydrogen phosphate. Dissolve 2.72 grams of 
potassium dihydrogen phosphate in 800 milliliters of distilled water and 
mix to dissolve the solid. Dilute to 1,000 milliliters with distilled 
water and mix.
    (C) Mobile phase. Mix 0.02M potassium dihydrogen phosphate and 
methanol, high performance liquid chromatography reagent grade (28:72 by 
volume). Adjust the pH to 6.5 with 0.4M potassium hydroxide. Filter the 
mobile phase through a suitable glass filter or equivalent which is 
capable of removing particulate matter contamination greater than 0.5 
micron in diameter. Degas the mobile phase just prior to its 
introduction into the chromatograph.
    (ii) Preparation of working standard and sample solutions. (A) 
Working standard solution. Dissolve an accurately weighed portion of the 
amikacin working standard with sufficient distilled water to obtain a 
solution containing approximately 1.0 milligram of amikacin activity per 
milliliter. This preparation is stable for 1 week. Transfer 50 
microliters of this solution directly to the bottom of a 50-milliliter, 
glass-stoppered centrifuge tube, using an automatic micropipetter. Add 
3.2 milliliters of pyridine and 2.0 milliliters of 1 percent 2,4,6-
trinitrobenzenesulphonic acid reagent just above the surface of the 
solution in the centrifuge tube. Close the tube tightly, mix and heat 
the tube in a water bath maintained at 75  
deg.Cplus-minus1 deg. for 45 minutes. Remove the tube from 
the bath and cool it at room temperature. Filter the contents through a 
0.5 micron membrane. Use the filtrate for the quantitative 
chromatographic determinations.
    (B) Preparation of sample solution. Dissolve an accurately weighed 
portion of sample with sufficient distilled water to obtain a solution 
containing 1.0 milligram of amikacin activity per milliliter 
(estimated). This preparation is stable for 1 week. Proceed as directed 
in paragraph (b)(1)(ii)(A) of this section, beginning at ``Transfer 50 
microliters * * *''.
    (C) Resolution test solution. Prepare an aqueous solution containing 
about 1.0 milligram per milliliter each of amikacin and kanamycin. 
Proceed as directed in paragraph (b)(1)(ii)(A) of this section, 
beginning at ``Transfer 50 microliters * * *''.
    (iii) System suitability requirements--(A) Asymmetry factor. The 
asymmetry factor (As) of the amikacin peak is satisfactory if 
it is not more than 1.3 at 10 percent of peak height.
    (B) Efficiency of the column. The absolute efficiency 
(hr) is satisfactory if it is not more than 20.0 for the 
amikacin peak.
    (C) Resolution. The resolution (R) between the amikacin peak and the 
kanamycin peak is satisfactory if it is not less than 5.0.

[[Page 703]]

    (D) Coefficent of variation (relative standard deviation). The 
coefficient of variation (Srin percent) of five replicate 
injections is satisfactory if it is not more than 2.0 percent. If the 
system suitability parameters have been met, then proceed as described 
in Sec. 436.216(b) of this chapter.
    (iv) Calculations. Calculate the micrograms of amikacin per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.224

where:
Au=Area of the amikacin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the amikcacin peak in the chromatogram of the 
          amikacin working standard;
Ps=Amikacin activity in the amikacin working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of the sample per milliliter of sample 
          solution; and
m=Percent loss on drying of the sample.

    (2) Loss on drying. Proceed as directed in Sec. 436.200(c) of this 
chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (4) Identity. Proceed as directed in Sec. 436.318 of this chapter.
    (5) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (6) Specific rotation. Proceed as directed in Sec. 436.210 of this 
chapter, using an aqueous solution containing 20 milligrams of amikacin 
sulfate per milliliter, and a 1.0 decimeter polarimeter tube. Calculate 
the specific rotation on the anhydrous basis.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[55 FR 38676, Sept. 20, 1990]



Sec. 444.10a  Dihydrostreptomycin sulfate, crystalline dihydrostreptomycin sulfate, dihydrostreptomycin hydrochloride.

    (a) Requirements for certification--(1) Dihydrostreptomycin sulfate 
is the hydrogenated sulfate salt of a kind of streptomycin or a mixture 
of two or more such salts; crystalline dihydrostreptomycin sulfate is 
the hydrogenated crystalline sulfate salt of a kind of streptomycin or a 
mixture of two or more such salts; dihydrostreptomycin hydrochloride is 
the hydrogenated hydrochloride salt of a kind of streptomycin or a 
mixture of two or more such salts. Each such drug conforms to all 
requirements prescribed by Sec. 444.70a(a) for streptomycin sulfate and 
streptomycin hydrochloride, and is subject to all procedures prescribed 
by Sec. 444.70a(a) for streptomycin sulfate and streptomycin 
hydrochloride, except that:
    (i) Its potency is not less than 650 micrograms per milligram, 
except that if it is crystalline dihydrostreptomycin sulfate its potency 
is not less than 725 micrograms per milligram.
    (ii) Its content of streptomycin sulfate or streptomycin 
hydrochloride is not more than 3.0 percent when calculated as 
streptomycin base, except that if it is crystalline dihydrostreptomycin 
sulfate its content of streptomycin sulfate is not more than 1.0 
percent.
    (iii) Its labeling shall conform to the requirements of 
Sec. 444.70a(a)(3)(iii).
    (b) Tests and methods of assay--(1) Potency. Using the 
dihydrostreptomycin working standard as a standard of comparison, 
proceed as directed in Sec. 444.70a(b)(1). Its potency is satisfactory 
if it contains not less than 90 percent of the number of milligrams that 
it is represented to contain.
    (2) Content of streptomycin sulfate or streptomycin hydrochloride--
(i) Reagents. (a) 10 percent ferric chloride stock solution. Dissolve 5 
grams of FeCl36H2O in 50 milliliters 0.1N 
HCl.
    (b) 0.25 percent ferric chloride solution. Dilute 2.5 milliliters of 
10 percent ferric chloride in 0.1N HCl to 100 milliliters with 0.01N 
MCl. Prepare the solution fresh daily.
    (ii) Standard curve. Keep the working standard (obtained from the 
Food and Drug Administration) at -20 deg. C. in tightly stoppered 
containers which in turn are kept in larger stoppered vials containing a 
suitable desiccant. Dry an appropriate amount of the working standard at 
100 deg. C. and a pressure of 5 millimeters or less for 4 hours. Prepare 
a stock aqueous solution containing 1.0 milligram of streptomycin base 
per milliliter. Store this standard solution

[[Page 704]]

in a refrigerator and use for no longer than 2 weeks. Transfer 1.0, 2.0, 
3.0, 4.0, and 5.0 milliliters of this standard solution and 10 
milliliters of distilled water to each of six 25-milliliter volumetric 
flasks. Add 9.0, 8.0, 7.0, 6.0, and 5.0 milliliters of distilled water 
to the five tubes, respectively, to give each a total volume of 10 
milliliters. To each add 2.0 milliliters of 1N NaOH and then heat the 
flasks in a boiling water bath for 10 minutes. Cool the flasks in ice 
water for 3 minutes and acidify the solutions with 2.0 milliliters of 
1.2N HCl. To each flask add 5.0 milliliters of 0.25 percent ferric 
chloride reagent, make to volume with distilled water, and mix 
thoroughly. Transfer the colored solutions to 2.0-centimeter absorption 
cells and measure the percent light transmission at 530 m in a 
suitable photoelectric colorimeter. Set the colorimeter at 100 percent 
light transmission for the zero concentration and then obtain the 
percent light transmission of the sample. Prepare a standard curve on 
semilog paper, plotting the percent light transmission on the 
logarithmic ordinate scale and the concentration of streptomycin base on 
the abscissa.
    (iii) Procedure. Dilute the contents of a vial or a sufficient 
amount of bulk material to give a concentration of approximately 20 
milligrams per milliliter. From the amount of streptomycin obtained, 
calculate the percent streptomycin as follows:

Percent streptomycin=(Milligrams of streptomycin x 100)/(Milligrams of 
          dihydrostreptomycin found in the sample used)

    (3) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (4) Toxicity, pyrogens, histamine, moisture, pH, crystallinity. 
Proceed as directed in Secs. 444.70a(b) (3), (4), (5), (6) and 
440.80a(b)(5)(iii) of this chapter.



Sec. 444.20  Gentamicin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Gentamicin sulfate is the sulfate salt of 
a kind of gentamicin or a mixture of two or more such salts. It is a 
powder, white to buff in color. It is readily soluble in water but 
insoluble in ethanol. It is so purified and dried that:
    (i) Its potency is not less than 590 micrograms of gentamicin per 
milligram on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 18.0 percent.
    (iv) Its pH in an aqueous solution containing 40 milligrams per 
milliliter is not less than 3.5 and not more than 5.5.
    (v) Its specific rotation in an aqueous solution containing 10 
milligrams per milliliter at 25 deg.C. is not less than +107 deg. and 
not more than +121 deg..
    (vi) Its content of gentamicin C1 is not less than 25 nor 
more than 50 percent; of gentamicin C1a, not less than 15 nor 
more than 40 percent; and of gentamicin C2, not less than 20 
nor more than 50 percent.
    (vii) It gives a positive identity test for gentamicin sulfate.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, specific rotation, content of gentamicins C1, 
C1a, and C2, and identity.
    (ii) Samples required. 10 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), to give a stock solution of 
convenient concentration. Further dilute the stock solution with 
solution 3 to the reference concentration of 0.1 microgram of gentamicin 
per milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(c) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 40 milligrams of gentamicin per 
milliliter.

[[Page 705]]

    (5) Specific rotation. Accurately weigh the sample to be tested in a 
volumetric flask and dilute with sufficient distilled water to give a 
solution containing approximately 10 milligrams per milliliter. Proceed 
as directed in Sec. 436.210 of this chapter, using a 1.0-decimeter 
polarimeter tube and calculate the specific rotation on an anhydrous 
basis.
    (6) Content of gentamicins C1, C1a, and 
C2. Proceed as directed in Sec. 444.20a(b)(8).
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 0.5 percent mixture of the sample in a potassium bromide disc 
prepared as described in paragraph (b)(1) of that section.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.20a  Sterile gentamicin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile gentamicin sulfate is the sulfate 
salt of a kind of gentamicin or a mixture of two or more such salts. It 
is a powder, white to buff in color. It is readily soluble in water but 
insoluble in ethanol. It is so purified and dried that:
    (i) Its potency is not less than 590 micrograms of gentamicin per 
milligram on an anhydrous basis.
    (ii) It is sterile.
    (iii) [Reserved]
    (iv) It is nonpyrogenic.
    (v) Its loss on drying is not more than 18.0 percent.
    (vi) Its pH in an aqueous solution containing 40 milligrams per 
milliliter is not less than 3.5 and not more than 5.5.
    (vii) Its specific rotation in an aqueous solution containing 10 
milligrams per milliliter at 25 deg. C. is not less than +107 deg. and 
not more than +121 deg..
    (viii) Its content of gentamicin C1 is not less than 25 
nor more than 50 percent; of gentamicin C1a, not less than 15 
nor more than 40 percent; and of gentamicin C2, not less than 
20 nor more than 50 percent.
    (ix) It gives a positive identity test for gentamicin sulfate.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, pH, specific rotation, content of gentamicins 
C1, C1a, and C2, and identity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), to give a stock solution of 
convenient concentration. Further dilute the stock solution with 
sufficient solution 3 to give a reference concentration of 0.1 microgram 
of gentamicin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) [Reserved]
    (4) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 10.0 milligrams of gentamicin per 
milliliter.
    (5) Loss on drying. Proceed as directed in Sec. 436.200(c) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 40 milligrams of gentamicin per 
milliliter.
    (7) Specific rotation. Accurately weigh the sample to be tested in a 
volumetric flask and dilute with sufficient distilled water to give a 
solution containing approximately 10 milligrams per milliliter. Proceed 
as directed in Sec. 436.210 of this chapter, using a 1-decimeter 
polarimeter tube and calculate the specific rotation on an anhydrous 
basis.
    (8) Content of gentamicins C1, C1a, and 
C2-(i) Equipment-(a) Chamber (chromatographic). Use a 
suitable chromatography jar with a tightly fitting, ground glass contact 
top for descending chromatography.
    (b) Sheets (chromatographic). Cut a 57  x  46-centimeter sheet of 
Whatman No. 2

[[Page 706]]

filter paper, or chromatographic paper that will produce similar 
results, into four strips of about 14.25  x  46 centimeters. Draw a 
starting line 9 centimeters from one end and mark two dots on this line, 
each 4 centimeters from each edge.
    (ii) Reagents. Use reagent grade solvents and chemicals.
    (iii) Solvent system. In each of two separators, equilibrate 200 
milliliters of chloroform and 100 milliliters of methanol with 100 
milliliters of 17 percent (9 molar) ammonium hydroxide. Without allowing 
the phases of one to separate, add the entire mixture to the 
chromatography jar and allow 24 hours for saturation. Allow the second 
separator to stand until the phases separate and use the lower phase 
only as the chromatographic solvent.
    (iv) Ninhydrin reagent. To 1 gram of ninhydrin and 0.1 gram of 
cadmium acetate, add 3 milliliters of water and 1.5 milliliters of 
gaacial acetic acid and shake. Add 100 milliliters of n-propanol and 
shake until solution is complete. Keep this solution in a brown bottle 
under refrigeration.
    (v) Procedure. Prepare an aqueous solution containing 40 milligrams 
of the sample per milliliter. Apply 5 microliters of this solution to 
each dot on the sheet. Prepare two such sheets and place them in the 
tank so that elution will take place from separate troughs. Fill the two 
troughs with the chromatographic solvent. Develop the sheets in a 
descending manner until the solvent front reaches the bottom of the 
paper (approximately 3\1/2\ hours at 25 deg. C.). Remove the sheets and 
dry in a hood for 30 minutes. Cut each sheet in half, lengthwise. Spray 
one half with ninhydrin reagent and place the sprayed strip in a drying 
oven at 100 deg. C. for 1 minute. The gentamicin fractions appear as 
reddish zones. The zone furthest from the origin is gentamicin 
C1, the one closest is gentamicin C1a, and the 
middle zone is gentamicin C2. Cut the corresponding zones out 
of the other unsprayed half of the sheet. Cut each portion of the sheet 
thus obtianed into small strips and put those from each zone into a 
separate 125-milliliter glass-stoppered flask. Add 50 milliliters of 
0.1M potassium phosphate buffer, pH 8, to each flask and swirl the flask 
mechanically for 30 minutes. Decant the solution from each flask into 
separate test tubes and allow the paper to settle. Pipet 4 milliliters 
of each clear solution into a 25-milliliter volumetric flask and make to 
volume with the pH 8 buffer. Assay these solutions as directed in 
paragraph (b)(1) of this section.
    (vi) Calculations.
    [GRAPHIC] [TIFF OMITTED] TC01AP94.060
    
where:

The assays are expressed in terms of the microgram equivalents of 
          gentamicin; and
The factors 0.786, 1.023, and 0.977 represent the activities of 
          gentamicins C1, C2, and C1a 
          relative to the gentamicin activity of the gentamicin master 
          standard.


[[Page 707]]


    (9) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 0.5 percent mixture of the sample in a potassium bromide disc 
prepared as described in paragraph (b)(2) of that section.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.30  Kanamycin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Kanamycin sulfate is the sulfate salt of 
a kind of kanamycin or a mixture of two or more such salts. It is so 
purified and dried that:
    (i) Its potency on an anhydrous basis is not less than 750 
micrograms of kanamycin per milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 4 percent.
    (iv) Its pH is an aqueous solution containing 10 milligrams per 
milliliter is not less than 6.5 and not more than 8.5
    (v) Its residue on ignition is not more than 1.0 percent.
    (vi) It gives a positive identity test for kanamycin.
    (vii) It contains not more than 5.0 percent kanamycin B.
    (viii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, residue on ignition, identity, kanamycin B content, and 
crystallinity.
    (ii) Samples required on the batch: 10 packages, each containing 
approximately 500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient sterile distilled 
water to give a stock solution of convenient concentration. Further 
dilute an aliquot of the stock solution with sterile distilled water to 
the reference concentration of 10 micrograms of kanamycin per milliliter 
(estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 10 milligrams per milliliter.
    (5) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (6) Identity. Dissolve about 10 milligrams of kanamycin sulfate in 1 
milliliter of water, and add 1 milliliter of a 1:500 solution of 
triketohydrindene hydrate in normal butyl alcohol; then add 0.5 
milliliter of pyridine. Heat in a steam bath for 5 minutes and add 10 
milliliters of water; a deep-purple color is produced.
    (7) Kanamycin B content--(i) Cylinders (cups). Use cylinders 
described under Sec. 440.80a(b)(1)(i) of this chapter.
    (ii) Culture medium. Use ingredients that conform to the standards 
prescribed by the U.S.P. or N.F. Make agar for the base and seed layers 
as follows:

Peptone..........................................................6.0 gm.
Yeast extract....................................................3.0 gm.
Beef extract.....................................................1.5 gm.
Agar............................................................15.0 gm.
pH 7.8 to 8.0 after sterilization.
Distilled water, q.s........................................1,000.00 ml.

    (iii) Working standard. Dissolve a suitable quantity of the 
kanamycin sulfate working standard, accurately weighed, in 0.1M 
potassium phosphate buffer, pH 8.0, to give a concentration equivalent 
to 1.0 milligram of kanamycin per milliliter.
    (iv) Preparation of sample. To 100 milligrams, accurately weighed, 
of kanamycin sulfate in a suitable container (such as a 7.5-milliliter 
serum vial) add 5.0 milliliters of 6N hydrochloric acid, and tightly 
close the container. Heat in a water bath at 100 deg. C. for 1 hour and 
cool. Add 4 milliliters of 6N sodium hydroxide, then dilute with sterile 
0.1M potassium phosphate buffer, pH 8.0, to obtain a concentration of 
the equivalent of 1 microgram of kanamycin per milliliter (estimated).

[[Page 708]]

    (v) Preparation of test organism. Use Bacillus subtilis (ATCC 6633) 
\1\ prepared as described in Sec. 436.103 of this chapter, using method 
2.
---------------------------------------------------------------------------

    \1\ Available from: American Type Culture Collection, 12301 Parklawn 
Drive, Rockville, MD 20852.
---------------------------------------------------------------------------

    (vi) Preparation of plates. Add 21 milliliters of the agar prepared 
as described in paragraph (b)(7) of this section to each Petri dish (20 
millimeters  x  100 millimeters). Distribute the agar evenly in the 
plates and allow to harden. Use the plates the same day they are 
prepared. Add 4.0 milliliters of the fresh daily inoculum described in 
paragraph (b)(7)(iv) of this section to each plate, tilting the plates 
back and forth to spread the inoculated agar evenly over the surface.
    (vii) Standard curve. Prepare on the day of testing in 0.1M 
potassium phosphate buffer, pH 7.8 to 8.0, from the standard stock 
solution, sufficient volumes of the following concentrations: 0.64, 0.8, 
1.0, 1.25, and 1.56 micrograms per milliliter. The 1.0 microgram-per-
milliliter solution is the reference point of the standard curve. On 
each of three plates fill three cylinders with the 1.0 microgram-per-
milliliter standard and the other three cylinders with the concentration 
under test. Thus, there will be thirty-six 1.0-microgram determinations 
for each of the other points on the curve. After the plates have 
incubated read the diameters of the circles of inhibition. Average the 
readings of the 1.0 microgram-per-milliliter concentration and the 
readings of the concentration test for each set of three plates and 
average also all 36 readings of the 1.0 microgram-per-milliliter 
concentration. The average of the 36 readings of the 1.0 microgram-per-
milliliter concentration is the correction point for the curve. Correct 
the average value obtained for each point to the figure it would be if 
the 1.0 microgram-per-milliliter reading for that set of three plates 
were the same as the correction point. Thus, if in correcting the 0.8-
microgram concentration, the average of the 36 readings of the 1.0 
microgram-per-milliliter concentration is 16.5 millimeters and the 
average of the 1.0 microgram-per-milliliter concentration of this set of 
three plates is 16.3 millimeters, the correction is +0.2 millimeter. If 
the average readings of the 0.8 microgram-per-milliliter concentration 
of these same three plates is 15.9 millimeters, the corrected value is 
16.1 millimeters. Plot these corrected values, including the average of 
the 1.0 microgram-per-milliliter concentration, on 2-cycle 
semilogarithmic paper, using the concentration in micrograms per 
milliliter as the ordinate and the diameter of the zone of inhibition as 
the abscissa. Draw the standard curve through these points, either by 
inspection or by means of the following equations:
[GRAPHIC] [TIFF OMITTED] TR01JA93.225

where:
L=Calculated zone diameter for the lowest concentration of the standard 
curve;
H=Calculated zone diameter for the highest concentration of the standard 
curve;
c=Average zone diameter of 36 readings of the 1.0 microgram-per-
milliliter standard;
a, b, d, e=Corrected average values for the 0.64, 0.8, 1.0, 1.25, and 
1.56 micrograms-per-milliliter solutions, respectively.


Plot the values obtained for L and H and connect with a straight line.
    (viii) Assay. Place six cylinders on the inoculated agar surface in 
each Petri dish prepared as described in paragraph (b)(7)(vi) of this 
section, so that they are at approximately 60 deg. intervals on a 2.8-
centimeter radius. Use three plates for each sample. Fill three 
cylinders on each plate with the 1.0 microgram-per-milliliter standard 
and three cylinders with the 1.0 microgram (estimated)-per-milliliter 
sample, alternating standard and sample. Incubate plates for 16 hours to 
18 hours at 32 deg. C. to 35 deg. C., and measure the diameter of each 
circle of inhibition.
    (ix) Estimation of kanamycin B content. Average the zone readings of 
the standard and average the zone readings of the sample on the three 
plates used. If the sample gives larger average zone size than the 
average of the standard, add the difference between them to the

[[Page 709]]

1.0-microgram zone size of the standard curve. If the average value is 
lower than the standard value, subtract the difference between them from 
the 1.0-microgram value on the curve. From the curve, read the kanamycin 
potencies corresponding to these corrected values of zone sizes. 
Multiply the observed potency by 100 and divide by 126 to obtain a value 
representing the potency in terms of the milligram equivalent of 
kanamycin B. The calculated amount of kanamycin B is not more than 5 
percent of the content of kanamycin found in paragraph (b)(1) of this 
section.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.30a  Sterile kanamycin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Kanamycin sulfate is the sulfate salt of 
a kind of kanamycin or a mixture of two or more such salts. It is so 
purified and dried that:
    (i) Its potency on an anhydrous basis is not less than 750 
micrograms of kanamycin per milligram.
    (ii) It is sterile.
    (iii) [Reserved]
    (iv) It is nonpyrogenic.
    (v) Its loss on drying is not more than 4 percent.
    (vi) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 6.5 and not more than 8.5.
    (vii) Its residue on ignition is not more than 1.0 percent.
    (viii) It gives a positive identity test for kanamycin.
    (ix) It contains not more than 5.0 percent kanamycin B.
    (x) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, pH, residue on ignition, identity, 
crystallinity, and kanamycin B content.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient sterile distilled 
water to give a stock solution of convenient concentration. Further 
dilute an aliquot of the stock solution with sterile distilled water to 
the reference concentration of 10 micrograms of kanamycin per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) [Reserved]
    (4) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 10 milligrams of kanamycin per milliliter.
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 10 milligrams per milliliter.
    (7) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (8) Identity. Dissolve about 10 milligrams of kanamycin sulfate in 1 
milliliter of water and add 1 milliliter of a 1:500 solution of 
triketohydrindene hydrate in normal butyl alcohol. Then add 0.5 
milliliter of pyridine. Heat in a steam bath for 5 minutes and add 10 
milliliters of water; a deep-purple color is produced.
    (9) Kanamycin B content. Proceed as directed in Sec. 444.30(b)(7).
    (10) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.42  Neomycin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate is the sulfate salt of a 
kind of neomycin or a mixture of two or more such salts. It is so 
purified and dried that:

[[Page 710]]

    (i) Its potency is not less than 600 micrograms of neomycin per 
milligram, calculated on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 8.0 percent.
    (iv) Its pH in an aqueous solution containing 33 milligrams per 
milliliter is not less than 5.0 and not more than 7.5.
    (v) It gives a positive identity test for neomycin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, and identity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), to give a stock solution of 
convenient concentration. Further dilute an aliquot of the stock 
solution with solution 3 to the reference concentration of 1.0 microgram 
of neomycin per milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 33 milligrams of neomycin per milliliter.
    (5) Identity--(i) Reagents. (a) Sulfuric acid solution: Mix 
concentrated sulfuric acid and distilled water in volumetric proportions 
of 40:60.
    (b) Xylene.
    (c) p-Bromoaniline: (Prepare and store this reagent in brown, 
nonactinic glassware.) Place 380 milliliters of thiourea-saturated 
glacial acetic acid solution in the bottle, add 10 milliliters of 20 
percent sodium chloride solution, 5 milliliters of 5 percent oxalic acid 
solution, and 5 milliliters of 10 percent disodium phosphate solution, 
and mix well. Add 8 grams of p-bromoaniline and mix well. Let this 
reagent stand overnight before use. Prepare the reagent once weekly.
    (ii) Procedure. Place about 10 milligrams of the sample into a test 
tube (19 millimeters  x  150 millimeters), dissolve with 1 milliliter of 
water, and then carefully add 5 milliliters of the sulfuric acid 
solution. Heat in a boiling water bath for 100 minutes. Cool to room 
temperature. Add 10 milliliters of xylene to the test tube. Stopper the 
tube and shake vigorously for about 1 minute. Let the two layers 
separate and then decant the xylene layer into a second test tube. Add 
10 milliliters of the p-bromoaniline reagent to the xylene solution, 
shake, and let stand. The development of a vivid pink-red color is a 
positive identity test for neomycin.
[40 FR 22252, May 22, 1975, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.42a  Sterile neomycin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate is the sulfate salt of a 
kind of neomycin or a mixture of two or more such salts. It is so 
purified and dried that:
    (i) It has a potency of not less than 600 micrograms of neomycin per 
milligram, calculated on an anhydrous basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 8.0 percent.
    (vi) Its pH in an aqueous solution containing 33 milligrams per 
milliliter is not less than 5.0 and not more than 7.5.
    (vii) It gives a positive identity test for neomycin.
    (2) Labeling. It is to be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Request for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, and identity.
    (ii) Samples required;

[[Page 711]]

    (a) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods:
    (i) Plate assay using Staphylococcus epidemmidis (ATCC 12228) \1\--
(a) Cylinders (cups). Use cylinders described in Sec. 440.80a(b)(1)(i) 
of this chapter.
---------------------------------------------------------------------------

    \1\ Available from: American Type Culture Collection, 12301 Parklawn 
Drive, Rockville, MD 20852.
---------------------------------------------------------------------------

    (b) Culture media. Using ingredients that conform to the standards 
prescribed by the U.S.P. or N.F.:
    (1) Make nutrient agar for carrying the test organism as follows:

Peptone..........................................................6.0 gm.
Pancreatic digest of casein......................................4.0 gm.
Yeast extract....................................................3.0 gm.
Beef extract.....................................................1.5 gm.
Dextrose.........................................................1.0 gm.
Agar............................................................15.0 gm.
Distilled water q.s..........................................1,000.0 ml.
pH 6.5 to 6.6 after sterilization.

    (2) Make nutrient agar for the base and seed layers as described in 
paragraph (b)(1)(i)(b)(1) of this section, except that its pH after 
sterilization is 7.8 to 8.0.

In lieu of preparing the media from the individual ingredients specified 
in paragraph (b)(1)(i)(b) of this section, they may be made from a 
dehydrated mixture that, when reconstituted with distilled water, has 
the same composition as such media. Minor modification of the individual 
ingredients specified in paragraph (b)(1)(i)(b) of this section are 
permissible if the resulting media possess growth-promoting properties 
at least equal to the media described.
    (c) Working standard. Dry a portion of the working standard for 3 
hours at 60 deg. C. and a pressure of 5 millimeters or less. Determine 
the dry weight, and dissolve in sufficient 0.1M potassium phosphate 
buffer, pH 8.0, to give a stock solution of convenient concentration. 
When stored under refrigeration, the stock solution may be used for a 
period not exceeding 2 weeks.
    (d) Preparation of sample. Dissolve an accurately weighed sample in 
sufficient 0.1M potassium phosphate buffer, pH 8.0 (solution 3), to give 
a stock solution of convenient concentration. Further dilute the stock 
solution with sufficient solution 3 to obtain a reference concentration 
of 1.0 microgram of neomycin per milliliter (estimated).
    (e) Preparation of test organism. The test organism is 
Staphylococcus epidermidis (ATCC 12228), 1 which is 
maintained on slants of nutrient agar described in (b)(1)(i)(b)(1) of 
this section. Using 3 milliliters of U.S.P. saline T.S., wash the 
organism from the nutrient agar slant (which has been incubated for 24 
hours at 32 deg. C.-35 deg. C.) onto a large nutrient agar surface such 
as that provided by a Roux bottle containing 300 milliliters of nutrient 
agar. Incubate for 24 hours at 32 deg. C.-35 deg. C. Wash the resulting 
growth from the nutrient surface, using 50 milliliters of sterile U.S.P. 
saline T.S. Adjust the volume of the suspension so that a 1:14 dilution 
will give 25 percent light transmission when measured with a suitable 
photo-electric colorimeter having a 580 m filter and a 13-
millimeter diameter test tube as an absorption cell. By the use of test 
plates, determine the appropriate inoculum of the adjusted suspension 
(usually 0.1 milliliter) to be inoculated to each 100 milliliters of 
seed layer agar in order to obtain satisfactory zones of inhibition. The 
suspension may be used for 1 week if stored under refrigeration.
    (f) Preparation of plates. Add 21 milliliters of the agar prepared 
as described in paragraph (b)(1)(i)(b)(2) of this section to each Petri 
dish (20 millimeters  x  100 millimeters). Distribute the agar evenly in 
the plates and allow to harden on a level surface. Accurately measure a 
sufficient quantity of the nutrient agar, cool to 48 deg. C., and add 
the appropriate inoculum of the adjusted suspension, prepared as 
described in paragraph (b)(1)(i)(e) of this section. Swirl the 
inoculated nutrient agar to obtain a homogeneous suspension, and add 4 
milliliters to each of the plates containing the 21 milliliters of 
uninoculated nutrient agar. Tilt the plates back and forth to spread the 
inoculated nutrient agar evenly, and allow to harden on a level surface. 
After the agar has hardened, place six cylinders described in paragraph

[[Page 712]]

(b)(1)(i)(a) of this section on the inoculated agar surface so that they 
are at approximately 60 deg. intervals on a 2.8-centimeter radius. Use 
the plates the same day they are prepared.
    (g) Standard curve. Using the stock solution of the working standard 
prepared as described in paragraph (b)(1)(i)(c) of this section, prepare 
solutions in 0.1M potassium phosphate buffer pH 8.0 of the following 
concentrations: 0.64, 0.8, 1.0, 1.25, 1.56 micrograms of neomycin per 
milliliter. The 1.0 microgram per milliliter concentration is the 
reference concentration of the assay. Use a total of 12 plates, three 
plates for each solution except the reference point solution which is 
included on each plate. On each of the three plates, fill three 
cylinders with the reference point solution and the other three 
cylinders with the concentrations under test. Thus, there will be 36 
reference point determinations and nine determinations for each of the 
other points on the curve. After the plates have incubated, read the 
diameters of the circles of inhibition. Average the readings of the 
reference point concentration and the readings of the point tested for 
each set of three plates and average also all 36 readings of the 
reference point concentration. The average of the 36 readings of the 
reference point concentration is the correction point of the curve. 
Correct the average value obtained for each point to the figure it would 
be if the reference point reading for that set of three plates were the 
same as the correction point. Thus, if in correcting the 0.8-microgram 
concentration, the average of the 36 readings of the 1.0 microgram per 
milliliter (reference point) concentration is 16.5 millimeters and the 
average of the 1.0 microgram per milliliter concentration of the set of 
three plates (the 0.8 microgram per milliliter set) is 16.3 millimeters, 
the correction is +0.2 millimeter. If the average readings of the 0.8 
microgram per milliliter concentration of these same three plates is 
15.9 millimeters, the corrected value is then 16.1 millimeters. Plot 
these corrected values, including the average of the 1.0 microgram per 
milliliter concentration, on 2-cycle semilog paper, using the 
concentration in micrograms per milliliter as the ordinate (the 
logarithmic scale) and the diameter of the zone of inhibition as the 
abscissa. Draw the standard curve through these points either by 
inspection or by means of the following equations:

                           H=(3a+2b+c-e)/(5),

                           L=(3e+2d+c-a)/(5),

where:
L=Calculated zone diameter for the lowest concentration of the standard 
curve;
H=Calculated zone diameter for the highest concentration of the standard 
curve;
c=Average zone diameter of 36 readings of the reference point standard 
solution;
a, b, d, e=Corrected average values for the other standard solutions, 
lowest to highest concentrations, respectively.

    (h) Assay procedure. Use three plates for each sample. Fill three 
cylinders on each plate with the standard and three cylinders with the 
sample, which has been diluted to the reference concentration, 
alternating standard and sample. Incubate the plates for 16 hours to 18 
hours at 32 deg. C.-35 deg. C., and then measure the diameter of each 
zone of inhibition. To estimate the potency of the sample, average the 
zone readings of the standard and the zone readings of the sample on the 
three plates used. If the sample gives a larger zone size than the 
average of the standard, add the difference between them to the 
reference point zone of the standard curve. If the average value is 
lower than the standard value, subtract the difference between them from 
the reference point value on the curve. From the curve, read the 
potencies corresponding to these corrected values of zone sizes.
    (ii) Plate assay using Staphylococcus aureus (ATCC 6538P).\1\ 
Proceed as directed in paragraph (b)(1)(i) of this section except that 
the reference concentration of the sample under test is 10.0 micrograms 
of neomycin per milliliter; the concentrations of the standard curve 
solutions are 6.4, 8.0, 10.0, 12.5, 15.6 micrograms of neomycin per 
milliliter; and the suspension of the test organism, staphylococcus 
aureus (ATCC 6538P),\1\ is adjusted so that a 1:19

[[Page 713]]

dilution will give 25 percent light transmission and the usual inoculum 
for each 100 milliliters of agar for the seed layer is 0.2 milliliter of 
diluted suspension.
---------------------------------------------------------------------------

    \1\ Available from: American Type Culture Collection, 12301 Parklawn 
Dr., Rockville, MD 20852.
---------------------------------------------------------------------------

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 440.80a(b)(3) of this 
chapter, using a test dose of 1.0 milliliter per kilogram of a solution 
containing 10 milligrams of neomycin per milliliter in pyrogen-free, 
sterile U.S.P. saline T.S.
    (4) [Reserved]
    (5) Moisture. In an atmosphere of about 10 percent relative 
humidity, transfer about 100 milligrams of the finely powdered sample to 
a tared weighing bottle equipped with ground-glass top and stopper. 
Weigh the bottle and place it in a vacuum oven, tilting the stopper on 
its side so that there is no closure during the drying period. Dry at a 
temperature of 60 deg. C. and a pressure of 5 millimeters of mercury or 
less for 3 hours. At the end of the drying period, fill the vacuum oven 
with air dried by passing it through a drying agent such as sulfuric 
acid or silica gel. Replace the stopper and place the weighing bottle in 
a desiccator over a desiccating agent such as phosphorus pentoxide or 
silica gel, allow to cool to room temperature, and reweigh. Calculate 
the percent loss.
    (6) pH. Proceed as directed in Sec. 440.80a(b)(5)(ii) of this 
chapter, using a solution containing 33 milligrams of neomycin per 
milliliter.
    (7) Identity--(i) Reagents. (a) Sulfuric acid solution: Mix 
concentrated sulfuric acid and distilled water in volumetric proportions 
of 40:60.
    (b) Xylene.
    (c) p-Bromoaniline: (Prepare and store this reagent in brown, 
nonactinic glassware.) Place 380 milliliters of thioureasaturated 
glacial acetic acid solution in the bottle, add 10 milliliters of 20 
percent sodium chloride solution, 5 milliliters of 5 percent oxalic acid 
solution, and 5 milliliters of 10 percent disodium phosphate solution, 
and mix well. Add 8 grams of p-bromoaniline and mix well. Let this 
reagent stand overnight before use. Prepare the reagent once weekly.
    (ii) Procedure. Place about 10 milligrams of the sample into a test 
tube (19 millimeters  x  150 millimeters), dissolve with 1 milliliter of 
water, and then carefully add 5 milliliters of the sulfuric acid 
solution. Heat in a boiling water bath for 100 minutes. Cool to room 
temperature. Add 10 milliliters of xylene to the test tube. Stopper the 
tube and shake vigorously for about 1 minute. Let the two layers 
separate and then decant the xylene layer into a second test tube. Add 
10 milliliters of the p-bromoaniline reagent to the xylene solution, 
shake, and let stand. The development of a vivid pink-red color is a 
positive identity test for neomycin.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985; 53 
FR 12660, Apr. 15, 1988]



Sec. 444.46  Netilmicin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Netilmicin sulfate is the sulfate salt of 
d-Streptamine,4-O-[3-amino-6-(aminomethyl)-3,4-dihydro-2H-pyran-2-yl]-2-
deoxy-6-O-[3-deoxy-4-C-methyl-3-(methylamino)--l-
arabinopyranosyl]-N \1\-ethyl-, (2S-cis)-, (2:5). It is a white-to-buff-
colored powder. It is so purified and dried that:
    (i) Its potency is not less than 595 micrograms of netilmicin per 
milligram on an anhydrous basis.
    (ii) Its loss on drying is not more than 15.0 percent.
    (iii) Its pH in an aqueous solution containing 40 milligrams per 
milliliter is not less than 3.5 and not more than 5.5.
    (iv) Its residue on ignition is not more than 1.0 percent.
    (v) Its specific rotation in an aqueous solution containing 30 
milligrams per milliliter at 25 deg. C is not less than +88 deg. and not 
more than +96 deg..
    (vi) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH,

[[Page 714]]

residue on ignition, specific rotation, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 12 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), to obtain a stock solution of 
convenient concentration. Dilute an aliquot of the stock solution with 
solution 3 to the reference concentration of 0.1 microgram of netilmicin 
per milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(c) of this 
chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 40 milligrams per milliliter.
    (4) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (5) Specific rotation. Use an aqueous solution containing 3 
milligrams of sample per milliliter. Proceed as directed in Sec. 436.210 
of this chapter, using a 1.0-decimeter tube, and calculate the specific 
rotation on an anhydrous basis.
    (6) Identity. Proceed as directed in Sec. 436.318 of this chapter, 
except:
    (i) Prepare sample and standard solutions containing 10 milligrams 
of netilmicin per milliliter;
    (ii) Use 5 microliters of the solutions to spot the chromatography 
plate;
    (iii) Remove the plate from the tank after 1.5 hours; and
    (iv) Netilmicin sulfate appears as a brown spot.
[48 FR 18800, Apr. 26, 1983; 48 FR 22144, May 17, 1983, as amended at 55 
FR 11584, Mar. 29, 1990]



Sec. 444.50  Paromomycin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Paromomycin sulfate is the sulfate salt 
of a kind of paromomycin or a mixture of two or more such salts. It is a 
creamy-white to light-yellow powder. It is so purified and dried that:
    (i) Its potency is not less than 675 micrograms per milligram on an 
anhydrous basis.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 5.0 percent.
    (iv) The pH of a 3.0 percent aqueous solution is not less than 5.0 
and not more than 7.5.
    (v) Its specific rotation at 25 deg. C. in water is not less than 
+50 deg. and not more than +55 deg. on an anhydrous basis.
    (vi) Its residue on ignition is not more than 2.0 percent.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, specific rotation, and residue on ignition.
    (ii) Samples of the batch: 10 packages, each containing 
approximately 500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), to give a stock solution of 
convenient concentration. Further dilute the stock solution with 
solution 3 to the reference concentration of 1.0 microgram of 
paromomycin per milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
3.0 percent aqueous solution.
    (5) Specific rotation. Accurately weigh approximately 1.25 grams of 
the sample into a 25-milliliter volumetric flask. Dissolve in a few 
milliliters of water, add water to volume, and mix. Proceed as directed 
in Sec. 436.210 of this chapter, using a 2.0-decimeter polarimeter tube. 
Calculate the specific rotation on an anhydrous basis.
    (6) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]

[[Page 715]]



Sec. 444.62  Sisomicin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sisomicin sulfate is the sulfate salt of 
O-3-deoxy-4-C-methyl-3-(methylamino)--L- 
arabinopyranosyl(14)-O-[2,6-diamino-2,3,4,6-tetradeoxy-
-D-glycero- hex-4-enopyranosyl(16)-2-deoxy-L-
streptamine. It is a hygroscopic powder. It is so purified and dried 
that:
    (i) Its potency is not less than 580 micrograms of sisomicin per 
milligram on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 15.0 percent.
    (iv) Its pH in an aqueous solution containing 40 milligrams per 
milliliter is not less than 3.5 and not more than 5.5.
    (v) Its residue on ignition is not more than 1.0 percent.
    (vi) Its specific rotation in an aqueous solution containing 10 
milligrams per milliliter at 25 deg. C is not less than +100 deg. and 
not more than +110 deg..
    (vii) It gives a positive identity test for sisomicin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, residue on ignition, specific rotation, and identity.
    (ii) Samples required: 12 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay. Sisomicin is hygroscopic and care 
should be exercised during storage and weighing of samples.
    (1) Potency. Proceed as directed in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Dissolve an accurately 
weighed sample in sufficient 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to give a stock solution of convenient concentration. 
Further dilute an aliquot of the stock solution with solution 3 to the 
reference concentration of 0.1 microgram of sisomicin per milliliter 
(estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(c) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 40 milligrams of sisomicin per 
milliliter.
    (5) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (6) Specific rotation. Accurately weigh the sample to be tested in a 
volumetric flask and dilute with sufficient distilled water to give a 
solution containing approximately 10 milligrams per milliliter. Proceed 
as directed in Sec. 436.210 of this chapter, using a 1.0 decimeter 
polarimeter tube and calculate the specific rotation on an anhydrous 
basis.
    (7) Identity. Proceed as directed in Sec. 436.318 of this chapter, 
except:
    (i) Prepare sample and standard solutions containing 10 milligrams 
of sisomicin per milliliter;
    (ii) Use 5 microliters of the solutions to spot the chromatographic 
plates;
    (iii) Remove the plate from the tank after 3 hours; and
    (iv) The compound appears as a brown spot.
[46 FR 2988, Jan. 13, 1981; 46 FR 16676, Mar. 13, 1981; 46 FR 22359, 
Apr. 20, 1981, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.70a  Sterile streptomycin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile streptomycin sulfate is the 
sulfate salt of a kind of streptomycin or a mixture or two or more such 
salts. It is so purified and dried that:
    (i) Its potency is not less than 650 micrograms and not more than 
850 micrograms of streptomycin per milligram. If it is packaged for 
dispensing, its content is satisfactory if it is not less than 90 
percent and not more than 115 percent of the number of milligrams of 
streptomycin that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) It contains no depressor substances.
    (vi) Its loss on drying is not more than 5.0 percent.
    (vii) Its pH in an aqueous solution containing 200 milligrams per 
milliliter

[[Page 716]]

is not less than 4.5 and not more than 7.0.
    (viii) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, depressor substances, loss on drying, pH, and identity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in 
manufacturing another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 12 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient sterile distilled 
water to give a stock solution of convenient concentration; and also, if 
it is packaged for dispensing, reconstitute as directed in the labeling. 
Then using a suitable hypodermic syringe and needle, remove all of the 
withdrawable contents from each container represented as a single-dose 
container; or, if the labeling specifies the amount of potency in a 
given volume of the resultant preparation, withdraw an accurately 
measured representative portion from each container. Accurately dilute 
the sample thus obtained with sterile distilled water to give a stock 
solution of convenient concentration. Further dilute an aliquot of the 
stock solution with sterile distilled water to the reference 
concentration of 30 micrograms of streptomycin per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 10 milligrams of streptomycin per 
milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 200 milligrams per milliliter.
    (8) Identity--(i) Reagents. (a) 10 percent ferric chloride stock 
solution: Dissolve 5 grams of FeCl36H2O in 
50 milliliters of 0.1N HCl.
    (b) 0.25 percent ferric chloride solution: Dilute 2.5 milliliters of 
10 percent ferric chloride in 0.1N HCl to 100 milliliters with 0.01N 
HCl. Prepare the solution fresh daily.
    (ii) Procedure. Using distilled water, dilute the sample to be 
tested to a concentration of approximately 1,000 micrograms per 
milliliter. To 5.0 milliliters of this solution, add 2.0 milliliters of 
1N NaOH and heat in a boiling water bath for 10 minutes. Cool in the ice 
water for 3 minutes and then acidify the solution by adding 2.0 
milliliters of 1.2N HCl. Add 5.0 milliliters of 0.25 percent ferric 
chloride reagent. A violet color indicates the presence of streptomycin.
[42 FR 21275, Apr. 26, 1977, as amended at 46 FR 60568, Dec. 11, 1981; 
50 FR 19919, May 13, 1985]



Sec. 444.80  Tobramycin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tobramycin is 0-3-amino-3-deoxy-
-D-glucopyranosyl-(14)-0-[2,6-diamino-2,3,6-trideoxy-
-D-ribo-hexopyranosyl-(16)]-2-deoxy-L-streptamine. It 
is so purified and dried that:
    (i) Its potency is not less than 900 micrograms of tobramycin per 
milligram on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 8 percent.
    (iv) Its pH in an aqueous solution containing 100 milligrams per 
milliliter is not less than 9 and not more than 11.

[[Page 717]]

    (v) It gives a positive identity test for tobramycin.
    (vi) Its residue on ignition is not more than 1.0 percent.
    (vii) Its heavy metals content is not more than 30 parts per 
million.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, identity, residue on ignition, and heavy metals.
    (ii) Samples required: 10 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological turbidimetric assay shall be conclusive:
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient distilled water to 
obtain a stock solution of convenient concentration. Further dilute an 
aliquot of the stock solution with distilled water to the reference 
concentration of 2.5 micrograms of tobramycin per milliliter 
(estimated).
    (ii) Nonaqueous titration. Proceed as directed in Sec. 436.213 of 
this chapter, using the titration procedure described in paragraph 
(e)(2) of that section. Calculate the tobramycin content as follows:

Micrograms tobramycin per milligram = [(A-B) x (normality of perchloric 
          acid reagent) x 93.4 x 100 x 1,000]/(Weight of sample in 
          milligrams x (100-m))

where:
A=Milliliters of perchloric acid reagent used in titrating the sample;
B=Milliliters of perchloric acid reagent used in titrating the blank;
m=Percent moisture of the sample.

    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (5) Identity. Proceed as directed in Sec. 436.318 of this chapter.
    (6) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (7) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
[40 FR 57798, Dec. 12, 1975, as amended at 45 FR 16476, Mar. 14, 1980; 
50 FR 19919, May 13, 1985]



Sec. 444.81a  Sterile tobramycin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile tobramycin sulfate is the sulfate 
salt of 0-3-amino-3-deoxy--D-glucopyranosyl-(14)-0-
[2,6-diamino-2,3,6-trideoxy--D-ribo-hexopyranosyl-
(16)]-2-deoxy-L-streptamine. It is a lyophilized powder. It is 
so purified and dried that:
    (i) Its potency is not less than 634 micrograms and not more than 
739 micrograms of tobramycin per milligram on an ``as is'' basis. If it 
is packaged for dispensing, its content is satisfactory if it is not 
less than 90 percent and not more than 115 percent of the number of 
milligrams of tobramycin that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its moisture content is not more than 2.0 percent.
    (vi) Its pH in an aqueous solution containing 40 milligrams per 
milliliter, or when reconstituted as directed in the labeling, is not 
less than 6.0 and not more than 8.0.
    (vii) It gives a positive identity test for tobramycin.
    (viii) Its residue on ignition is not more than 1.0 percent.
    (ix) Its heavy metals content is not more than 30 parts per million.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, identity, residue on ignition, and heavy metals.
    (ii) Samples required:

[[Page 718]]

    (a) If the batch is packaged for repacking or for use in the 
manufcture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 14 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample with sufficient sterile distilled 
water to obtain a stock solution of convenient concentration; also, if 
it is packaged for dispensing, reconstitute as directed in the labeling. 
Then, using a suitable hypodermic needle and syringe, remove all of the 
withdrawable contents if it is represented as a single dose container; 
or if the labeling specifies the amount of potency in a given volume of 
the resultant preparation, remove an accurately measured representative 
portion from each container. Dilute with sterile distilled water to 
obtain a stock solution of convenient concentration. Further dilute a 
portion of the stock solution with sterile distilled water to the 
reference concentration of 2.5 micrograms of tobramycin per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 10 milligrams of tobramycin per milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 40 milligrams per milliliter, or if it is 
packaged for dispensing, reconstitute as directed in the labeling.
    (7) Identity. Proceed as directed in Sec. 436.318 of this chapter.
    (8) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (9) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
[44 FR 26072, May 4, 1979, as amended at 45 FR 16476, Mar. 14, 1980; 50 
FR 19919, May 13, 1985]



                      Subpart B--Oral Dosage Forms



Sec. 444.130  Kanamycin sulfate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Kanamycin sulfate capsules are composed 
of crystalline kanamycin sulfate, with or without one or more suitable 
and harmless buffer substances, vegetable oils, preservatives, diluents, 
binders, lubricants, colorings, and flavorings, enclosed in gelatin 
capsules. Each capsule contains 500 milligrams of kanamycin. Its potency 
is satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of milligrams of kanamycin that it is represented 
to contain. The loss on drying is not more than 4.0 percent. The 
crystalline kanamycin sulfate used conforms to the standards prescribed 
by Sec. 444.30(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The kanamycin sulfate used in making the batch for potency, loss 
on drying, pH, residue on ignition, identity, kanamycin B content, and 
crystallinity.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:
    (a) Kanamycin sulfate used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch: Minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed

[[Page 719]]

glass blender jar with sufficient sterile distilled water to give a 
stock solution of convenient concentration. Blend for 3 to 5 minutes. 
Remove an aliquot and further dilute with sterile distilled water to the 
reference concentration of 10 micrograms of kanamycin per milliliter 
(estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.142  Neomycin sulfate oral dosage forms.



Sec. 444.142a  Neomycin sulfate tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate tablets are tablets 
composed of neomycin sulfate with one or more suitable and harmless 
binders, and with or without one or more suitable and harmless fillers, 
buffers, lubricants, and colorings. Each tablet contains 150 milligrams, 
175 milligrams, or 350 milligrams of neomycin. The moisture content is 
not more than 10.0 percent. Tablets shall disintegrate within 1 hour. 
The neomycin sulfate used conforms to the standards prescribed by 
Sec. 444.42a(a)(1)(i), (v), (vi), and (vii). Each other substance used, 
if its name is recognized in the U.S.P. or N.F., conforms to the 
standards prescribed therefor by such official compendium.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. Its expiration date is 12 
months.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, 
moisture, pH, and identity.
    (b) The batch for potency, moisture, and disintegration time.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except disintegration time: Minimum 30 tablets.
    (2) For disintegration time: Six tablets.
    (c) In the case of an initial request for certification, each other 
ingredient used in making the batch: One package of each containing 
approximately 5 grams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 444.42a(b)(1), except prepare the sample as follows: Place a 
representative number of tablets into a high-speed glass blender, add a 
sufficient quantity of 0.1M potassium phosphate buffer, pH 8.0, to give 
a stock solution of convenient concentration. Blend 3 to 5 minutes. 
Further dilute in 0.1M potassium phosphate buffer, pH 8.0, to the proper 
prescribed reference concentration. Its neomycin content is satisfactory 
if it contains not less than 90 percent and not more than 125 percent of 
the number of milligrams of neomycin that it is represented to contain.
    (2) Moisture. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Disintegration time. Proceed as directed in Sec. 440.180a(b)(3) 
of this chapter.
[39 FR 19046, May 30, 1974, as amended at 46 FR 25608, May 8, 1981; 50 
FR 19919, May 13, 1985]



Sec. 444.142b  Neomycin sulfate oral solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate oral solution is 
neomycin sulfate with or without one or more suitable and harmless 
flavorings, colorings, and preservatives in an aqueous vehicle. Each 
milliliter contains 17.5 milligrams of neomycin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 125 
percent of the number of milligrams of neomycin that it is represented 
to contain. Its pH is not less than 5.0 and not more than 7.5. The 
neomycin sulfate used conforms to the standards prescribed by 
Sec. 444.42a(a)(1) (i), (v), (vi), and (vii).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:

[[Page 720]]

    (a) The neomycin sulfate used in making the batch for potency, 
moisture, pH, and identity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, except prepare the sample as follows: 
Remove an accurately measured representative portion with a suitable 
syringe, and dilute with sufficient 0.1 M potassium phosphate buffer, pH 
8.0 (solution 3), to give a stock solution of convenient concentration. 
Further dilute with solution 3 to the reference concentration of 1.0 
microgram of neomycin per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.150  Paromomycin sulfate oral dosage forms.



Sec. 444.150a  Paromomycin sulfate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Paromomycin sulfate capsules are 
paromomycin sulfate enclosed in a suitable and harmless gelatin capsule. 
Each capsule contains 250 milligrams of paromomycin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 125 
percent of the number of milligrams of paromomycin that it is 
represented to contain. The loss on drying is not more than 7.0 percent. 
The paromomycin sulfate used conforms to the standards prescribed 
therefor by Sec. 444.50(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The paromomycin sulfate used in making the batch for potency, 
loss on drying, pH, specific rotation, and residue on ignition.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:
    (a) The paromomycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Blend a representative number of capsules for 3 to 5 minutes in a high-
speed glass blender with sufficient 0.1M potassium phosphate buffer, pH 
8.0 (solution 3), to give a stock solution of convenient concentration. 
Further dilute the stock solution with solution 3 to the reference 
concentration of 1.0 microgram of paromomycin per milliliter 
(estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.150b  Paromomycin sulfate sirup.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Paromomycin sulfate sirup contains the 
equivalent of 25 milligrams of paromomycin per milliliter. Its potency 
is satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of milligrams of paromomycin that it is 
represented to contain. It may contain one or more suitable and harmless 
solvents, flavorings, colorings, preservatives, and buffers in water. 
Its pH is not less than 7.5 and not more than 8.5. The paromomycin 
sulfate used conforms to the requirements of Sec. 444.50(a)(1) (i), 
(ii), (iv), (v), and (vi).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays for:
    (a) The paromomycin sulfate used in making the batch for potency, 
pH, specific rotation, and residue on ignition.
    (b) The batch for potency and pH.

[[Page 721]]

    (ii) Samples required:
    (a) The paromomycin sulfate used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Remove an appropriate aliquot of the sirup and transfer to an 
appropriate-sized volumetric flask. Dilute to volume with 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), and mix well. Further dilute with 
solution 3 to the reference concentration of 1.0 microgram of 
paromomycin per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



                   Subpart C--Injectable Dosage Forms



Sec. 444.206  Amikacin sulfate injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amikacin sulfate injection is an aqueous 
solution of amikacin with suitable and harmless buffer substances and 
preservatives. Each milliliter contains amikacin sulfate equivalent to 
either 50 milligrams or 250 milligrams of amikacin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of amikacin that it is represented 
to contain. It is sterile. It is nonpyrogenic. Its pH is not less than 
3.5 and not more than 5.5. The amikacin used conforms to the standards 
prescribed by Sec. 444.6(a)(1) or, if amikacin sulfate is used, to the 
standards prescribed by Sec. 444.7(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The amikacin used in making the batch for potency, moisture, pH, 
identity, residue on ignition, specific rotation, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, and pH.
    (ii) Samples required:
    (a) The amikacin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 12 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place an accurately measured representative portion of the sample into 
an appropriate-sized volumetric flask and dilute to volume with sterile 
distilled water to give a stock solution of convenient concentration. 
Further dilute an aliquot of the stock solution with sterile distilled 
water to the reference concentration of 10.0 micrograms of amikacin per 
milliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 25 milligrams of amikacin per milliliter.
    (4) [Reserved]
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[41 FR 49483, Nov. 9, 1976, as amended at 50 FR 19919, May 13, 1985; 55 
FR 38677, Sept. 20, 1990]



Sec. 444.220  Gentamicin sulfate injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Gentamicin sulfate injection is an 
aqueous solution of gentamicin sulfate with or without one or more 
suitable buffers, sequestering agents, tonicity agents, or 
preservatives. Each milliliter contains gentamicin sulfate equivalent to 
either 0.4, 0.6, 0.7, 0.8, 0.9, 1.0, 1.2, 1.6, 2.0, 2.4, 10.0, or 40 
milligrams of gentamicin. Its potency is satisfactory if it contains

[[Page 722]]

not less than 90 percent nor more than 125 percent of the number of 
milligrams of gentamicin that it is represented to contain. It is 
sterile. It is nonpyrogenic. Its pH is not less than 3.0 nor more than 
5.5. The gentamicin sulfate used conforms to the standards prescribed by 
Sec. 444.20(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The gentamicin sulfate used in making the batch for potency, 
loss on drying, pH, specific rotation, content of gentamicins 
C1,C1a,C2, and identity.
    (b) The batch for gentamicin potency, sterility, pyrogens, and pH.
    (ii) Samples required:
    (a) The gentamicin sulfate used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 40 containers if 
each milliliter contains the equivalent of 2.0 milligrams or 10.0 
milligrams of gentamicin; a minimum of 12 containers if each milliliter 
contains the equivalent of 40.0 milligrams of gentamicin; or, a minimum 
of 10 containers if each milliliter contains the equivalent of 1.0 
milligram of gentamicin.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Using 0.1M potassium phosphate buffer, pH 8.0 (solution 3), dilute an 
accurately measured representative portion of the product to the 
reference concentration of 0.1 microgram of gentamicin per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) [Reserved]
    (4) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
except inject a sufficient volume of the undiluted solution to deliver 
10 milligrams of gentamicin per kilogram, but not to exceed 10 
milliliters per kilogram.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[39 FR 19046, May 30, 1974, as amended at 46 FR 2994, Jan. 13, 1981; 46 
FR 16683, Mar. 13, 1981; 46 FR 31009, June 12, 1981; 47 FR 56490, Dec. 
17, 1982; 48 FR 44775, Sept. 30, 1983; 49 FR 49287, Dec. 19, 1984; 50 FR 
10754, Mar. 18, 1985; 50 FR 19919, May 13, 1985]



Sec. 444.230  Kanamycin sulfate injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Kanamycin sulfate injection is an aqueous 
solution of kanamycin sulfate with suitable and harmless buffer 
substances and preservatives. It contains either 75 milligrams of 
kanamycin per 2.0 milliliters, or 250 milligrams of kanamycin per 
milliliter, or 1.0 gram of kanamycin per 3.0 milliliters. Its potency is 
satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of milligrams of kanamycin that it is represented 
to contain. It is sterile. It is nonpyrogenic. Its pH is not less than 
3.5 and not more than 5.0. The kanamycin sulfate used conforms to the 
standards prescribed by Sec. 444.30a(a)(1)(i), (v), (vii), (viii), (ix), 
and (x).
    (2) Labeling. In addition to the requirements prescribed by 
Sec. 432.5 of this chapter, the labeling of each package shall bear a 
warning to the effect that older patients and patients receiving a total 
dose of more than 20 grams of the drug should be carefully observed for 
signs of eighth-nerve damage. In patients with impaired kidney function 
or with prerenal azotemia, the risk of severe ototoxic reaction that may 
result in permanent deafness is sharply increased.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The kanamycin sulfate used in making the batch for potency, 
residue on ignition, loss on drying, identity, crystallinity, and 
kanamycin B content.

[[Page 723]]

    (b) The batch for potency, sterility, pyrogens, and pH.
    (ii) Samples required:
    (a) The kanamycin sulfate used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: Minimum of 12 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place an accurately measured representative aliquot of the sample into 
an appropriate-sized volumetric flask and dilute to volume with sterile 
distilled water to give a stock solution of convenient concentration. 
Further dilute an aliquot of the stock solution with sterile distilled 
water to the reference concentration of 10 micrograms of kanamycin per 
milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) [Reserved]
    (4) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 10 milligrams of kanamycin per milliliter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.246  Netilmicin sulfate injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Netilmicin sulfate injection is an 
aqueous solution of netilmicin sulfate and one or more buffers, 
chelating agents, antioxidants, and preservatives. Each milliliter 
contains netilmicin sulfate equivalent to 10 milligrams, 25 milligrams, 
or 100 milligrams of netilmicin. Its potency is satisfactory if it is 
not less than 90 percent and not more than 115 percent of the number of 
milligrams of netilmicin that it is represented to contain. It is 
sterile. It is nonpyrogenic. Its pH is not less than 3.5 and not more 
than 6.0. The netilmicin sulfate used conforms to the standards 
prescribed by Sec. 444.46(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The netilmicin sulfate used in making the batch for potency, 
loss on drying, pH, residue on ignition, specific rotation, and 
identity.
    (b) The batch for potency, sterility, pyrogens, and pH.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The netilmicin sulfate used in making the batch: 12 packages, 
each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 12 immediate 
containers.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dilute an accurately measured representative portion of the product with 
0.1M potassium phosphate buffer, pH 8.0 (solution 3), to the reference 
concentration of 0.1 microgram of netilmicin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 10 milligrams of netilmicin per milliliter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[48 FR 18801, Apr. 26, 1983, as amended at 55 FR 11584, Mar. 29, 1990]



Sec. 444.262  Sisomicin sulfate injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sisomicin sulfate injection is an aqueous 
solution of sisomicin sulfate and one or more suitable buffers, 
chelating agents, and preservatives.

[[Page 724]]

Each milliliter contains sisomicin sulfate equivalent to 50 milligrams 
of sisomicin. Its potency is satisfactory if it contains not less than 
90 percent and not more than 120 percent of the number of milligrams of 
sisomicin that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its pH is not less than 2.5 and not more than 5.5. The 
sisomicin sulfate used conforms to the standards prescribed by 
Sec. 444.62(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The sisomicin sulfate used in making the batch for potency, loss 
on drying, pH, residue on ignition, specific rotation, and identity.
    (b) The batch for potency, sterility, pyrogens, and pH.
    (ii) Samples required:
    (a) The sisomicin sulfate used in making the batch: 12 packages, 
each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 12 vials.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dilute an accurately measured representative portion of the product with 
0.1M potassium phosphate buffer, pH 8.0 (solution 3), to the reference 
concentration of 0.1 microgram of sisomicin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 10 milligrams of sisomicin per milliliter.
    (4) [Reserved]
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[46 FR 2989, Jan. 13, 1981, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.270  Streptomycin sulfate injectable dosage forms.



Sec. 444.270a  Sterile streptomycin sulfate.

    The requirements for certification and the tests and methods of 
assay for sterile streptomycin sulfate, packaged for dispensing, are 
described in Sec. 444.70a.
[42 FR 21275, Apr. 26, 1977]



Sec. 444.270b  Streptomycin sulfate injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Streptomycin sulfate injection is an 
aqueous solution of streptomycin sulfate. It may contain one or more 
suitable and harmless preservatives, buffer substances and stabilizing 
agents. Each milliliter contains streptomycin sulfate equivalent to 400 
milligrams, 420 milligrams, or 500 milligrams of streptomycin. Its 
potency is satisfactory if it is not less than 90 percent and not more 
than 115 percent of the number of milligrams of streptomycin that it is 
represented to contain. It is sterile. It is nonpyrogenic. It contains 
no depressor substances. Its pH is not less than 5.0 and not more than 
8.0. The streptomycin sulfate used conforms to the standards prescribed 
by Sec. 444.70a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain;
    (i) Results of tests and assays on:
    (a) The streptomycin sulfate used in making the batch for potency, 
depressor substances, loss on drying, pH, and identity.
    (b) The batch for potency, sterility, pyrogens, depressor substances 
(except that the results of this test performed on the streptomycin 
sulfate used in making the batch may be submitted instead), and pH.
    (ii) Samples required:
    (a) The streptomycin sulfate used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch:

[[Page 725]]

    (1) If the batch is packaged for use in the manufacture of another 
drug:
    (i) For all tests except sterility: Five containers, each containing 
not less than 2.0 milliliters.
    (ii) For sterility testing: 20 containers, each containing not less 
than 2.0 milliliters.
    (2) If the batch is packaged for dispensing:
    (i) For all tests except sterility: A minimum of eight immediate 
containers.
    (ii) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and method of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Using a suitable hypodermic syringe and needle, remove all of the 
withdrawable contents if it is represented as a single-dose container; 
or if the labeling specifies the amount of potency in a given volume of 
the resultant preparation, remove an accurately measured representative 
portion from each container. Accurately dilute the portion with sterile 
distilled water to give a stock solution of convenient concentration. 
Further dilute an aliquot of the stock solution with sterile distilled 
water to the reference concentration of 30 micrograms of streptomycin 
per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 10 milligrams of streptomycin per 
milliliter.
    (4) [Reserved]
    (5) Depressor substances (the depressor substances test may be 
omitted if it is performed on the streptomycin sulfate used in preparing 
the injection). Proceed as directed in Sec. 436.35 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[42 FR 21275, Apr. 26, 1977; 42 FR 37543, July 7, 1977, as amended at 46 
FR 60568, Dec. 11, 1981; 50 FR 19919, May 13, 1985]



Sec. 444.280  Tobramycin sulfate injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tobramycin sulfate injection is 
tobramycin solubilized with sulfuric acid in an aqueous solution 
containing one or more suitable buffers, chelating agents, and 
preservatives. Each milliliter contains tobramycin sulfate equivalent to 
either 10 milligrams or 40 milligrams of tobramycin. Its potency is 
satisfactory if it contains not less than 90 percent and not more than 
120 percent of the number of milligrams of tobramycin that it is 
represented to contain. It is sterile. It is nonpyrogenic. Its pH is not 
less than 3.0 and not more than 6.5. The tobramycin used conforms to the 
standards prescribed by Sec. 444.80(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The tobramycin used in making the batch for potency, moisture, 
pH, identity, residue on ignition, and heavy metals.
    (b) The batch for potency, sterility, pyrogens, and pH.
    (ii) Samples required:
    (a) The tobramycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 40 vials if each 
milliliter contains 10 milligrams of tobramycin per milliliter, or a 
minimum of 12 vials if each milliliter contains the equivalent of 40 
milligrams of tobramycin.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
If the immediate container is a single-dose vial, use a suitable 
hypodermic needle and syringe and remove all the withdrawable contents; 
or, if the labeling specifies the amount of potency in a given volume,

[[Page 726]]

remove an accurately measured representative portion from each 
container. Dilute this portion with sufficient distilled water to give a 
stock solution of convenient concentration. If the preparation is 
packaged in a prefilled syringe, eject the entire contents of the 
syringe and dilute with distilled water to obtain a stock solution of 
convenient concentration. Further dilute the stock solution to the 
reference concentration of 2.5 micrograms of tobramycin per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 10 milligrams of tobramycin per milliliter.
    (4) [Reserved]
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[40 FR 57798, Dec. 12, 1975, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.281  Sterile tobramycin sulfate.

    The requirements for certification and the tests and methods of 
assay for sterile tobramycin sulfate packaged for dispensing are 
described in Sec. 444.81a.
[44 FR 26072, May 4, 1979]



                   Subpart D--Ophthalmic Dosage Forms



Sec. 444.320  Gentamicin sulfate ophthalmic dosage forms.



Sec. 444.320a  Gentamicin sulfate ophthalmic solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Gentamicin sulfate ophthalmic solution 
contains in each milliliter the equivalent of 3.0 milligrams of 
gentamicin and suitable buffers and preservatives. Its potency is 
satisfactory if it is not less than 90 and not more than 135 percent of 
the number of milligrams of gentamicin it is represented to contain. It 
is sterile. Its pH is not less than 6.5 nor more than 7.5. The 
gentamicin sulfate conforms to the standards prescribed by 
Sec. 444.20(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The gentamicin sulfate used in making the batch for potency, 
loss on drying, pH, specific rotation, content of gentamicins 
C1, C1a, and C2, and identity.
    (b) The batch for potency, sterility, and pH.
    (ii) Samples required:
    (a) The gentamicin sulfate used in making the batch: 10 packages, 
each containing not less than 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of five immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dilute an accurately measured representative portion of the product with 
0.1M potassium phosphate buffer, pH 8.0 (solution 3), to the reference 
concentration of 0.1 microgram of gentamicin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.320b  Gentamicin sulfate ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Gentamicin sulfate ointment contains in 
each gram the equivalent of 3.0 milligrams of gentamicin with suitable 
preservatives in a white petrolatum base. Its potency is satisfactory if 
it is not less than 90 percent and not more than 135 percent of the 
number of milligrams of gentamicin

[[Page 727]]

that it is represented to contain. It is sterile. Its moisture content 
is not more than 1.0 percent. It passes the test for particulate 
contamination. The gentamicin sulfate used conforms to the standards 
prescribed therefor by Sec. 444.20a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The gentamicin sulfate used in making the batch for potency, 
loss on drying, pH, specific rotation, content of gentamicins 
C1, C1a, and C2, and identity.
    (b) The batch for gentamicin potency, sterility, moisture, and 
particulate contamination.
    (ii) Samples required:
    (a) The gentamicin sulfate used in making the batch: 10 packages, 
each containing not less than 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, except prepare the sample as follows: 
Place an accurately weighed representative portion of the ointment into 
a separatory funnel containing 50 milliliters of peroxide-free ether. 
Shake the sample and ether until homogeneous. Add 20 to 25 milliliters 
of 0.1M potassium phosphate buffer, pH 8.0 (solution 3), and shake well. 
Allow the layers to separate. Remove the buffer layer and repeat the 
extraction with new portions of solution 3. Repeat any additional times 
necessary to insure complete extraction of the antibiotic. Combine the 
extractives and adjust to an appropriate volume to give a stock solution 
of convenient concentration. Further dilute with solution 3 to the 
reference concentration of 0.1 microgram of gentamicin per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) Particulate contamination. Proceed as directed in Sec. 436.206 
of this chapter.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.320c  Gentamicin sulfate-prednisolone acetate ophthalmic suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Gentamicin sulfate-prednisolone acetate 
ophthalmic suspension is an aqueous suspension containing in each 
milliliter gentamicin sulfate equivalent to 3.0 milligrams of gentamicin 
and 10.0 milligrams of prednisolone acetate. It contains suitable and 
harmless chelating agents, tonicity agents, buffers, and preservatives. 
Its gentamicin content is satisfactory if it is not less than 90 percent 
and not more than 130 percent of the number of milligrams of gentamicin 
that it is represented to contain. Its prednisolone acetate content is 
satisfactory if it is not less than 90 percent and not more than 110 
percent of the number of milligrams of prednisolone acetate that it is 
represented to contain. Its pH is not less than 5.4 and not more than 
6.6. It is sterile. The gentamicin sulfate used conforms to the 
standards prescribed by Sec. 444.20(a)(1). The prednisolone acetate used 
conforms to the standards prescribed by the USP XXI.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The gentamicin sulfate used in making the batch for potency, 
loss on drying, pH, specific rotation, content of gentamicin 
C1, Cla, C2, and identify.
    (B) The prednisolone acetate used in making the batch for all USP 
XXI specifications.
    (C) The batch for gentamicin content, prednisolone acetate content, 
sterility, and pH.

[[Page 728]]

    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The gentamicin sulfate used in making the batch: 10 packages, 
each containing not less than 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Gentamincin content. Proceed as 
directed in Sec. 436.105 of this chapter, preparing the sample for assay 
as follows: Dilute an accurately measured representative portion of the 
sample with 0.1M potassium phosphate buffer, pH 8.0 (solution 3), to the 
reference concentration of 0.1 microgram of gentamicin per milliliter 
(estimated).
    (2) Prednisolone acetate content. Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 254 nanometers, a column 
packed with octadecyl hydrocarbon bonded silicas, a flow rate of 2.0 
milliliters per minute, and an injection volume of 30 microliters. 
Mobile phase, reference standard and sample solutions, system 
suitability requirements, and calculations are as follows:
    (i) Mobile phase. Mix acetonitrile distilled deionized water 
(40:60). Filter the mobile phase through a suitable glass fiber filter 
or equivalent which is capable of removing particulate contamination to 
1 micron in diameter.
    (ii) Reference standard and sample solutions--(A) Preparation of 
reference standard solution. Accurately weigh approximately 60 
milligrams of prednisolone acetate reference standard into a 50-
milliliter volumetric flask. Dissolve and dilute to volume with methyl 
alcohol and mix well. Transfer 8 milliliters of this solution into a 50-
milliliter volumetric flask, dilute to volume with 70 percent methyl 
alchohol, and mix well.
    (B) Preparation of sample solution. Transfer 1.0 milliliter of the 
sample into a 50-milliliter volumetric flask, dilute to volume with 70 
percent methyl alcohol, and mix well.
    (iii) System suitability requirements--(A) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.25 at 5 
percent of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 2,000 theoretical plates.
    (C) Coefficient of variation. The coefficient of variation (SR 
in percent) of five replicate injections is satisfactory if it is not 
more than 2.0 percent. If the system suitability requirements have been 
met, then proceed as described in Sec. 436.216(b) of this chapter.
    (iv) Calculations. Calculate the milligrams of prednisolone acetate 
per milliliter of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.226

where:
Au=Area of the prednisolone acetate peak in the chromatogram 
          of the sample (at a retention time equal to that observed for 
          the standard);
As=Area of the prednisolone acetate peak in the chromatogram 
          of the prednisolone acetate reference standard;
Cs=Concentration of prednisolone acetate in the reference 
          standard solution in milligrams per milliliter; and
d=Dilution factor of the sample.

    (3) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[53 FR 40725, Oct. 18, 1988, as amended at 59 FR 8398, Feb. 22, 1994]



Sec. 444.320d  Gentamicin sulfate-prednisolone acetate ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Gentamicin sulfate-prednisolone acetate 
ophthalmic ointment contains in each gram gentamicin sulfate equivalent 
to 3.0 milligrams of gentamicin and 6.0 milligrams of prednisolone 
acetate, with a suitable lubricant and preservative in a suitable and 
harmless white petrolatum base. Its gentamicin content is satisfactory 
if it is not less than 90 percent and not more than 120 percent of the 
number of milligrams of gentamicin that it is represented to contain. 
Its prednisolone acetate content is satisfactory if it is

[[Page 729]]

not less than 90 percent and not more than 110 percent of the number of 
milligrams of prednisolone acetate that it is represented to contain. It 
is sterile. Its moisture content is not more than 2.0 percent. It passes 
the test for metal particles. The gentamicin sulfate used conforms to 
the standards prescribed by Sec. 444.20(a)(1). The prednisolone acetate 
used conforms to the standards prescribed by the United States 
Pharmacopeia.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The gentamicin sulfate used in making the batch for potency, 
loss on drying, pH, specific rotation, content of gentamicins 
C1, C1a, C2, and identity.
    (B) The prednisolone acetate used in making the batch for all USP 
XXI specifications.
    (C) The batch for gentamicin content, prednisolone acetate content, 
sterility, moisture, and metal particles.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research:
    (A) The gentamicin sulfate used in making the batch: 10 packages, 
each containing not less than 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Gentamicin content. Proceed as 
directed in Sec. 436.105 of this chapter, except prepare the sample as 
follows: Place an accurately weighed representative portion of the 
ointment into a separatory funnel containing 50 milliliters of peroxide-
free ether. Shake the sample and ether until homogeneous. Add 20 to 25 
milliliters of 0.1M potassium phosphate buffer, pH 8.0 (solution 3), and 
shake well. Allow the layers to separate. Remove the buffer layer and 
repeat the extraction with new portions of solution 3. Repeat any 
additional times necessary to insure complete extraction of the 
antibiotic. Combine the extractives and adjust to an appropriate volume 
to give a stock solution of convenient concentration. Further dilute 
with solution 3 to the reference concentration of 0.1 microgram of 
gentamicin per milliliter (estimated).
    (2) Prednisolone acetate content. Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 254 nanometers, a column 
packed with octadecyl hydrocarbon bonded silicas 3 to 10 micrometers in 
diameter, a flow rate of 2.0 milliliters per minute, and an injection 
volume of 30 microliters. Reagents, working standard and sample 
solutions, system suitability requirements, and calculations are as 
follows:
    (i) Reagents--(A) Mobile phase. Mix acetonitrile distilled deionized 
water (40:60). Filter the mobile phase through a suitable glass fiber 
filter or equivalent which is capable of removing particulate 
contamination to 1 micron in diameter. Degas the mobile phase just prior 
to its introduction into the chromatograph.
    (B) Internal standard solution. Accurately weigh 135 milligrams 
 10 milligrams of fluorometholone acetate into a 50-
milliliter volumetric flask. Dissolve and dilute to volume with methyl 
alcohol.
    (ii) Preparation of working standard and sample solutions--(A) 
Working standard solution. Prepare the working standard solution fresh 
before injection by dissolving approximately 40 miligrams  2 
milligrams of prednisolone acetate, accurately weighed, into a 100-
milliliter volumetric flask with 25 milliliters of methyl alcohol. 
Sonicate to dissolve and dilute to volume with methyl alcohol and mix 
well. Transfer 8 milliliters of this solution into a 50-milliliter 
volumetric flask. Add 25 milliliters of hexane and shake. Add 2.0 
milliliters of internal standard as described in paragraph (b)(2)(i)(B) 
of this section, and dilute to volume with methyl alcohol. Shake 
vigorously for 30 seconds, allow the phases to separate, then aspirate 
the upper hexane layer and dilute to volume with methyl alcohol. 
Centrifuge for 10 minutes at 5,700 revolutions per minute.
    (B) Sample solution. Accurately weigh 500 milligrams  20 
milligrams of the sample into a 50-milliliter volumetric

[[Page 730]]

flask. Add 25 milliliters of hexane and sonicate. Add 2.0 milliliters of 
the internal standard. Dilute to volume with methyl alcohol. Shake 
vigorously for 30 seconds and allow the phase to separate. Aspirate the 
upper hexane and cloudy layers. Dilute to volume with methyl alcohol. 
Centrifuge for 10 minutes at 5,700 revolutions per minute.
    (iii) System suitability requirements--(A) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.50 at 5 
percent of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 2,500 theoretical plates.
    (C) Resolution. The resolution (R) between the peak for prednisolone 
acetate and the internal standard is satisfactory if it is not less than 
2.0.
    (D) Coefficient of variation. The coefficient of variation 
(SR in percent) of five replicate injections is satisfactory 
if it is not more than 2.0 percent. If the system suitability 
requirements have been met, then proceed as described in Sec. 436.216(b) 
of this chapter. Alternate chromatographic conditions are acceptable 
provided comparable system suitability requirements are met. However, 
the sample preparation described in paragraph (b)(2)(ii)(B) of this 
section should not be changed.
    (iv) Calculations. Calculate the percent of prednisolone acetate as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.227

where:
Ru=Area of the prednisolone acetate peak in the chromatogram 
          of the sample (at a retention time equal to that observed for 
          the standard)/Area of internal standard peak;
Rs=Area of the prednisolone acetate peak in the chromatogram 
          of the prednisolone acetate working standard /Area of internal 
          standard peak;
Ps=Prednisolone acetate activity in the prednisolone acetate 
          working standard solution in milligrams per milliliter;
Wu=Weight of sample in milligrams; and
d=Dilution factor of the sample.

    (3) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in Sec. 436.20(e)(3).
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) Metal particles. Proceed as directed in Sec. 436.206 of this 
chapter.
[55 FR 2643, Jan. 26, 1990]



Sec. 444.342  Neomycin sulfate ophthalmic dosage forms.



Sec. 444.342a  Neomycin sulfate- ------------ ophthalmic suspension; neomycin sulfate- ------------ ophthalmic solution (the blanks being filled in with the 

          established name(s) of the other active ingredient(s) present 
          in accordance with paragraph (a)(1) of this section).

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. The drug is a suspension or a solution 
containing, in each milliliter, 3.5 milligrams of neomycin and the 
following other active ingredients in a suitable and harmless vehicle:
    (i) 15 milligrams of cortisone acetate; or
    (ii) 5 milligrams or 25 milligrams of hydrocortisone acetate; or
    (iii) 1 milligram or 2 milligrams of prednisolone; or
    (iv) 1 milligram of sodium dexa-methasone phosphate; or
    (v) 5 milligrams of prednisolone phosphate.

It contains suitable and harmless buffers, dispersants, and 
preservatives. It is sterile. Its pH is not less than 6.0 and not more 
than 8.0. The neomycin sulfate used conforms to the standards prescribed 
by Sec. 444.42a(a)(1) (i), (vi), and (vii). Each other substance used, 
if its name is recognized in the U.S.P. or N.F., conforms to the 
standards prescribed therefor by such official compendium.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. Its expiration date is 12 
months.
    (3) Request for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, pH, 
and identity.
    (b) The batch for potency, sterility, and pH.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 containers, 
each

[[Page 731]]

containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 5 immediate 
containers.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (c) In case of an initial request for certification, each other 
ingredient used in making the batch: One package of each containing 
approximately 5 grams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 444.442a(b)(1). Its neomycin content is satisfactory if it contains 
not less than 90 percent and not more than 130 percent of the number of 
milligrams of neomycin that it is represented to contain.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
if the steroid prevents solubilization, use 0.25 milliliter of sample in 
lieu of 1 milliliter and proceed as directed in paragraph (e)(2) of that 
section.
    (3) pH. Proceed as directed in Sec. 440.80a(b)(5)(ii) of this 
chapter, using the undiluted sample.
[39 FR 19046, May 30, 1974, as amended at 47 FR 23441, May 28, 1982; 50 
FR 19919, May 13, 1985; 59 FR 8398, Feb. 22, 1994]



Sec. 444.342b  Neomycin sulfate-polymyxin B sulfate-gramicidin ophthalmic solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-polymyxin B sulfate-
gramicidin ophthalmic solution is a solution containing in each 
milliliter, 1.75 milligrams of neomycin, 10,000 units of polymyxin B and 
0.025 milligram of gramicidin, and with one or more suitable and 
harmless buffers, dispersants, and preservatives in a suitable and 
harmless isotonic aqueous vehicle. It is sterile. Its pH is not less 
than 4.7 and not more than 6.0. The neomycin sulfate used conforms to 
the standards prescribed by Sec. 444.42a(a)(1)(i), (vi), and (vii). The 
polymyxin B sulfate used conforms to the standards prescribed by 
Sec. 448.30a(a)(1)(i), (vi), (vii), and (ix) of this chapter. The 
gramicidin used conforms to the standards prescribed by 
Sec. 448.25(a)(1)(i), (iv), (v), and (vi) of this chapter. Each other 
substance used, if its name is recognized in the U.S.P. or N.F., 
conforms to the standards prescribed therefor by such official 
compendium.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. Its expiration date is 12 
months.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, pH, 
and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
pH, residue on ignition, and identity.
    (c) The gramicidin used in making the batch for potency, residue on 
ignition, melting point, crystallinity, and identity.
    (d) The batch for neomycin content, polymyxin content, gramicidin 
content, sterility, and pH.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The gramicidin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (d) The batch:
    (1) For all tests except sterility: A minimum of 7 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation, except that if the 
product is sterilized after filling, a representative sample consisting 
of 10 immediate containers from each sterilizer load. If only 1 
sterilizer load is involved, the sample shall consist of 20 immediate 
containers.
    (e) In case of an initial request for certification, each other 
ingredient used in making the batch: One package of each containing 
approximately 5 grams.

[[Page 732]]

    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 444.42a(b)(1), except prepare the sample as 
follows: Remove an accurately measured portion and dilute with 0.1M 
potassium phosphate buffer, pH 8.0, to the proper prescribed reference 
concentration. The neomycin content is satisfactory if it is not less 
than 90 percent and not more than 130 percent of the number of 
milligrams of neomycin that it is represented to contain.
    (ii) Polymyxin content. Remove an accurately measured portion and 
dilute with 10 percent potassium phosphate buffer, pH 6.0, to a 
reference concentration of 10 units of polymyxin per milliliter. Proceed 
as directed in Sec. 448.30a(b)(1) of this chapter, except add to each 
concentration of the polymyxin standard curve a quantity of neomycin to 
yield the same concentration of neomycin as that present when the sample 
is diluted to contain 10 units of polymyxin per milliliter. The 
polymyxin content is satisfactory if it is not less than 90 percent and 
not more than 130 percent of the number of units of polymyxin that it is 
represented to contain.
    (iii) Gramicidin content. Proceed as directed in Sec. 448.25(b)(1) 
of this chapter, except to prepare the sample for assay remove a 
representative sample with a suitable syringe, place into an appropriate 
volumetric flask, and dilute with alcohol U.S.P. XX to obtain a stock 
solution of convenient concentration. Make proper estimated dilutions in 
alcohol U.S.P. XX to the reference concentration. The gramicidin content 
is satisfactory if it contains not less than 90 percent and not more 
than 130 percent of the number of milligrams of gramicidin that it is 
represented to contain.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) pH. Proceed as directed in Sec. 440.80a(b)(5)(ii) of this 
chapter, using the undiluted sample.
[39 FR 19046, May 30, 1974, as amended at 47 FR 22515, May 25, 1982; 47 
FR 23441, May 28, 1982; 47 FR 23709, June 1, 1982; 50 FR 19919, May 13, 
1985]



Sec. 444.342c  Neomycin sulfate-gramicidin ------------ ophthalmic solution; neomycin sulfate-gramicidin ------------ ophthalmic suspension (the blanks being 

          filled in with the established name(s) of the other 
          ingredient(s) present in accordance with paragraph (a)(1) of 
          this section).

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. The drug is a solution or suspension in a 
suitable and harmless aqueous vehicle containing, in each milliliter, 
the following:
    (i) 2.5 milligrams of neomycin, 0.025 milligram of gramicidin, and 1 
milligram of fluorocortisone acetate; or
    (ii) 2.5 milligrams of neomycin, 0.025 milligram of gramicidin, and 
1.14 milligrams of fluorocortisone hemisuccinate.

It contains suitable and harmless buffers, dispersants, irrigants, and 
preservatives. It is sterile. Its pH is not less than 5.0 nor more than 
7.5. The neomycin sulfate used conforms to the standards prescribed by 
Sec. 444.42a(a)(1)(i), (vi), and (vii). The gramicidin used conforms to 
the standards prescribed by Sec. 448.25(a)(1)(i), (ii), and (vi) of this 
chapter. Each other substance used, if its name is recognized in the 
U.S.P. or N.F., conforms to the standards prescribed therefor by such 
official compendium.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. Its expiration date is 12 
months.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, pH, 
and identity.
    (b) The gramicidin used in making the batch for potency, 
crystallinity, residue on ignition, melting point, and identity.
    (c) The batch for neomycin content, gramicidin content, sterility, 
and pH.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.

[[Page 733]]

    (b) The gramicidin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (c) The batch:
    (1) For all tests except sterility: A minimum of 6 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (d) In case of an initial request for certification, each other 
ingredient used in making the batch: One package of each containing 
approximately 5 grams.
    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 444.342b(b)(1)(i). The neomycin content is 
satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of milligrams of neomycin that it is represented 
to contain.
    (ii) Gramicidin content. Proceed as directed in 
Sec. 444.342b(b)(1)(iii). The content of gramicidin is satisfactory if 
it is not less than 90 percent and not more than 130 percent of the 
number of milligrams of gramicidin that it is represented to contain.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section, except 
use 0.25 milliliter of sample in lieu of 1.0 milliliter.
    (3) pH. Proceed as directed in Sec. 440.80a(b)(5)(ii) of this 
chapter, using the undiluted sample.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985; 59 
FR 8398, Feb. 22, 1994]



Sec. 444.342d  Neomycin sulfate-polymyxin B sulfate ------------ ophthalmic suspension (the blank being filled in with the established name(s) of the other 

          active ingredient(s) present in accordance with paragraph 
          (a)(1) of this section).

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. The drug is a suspension in a suitable 
and harmless aqueous vehicle containing, in each milliliter, neomycin 
sulfate, polymyxin B sulfate, and other active ingredients in the 
following amounts:
    (i) 3.5 milligrams of neomycin, 16,250 units of polymyxin B, and 
either 5 milligrams or 15 milligrams of hydrocortisone acetate; or
    (ii) 5 milligrams of neomycin, 15,000 units of polymyxin B, and 2.5 
milligrams of hydrocortisone; or
    (iii) 3.5 milligrams of neomycin, 10,000 units of polymyxin B, and 
10.0 milligrams of hydrocortisone; or
    (iv) 3.5 milligrams of neomycin, 10,000 units of polymyxin B, and 
5.0 milligrams of prednisolone acetate.

It contains suitable and harmless buffers, dispersants, irrigants, and 
preservatives. It is sterile. Its pH is not less than 5.0 and not more 
than 7.0; except if it contains 10 milligrams per milliliter of hydro-
cortisone, its pH is not less than 4.1 and not more than 7.0. The 
neomycin sulfate used conforms to the standards prescribed by 
Sec. 444.42a(a)(1)(i), (vi), and (vii). The polymyxin B sulfate used 
conforms to the standards prescribed by Sec. 448.30a(a)(1)(i), (vi), 
(vii), and (ix) of this chapter. Each other substance used, if its name 
is recognized in the U.S.P. or N.F., conforms to the standards 
prescribed therefor by such official compendium.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. Its expiration date is 12 
months.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, pH, 
and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
pH, residue on ignition, and identity.
    (c) The batch for neomycin content, polymyxin content, sterility, 
and pH.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch for:
    (1) All tests except sterility: A minimum of 6 immediate containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.

[[Page 734]]

    (d) In case of an initial request for certification, each other 
ingredient used in making the batch: One package of each containing 
approximately 5 grams.
    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 444.42a(b)(1) except prepare the sample as 
follows: Remove an accurately measured representative portion of the 
sample with a suitable syringe, place into an appropriate volumetric 
flask to yield a convenient stock solution. Dilute to volume with 0.1M 
potassium phosphate buffer, pH 8.0. Further dilute with 0.1 M potassium 
phosphate buffer, pH 8.0, to the proper prescribed reference 
concentration. Its content of neomycin is satisfactory if it is not less 
than 90 percent and not more than 130 percent of the number of 
milligrams of neomycin that it is represented to contain.
    (ii) Polymyxin content. Remove an accurately measured representative 
portion with a suitable syringe, dilute to a convenient concentration 
with 10 percent potassium phosphate buffer, pH 6.0. Further dilute to a 
concentration of 10 units of polymyxin per milliliter with 10 percent 
potassium phosphate buffer, pH 6.0, and proceed as directed in 
Sec. 448.30a(b)(1) of this chapter, except add to each concentration of 
the polymyxin standard curve a quantity of neomycin to yield the same 
concentration of neomycin as that present when the sample is diluted to 
contain 10 units of polymyxin per milliliter. Its content of polymyxin 
is satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of units of polymyxin that it is represented to 
contain.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
if the steroid prevents solubilization, use 0.25 milliliter of sample in 
lieu of 1 milliliter and proceed as directed in paragraph (e)(2) of that 
section.
    (3) pH. Proceed as directed in Sec. 440.80a(b)(5)(ii) of this 
chapter, using the undiluted sample.
[39 FR 19045, May 30, 1974, as amended at 42 FR 37975, July 26, 1977; 47 
FR 23441, May 28, 1982; 49 FR 5097, Feb. 10, 1984; 49 FR 34351, Aug. 30, 
1984; 50 FR 19919, May 13, 1985; 59 FR 8399, Feb. 22, 1994]



Sec. 444.342e  Neomycin sulfate ointment; neomycin sulfate- ------------ ointment (the blank being filled in with the established name(s) of certain other 

          active ingredient(s)).

    The requirements for certification and the tests and methods of 
assay for neomycin sulfate ointment and for neomycin sulfate- ----------
-- ointment are described in Sec. 444.542a.



Sec. 444.342f  Neomycin sulfate-gramicidin topical ointment; neomycin sulfate-gramicidin-triamcinolone acetonide ointment; neomycin sulfate-gramicidin-
          fludrocortisone acetate ointment.

    The requirements for certification and the tests and methods of 
assay for neomycin sulfate-gramicidin topical ointment; neomycin 
sulfate-gramicidin-triamcinolone acetonide ointment; neomycin sulfate-
gramicidin-fludrocortisone acetate ointment are described in 
Sec. 444.542f.



Sec. 444.342g  Neomycin sulfate-hydrocortisone acetate ophthalmic suspension; neomycin sulfate-prednisolone acetate ophthalmic suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-hydrocortisone acetate 
ophthalmic suspension is an aqueous suspension containing in each 
milliliter 3.5 milligrams of neomycin and 5 milligrams or 15 milligrams 
of hydrocortisone acetate. Neomycin sulfate-prednisolone acetate 
ophthalmic suspension is an aqueous suspension containing in each 
milliliter 3.5 milligrams of neomycin and 2.5 milligrams of prednisolone 
acetate. The vehicle contains one or more suitable and harmless buffers, 
preservatives, and dispersants. It is sterile. Its

[[Page 735]]

pH is not less than 5.5 and not more than 7.5. The neomycin sulfate used 
conforms to the standards prescribed by Sec. 444.42a(a)(1) (i), (v), 
(vi), and (vii). Each other substance used, if its name is recognized in 
the U.S.P. or N.F., conforms to the standards prescribed therefor by 
such official compendium.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. Its expiration date is 12 
months.
    (3) Requests for certification. In addition to the requirements of 
Sec. 431.1 of this chapter, each such request shall contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, 
moisture, pH, and identity.
    (b) The batch for potency, sterility, and pH.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 5 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (c) In case of an initial request for certification, each other 
ingredient used in making the batch: One package of each containing 
approximately 5 grams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 444.42a(b)(1), except prepare the sample for assay as follows: 
Remove 1.0 milliliter with a suitable syringe, place into an 
appropriate-sized volumetric flask and dilute to volume with 0.1M 
potassium phosphate buffer, pH 8.0, to give a stock solution of 
convenient concentration. Make proper estimated dilutions to the 
prescribed reference concentration with 0.1M potassium phosphate buffer, 
pH 8.0. The content of neomycin is satisfactory if it is not less than 
90 percent and not more than 130 percent of the number of milligrams of 
neomycin that it is represented to contain.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
if the steriod prevents solubilization, use 0.25 milliliter in lieu of 1 
milliliter and proceed as directed in paragraph (e)(2) of that section.
    (3) pH. Proceed as directed in Sec. 440.80a(b)(5)(ii) of this 
chapter, using the undiluted sample.
[39 FR 19045, May 30, 1974, as amended at 39 FR 33666, Sept. 19, 1974; 
50 FR 19919, May 13, 1985]



Sec. 444.342h  Neomycin sulfate-polymyxin B sulfate ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-polymyxin B sulfate 
ophthalmic ointment contains in each gram, neomycin sulfate equivalent 
to 3.5 milligrams of neomycin and polymyxin B sulfate equivalent to 
10,000 units of polymyxin B with suitable preservatives in a suitable 
and harmless ointment base. Its neomycin sulfate content is satisfactory 
if it is not less than 90 percent and not more than 130 percent of the 
number of milligrams of neomycin that it is represented to contain. Its 
polymyxin B sulfate content is satisfactory if it is not less than 90 
percent and not more than 130 percent of the number of milligrams of 
polymyxin B that it is represented to contain. It is sterile. Its 
moisture content is not more than 0.5 percent. It passes the test for 
metal particles. The neomycin sulfate used conforms to the standards 
prescribed by Sec. 444.42a(a)(1) except sterility and pyrogens. The 
polymyxin B sulfate used conforms to the standards prescribed by 
Sec. 448.30a(a)(1) of this chapter except sterility, pyrogens, and heavy 
metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, residue on ignition, and identity.

[[Page 736]]

    (c) The batch for neomycin content, polymyxin B content, sterility, 
moisture, and metal particles.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch:
    (1) For all tests except sterility: A minimum of 16 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Place an accurately weighed representative 
portion of the sample into a separatory funnel containing approximately 
50 milliliters of peroxide-free ether. Shake the sample and ether until 
homogeneous. Add 20 to 25 milliliters of 0.1M potassium phosphate 
buffer, pH 8.0 (solution 3), and shake well. Allow the layers to 
separate. Remove the buffer layer and repeat the extraction procedure 
with each of three more 20- to 25-milliliter quantities of solution 3. 
Combine the buffer extractives in a suitable volumetric flask and dilute 
to volume with solution 3. Remove an aliquot and further dilute with 
solution 3 to the reference concentration of 1.0 microgram of neomycin 
per milliliter (estimated).
    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, except add to each concentration of the polymyxin B 
standard response line a quantity of neomycin to yield the same 
concentration of neomycin as that present when the sample is diluted to 
contain 10 units of polymyxin B per milliliter. Prepare the sample for 
assay as follows: Place an accurately weighed representative portion of 
the sample into a separatory funnel containing approximately 50 
milliliters of peroxide-free ether. Shake the sample and ether until 
homogeneous. Add 20 to 25 milliliters of 10 percent potassium phosphate 
buffer, pH 6.0 (solution 6), and shake well. Allow the layers to 
separate. Remove the buffer layer and repeat the extraction procedure 
with each of three more 20- to 25-milliliter quantities of solution 6. 
Combine the buffer extractives in a suitable volumetric flask and dilute 
to volume with solution 6. Remove an aliquot and further dilute with 
solution 6 to the reference concentration of 10 units of polymyxin B per 
milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) Metal particles. Proceed as directed in Sec. 436.206 of this 
chapter.
[39 FR 19046, May 30, 1974, as amended at 47 FR 23441, May 28, 1982; 50 
FR 19919, May 13, 1985; 55 FR 14969, Apr. 20, 1990]



Sec. 444.342i  Neomycin sulfate-polymyxin B sulfate ophthalmic solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-polymyxin B sulfate 
ophthalmic solution is neomycin sulfate and polymyxin B sulfate in a 
suitable and harmless aqueous vehicle. Each milliliter contains: (i) 
Neomycin sulfate equivalent to 3.5 milligrams of neomycin and polymyxin 
B sulfate equivalent to 10,000 units of polymyxin B; or
    (ii) Neomycin sulfate equivalent to 3.5 milligrams of neomycin and 
polymyxin B sulfate equivalent to 16,250 units of polymyxin B. It 
contains suitable and harmless buffers, dispersants, irrigants, and 
preservatives. Its neomycin sulfate content is satisfactory if it is not 
less than 90 percent and not more than 130 percent of the number of 
milligrams of neomycin that it is represented to contain. Its polymyxin 
B sulfate content is satisfactory if it is not less than 90 percent and 
not more than 130 percent of the number of milligrams of polymyxin B 
that it is represented to contain. It is sterile. Its pH is not less 
than 5.0 and not more than 7.0. The neomycin sulfate used conforms to 
the standards prescribed by Sec. 444.42a(a)(1) except sterility and 
pyrogens. The polymyxin B sulfate used conforms to the standards 
prescribed by Sec. 448.30a(a)(1) of this chapter

[[Page 737]]

except sterility, pyrogens, and heavy metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, 
moisture, pH, and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
moisture, pH, residue on ignition, and identity.
    (c) The batch for neomycin content, polymyxin B content, sterility, 
and pH.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch:
    (1) For all tests except sterility: A minimum of 6 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Place an accurately measured representative 
portion of the sample into an appropriate-sized volumetric flask with 
sufficient 0.1M potassium phosphate buffer, pH 8.0 (solution 3), to give 
a stock solution of convenient concentration. Remove an aliquot and 
further dilute with solution 3 to the reference concentration of 1.0 
microgram of neomycin per milliliter (estimated).
    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, except add to each concentration of the polymyxin B 
standard response line a quantity of neomycin to yield the same 
concentration of neomycin as that present when the sample is diluted to 
contain 10 units of polymyxin B per milliliter. Prepare the sample for 
assay as follows: Place an accurately measured representative portion of 
the sample into an appropriate-sized volumetric flask with sufficient 10 
percent potassium phosphate buffer, pH 6.0 (solution 6), to give a stock 
solution of convenient concentration. Remove an aliquot and further 
dilute with solution 6 to the reference concentration of 10.0 units of 
polymyxin B per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[39 FR 19045, May 30, 1974, as amended at 39 FR 36472, Oct. 10, 1974; 47 
FR 23441, May 28, 1982; 50 FR 19919, May 13, 1985; 55 FR 14969, Apr. 20, 
1990; 59 FR 8399, Feb. 22, 1994]



Sec. 444.342j  Neomycin sulfate-polymyxin B sulfate-dexamethasone ophthalmic suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-polymyxin B sulfate-
dexamethasone ophthalmic suspension is an aqueous suspension containing 
in each milliliter 3.5 milligrams of neomycin, 10,000 units of polymyxin 
B, and 1.0 milligram of dexamethasone. It contains suitable and harmless 
buffers, dispersants, irrigants, and preservatives. Its neomycin sulfate 
content is satisfactory if it is not less than 90 percent and not more 
than 130 percent of the number of milligrams of neomycin that it is 
represented to contain. Its polymyxin B sulfate content is satisfactory 
if it is not less than 90 percent and not more than 130 percent of the 
number of milligrams of polymyxin B that it is represented to contain. 
It is sterile. Its pH is not less than 5.2 and not more than 5.8. The 
neomycin sulfate used conforms to the standards prescribed by 
Sec. 444.42(a)(1). The polymyxin B sulfate used conforms to the 
standards prescribed by Sec. 448.30(a)(1) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:

[[Page 738]]

    (a) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (c) The batch for neomycin content, polymyxin B content, sterility, 
and pH.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages 
each containing approximately 300 milligrams.
    (c) The batch:
    (1) For all tests except sterility: A minimum of 6 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Place an accurately measured representative 
portion of the sample into an appropriate-sized volumetric flask with 
sufficient 0.1 M potassium phosphate buffer, pH 8.0 (solution 3), to 
obtain a stock solution of convenient concentration. Remove an aliquot 
and further dilute with solution 3 to the reference concentration of 1.0 
microgram of neomycin per milliliter (estimated).
    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, except add to each concentration of the polymyxin B 
standard response line a quantity of neomycin to yield the same 
concentration of neomycin as that present when the sample is diluted to 
contain 10 units of polymyxin B per milliliter. Prepare the sample for 
assay as follows: Place an accurately measured representative portion of 
the sample into an appropriate-sized volumetric flask with sufficient 10 
percent potassium phosphate buffer, pH 6.0 (solution 6), to obtain a 
stock solution of convenient concentration. Remove an aliquot and 
further dilute with solution 6 to the reference concentration of 10 
units of polymyxin B per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use 0.25 milliliter in lieu of 1.0 milliliter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[47 FR 23441, May 28, 1982; 47 FR 25320, June 11, 1982, as amended at 50 
FR 19919, May 13, 1985; 55 FR 14969, Apr. 20, 1990; 59 FR 8399, Feb. 22, 
1994]



Sec. 444.342k  Neomycin sulfate-polymyxin B sulfate-dexamethasone ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-polymyxin B sulfate-
dexamethasone ophthalmic ointment contains in each gram neomycin sulfate 
equivalent to 3.5 milligrams of neomycin, polymyxin B sulfate equivalent 
to 10,000 units of polymyxin B and 1.0 milligram of dexamethasone with 
suitable preservatives in a suitable and harmless ointment base. Its 
neomycin sulfate content is satisfactory if it is not less than 90 
percent and not more than 130 percent of the number of milligrams of 
neomycin that it is represented to contain. Its polymyxin B sulfate 
content is satisfactory if it is not less than 90 percent and not more 
than 130 percent of the number of milligrams of polymyxin B that it is 
represented to contain. It is sterile. Its moisture content is not more 
than 0.5 percent. It passes the test for metal particles. The neomycin 
sulfate used conforms to the standards prescribed by Sec. 444.42(a)(1). 
The polymyxin B sulfate used conforms to the standards prescribed by 
Sec. 448.30(a)(1) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.

[[Page 739]]

    (c) The batch for neomycin content, polymyxin B content, moisture, 
and metal particles.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch:
    (1) For all tests except sterility: A minimum of 16 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Place an accurately weighed representative 
portion of the sample into a separatory funnel containing approximately 
50 milliliters of peroxide-free ether. Shake the sample and ether until 
homogeneous. Add 20 to 25 milliliters of 0.1 M potassium phosphate 
buffer, pH 8.0 (solution 3), and shake well. Allow the layers to 
separate. Remove the buffer layer and repeat the extraction procedure 
with each of three more 20- to 25-milliliter quantities of solution 3. 
Combine the buffer extractives in a suitable volumetric flask and dilute 
to volume with solution 3. Remove an aliquot and further dilute with 
solution 3 to the reference concentration of 1.0 microgram of neomycin 
per milliliter (estimated).
    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, except add to each concentration of the polymyxin B 
standard response line a quantity of neomycin to yield the same 
concentration of neomycin as that present when the sample is diluted to 
contain 10 units of polymyxin B per milliliter. Prepare the sample for 
assay as follows: Place an accurately weighed representative portion of 
the sample into a separatory funnel containing approximately 50 
milliliters of peroxide-free ether. Shake the sample and ether until 
homogeneous. Add 20 to 25 milliliters of 10 percent potassium phosphate 
buffer, pH 6.0 (solution 6), and shake well. Allow the layers to 
separate. Remove the buffer layer and repeat the extraction procedure 
with each of three more 20- to 25-milliliter quantities of solution 6. 
Combine the buffer extractives in a suitable volumetric flask and dilute 
to volume with solution 6. Remove an aliquot and further dilute with 
solution 6 to the reference concentration of 10 units of poylmyxin B per 
milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) Metal particles. Proceed as directed in Sec. 436.206 of this 
chapter.
[47 FR 23442, May 28, 1982; 47 FR 25320, June 11, 1982, as amended at 50 
FR 19919, May 13, 1985; 55 FR 14969, Apr. 20, 1990]



Sec. 444.380  Tobramycin ophthalmic dosage forms.



Sec. 444.380a  Tobramycin ophthalmic solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tobramycin ophthalmic solution contains 
in each milliliter 3.0 milligrams of tobramycin in a suitable and 
harmless aqueous vehicle. It contains suitable and harmless buffers, 
dispersants, preservatives, and tonicity agents. Its potency is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of tobramycin that it is represented 
to contain. It is sterile. Its pH is not less than 7.0 and not more than 
8.0. The tobramycin used conforms to the standards prescribed by 
Sec. 444.80(a)(1), except heavy metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tobramycin used in making the batch for potency, moisture, 
pH, identity, and residue on ignition.
    (b) The batch for potency, sterility, and pH.
    (ii) Samples required:

[[Page 740]]

    (a) The tobramycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of five immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dilute an accurately measured representative portion of the sample with 
sufficient distilled water to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of the stock solution with 
distilled water to the reference concentration of 2.5 micrograms of 
tobramycin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[46 FR 16681, Mar. 13, 1981; 46 FR 22359, Apr. 17, 1981. Redesignated at 
47 FR 7827, Feb. 23, 1982, and amended at 50 FR 19919, May 13, 1985; 59 
FR 8399, Feb. 22, 1994]



Sec. 444.380b  Tobramycin ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tobramycin ophthalmic ointment contains, 
in each gram, 3.0 milligrams of tobramycin with a suitable preservative 
in a suitable and harmless ointment base. Its potency is satisfactory if 
it is not less than 90 percent and not more than 120 percent of the 
number of milligrams of tobramycin that it is represented to contain. It 
is sterile. Its moisture content is not more than 1.0 percent. It passes 
the test for metal particles. The tobramycin used conforms to the 
standards prescribed by Sec. 444.80(a)(1), except heavy metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tobramycin used in making the batch for potency, moisture, 
pH, identity, and residue on ignition.
    (b) The batch for potency, sterility, moisture, and metal particles.
    (ii) Samples required:
    (a) The tobramycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the sample into a 
separatory funnel containing approximately 50 milliliters of peroxide-
free ether. Shake the sample and ether until homogeneous. Add 20 to 25 
milliliters of distilled water, and shake well. Allow the layers to 
separate. Remove the distilled water layer and repeat the extraction 
procedure with each of three more 20- to 25-milliliter quantities of 
distilled water. Combine the extractives in a suitable volumetric flask 
and dilute to volume with distilled water. Further dilute an aliquot 
with distilled water to the reference concentration of 2.5 micrograms of 
tobramycin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) Metal particles. Proceed as directed in Sec. 436.206 of this 
chapter.
[47 FR 7827, Feb. 23, 1982; 47 FR 16320, Apr. 16, 1982, as amended at 50 
FR 19919, May 13, 1985]



Sec. 444.380c  Tobramycin-dexamethasone ophthalmic suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tobramycin-dexamethasone ophthalmic 
suspension is an aqueous suspension containing, in each milliliter, 3.0 
milligrams of tobramycin and

[[Page 741]]

1.0 milligram of dexamethasone in a suitable and harmless aqueous 
vehicle. It contains suitable and harmless buffers, dispersants, 
preservatives, and tonicity agents. Its tobramycin potency is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of tobramycin that it is represented 
to contain. Its dexamethasone content is satisfactory if it is not less 
than 90 percent and not more than 110 percent of the number of 
milligrams of dexamethasone that it is represented to contain. It is 
sterile. Its pH is not less than 5.0 and not more than 6.0. It passes 
the identity tests for tobramycin and dexamethasone. The tobramycin used 
conforms to the standards prescribed by Sec. 444.80(a)(1), except heavy 
metals. The dexamethasone used conforms to the standards prescribed by 
the USP XXI.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The tobramycin used in making the batch for potency, moisture, 
pH, identity, and residue on ignition.
    (B) The dexamethasone used in making the batch for all USP XXI 
specifications.
    (C) The batch for tobramycin potency, dexamethasone content, 
sterility, pH, tobramycin identity, and dexamethasone identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The tobramycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Tobramycin potency. Proceed as 
directed in Sec. 436.106 of this chapter, preparing the sample for assay 
as follows: Dilute an accurately measured representative portion of the 
sample with sufficient distilled water to obtain a stock solution of 
convenient concentration. Further dilute an aliquot of the stock 
solution with distilled water to the reference concentration of 2.5 
micrograms of tobramycin per milliliter (estimated).
    (2) Dexamethasone content. Proceed as directed in Sec. 436.216 of 
this chapter, using ambient temperature, an ultraviolet detection system 
operating at a wavelength of 254 nanometers, a column packed with 
microparticulate (3 to 10 micrometers in diameter) reversed phase 
packing material such as octadecyl hydrocarbon bonded silicas, a flow 
rate of 1.5 milliliters per minute, and an injection volume of 20 
microliters. Mobile phase, working standard and sample solutions, system 
suitability requirements, and calculations are as follows:
    (i) Mobile phase. Mix acetonitrile:water (45:55). Filter the mobile 
phase through a suitable glass fiber filter or equivalent that is 
capable of removing particulate contamination to 1 micron in diameter. 
Degas the mobile phase just prior to its introduction into the 
chromatograph pumping system.
    (ii) Preparation of working standard and sample solutions--(A) 
Working standard solution. Accurately weigh approximately 25 milligrams 
of the dexamethasone working standard into a 25-milliliter volumetric 
flask containing methanol. Shake until dissolved. Dilute to volume with 
methanol. Further dilute 4.0 milliliters of this solution to 100 
milliliters with methanol to obtain a solution containing approximately 
40 micrograms of dexamethasone per milliliter. Mix well.
    (B) Sample solutions. Remove an accurately measured representative 
portion from each container. Dilute the solution thus obtained with 
sufficient methanol to obtain a solution containing 40 micrograms of 
dexamethasone per milliliter (estimated).
    (iii) System suitability requirements--(A) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.6 at 10 
percent of peak height in lieu of 5 percent of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory

[[Page 742]]

if it is greater than 5,500 theoretical plates.
    (C) Resolution. The resolution (R) between the peak for 
dexamethasone and its nearest eluting impurity is satisfactory if it is 
not less than 1.1.
    (D) Coefficient of variation. The coefficient of variation 
(SRin percent) of 5 replicate injections is satisfactory if 
it is not more than 2.0 percent. If the system suitability requirements 
have been met, then proceed as described in Sec. 436.216(b) of this 
chapter. Alternate chromatographic conditions are acceptable provided 
reproducibility and resolution are comparable to the system. However, 
the sample preparation described in paragraph(b)(2)(ii)(B) of this 
section should not be changed.
    (iv) Calculations. Calculate the dexamethasone content of the 
container as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.228

where:
    Au=Area of the dexamethasone peak in the chromatogram of 
the sample (at a retention time equal to that observed for the 
standard);
    As=Area of the dexamethasone peak in the chromatogram of 
the dexamethasone working standard;
    Ps=Dexamethasone content in the dexamethasone working 
standard solution in micrograms per milliliter; and
    d=Dilution factor of the sample.

    (3) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (5) Tobramycin identity. Proceed as directed in Sec. 436.318 of this 
chapter, except prepare the sample for assay as follows; decant 1 
milliliter into a test tube. Add 100 milligrams of sodium sulfate to the 
test tube and shake until the sodium sulfate has been dispersed. 
Centrifuge to obtain a clear supernatant. Use the supernatant as the 
sample solution.
    (6) Dexamethasone identity. The high-pressure liquid chromatogram of 
the sample determined as directed in paragraph (b)(2) of this section, 
compares qualitatively to that of the dexamethasone working standard.
[54 FR 13879, Apr. 6, 1989, as amended at 59 FR 8399, Feb. 22, 1994]



Sec. 444.380d  Tobramycin-dexamethasone ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tobramycin-dexamethasone ophthalmic 
ointment contains in each gram, tobramycin equivalent to 3.0 milligrams 
of tobramycin and 1.0 milligram of dexamethasone, with a suitable 
preservative in a suitable and harmless white petrolatum base. Its 
tobramycin potency is satisfactory if it is not less than 90 percent and 
not more than 120 percent of the number of milligrams of tobramycin that 
it is represented to contain. Its dexamethasone content is satisfactory 
if it is not less than 90 percent and not more than 110 percent of the 
number of milligrams of dexamethasone that it is represented to contain. 
It is sterile. Its moisture content is not more than 1.0 percent. It 
passes the test for metal particles. It passes the identity tests for 
tobramycin and dexamethasone. The tobramycin used conforms to the 
standards prescribed by Sec. 444.80(a)(1), except heavy metals. The 
dexamethasone used conforms to the standards prescribed by the U.S. 
Pharmacopeia XXII.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The tobramycin used in making the batch for potency, moisture, 
pH, identity, and residue on ignition.
    (B) The dexamethasone used in making the batch for all U.S. 
Pharmacopeia XXII specifications.
    (C) The batch for tobramycin potency, dexamethasone content, 
sterility, moisture, metal particles, tobramycin identity, and 
dexamethasone identity.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research:

[[Page 743]]

    (A) The tobramycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Tobramycin potency. Proceed as 
directed in Sec. 436.106 of this chapter, preparing the sample for assay 
as follows: Place an accurately weighed representative portion of the 
sample into a separatory funnel containing approximately 50 milliliters 
of peroxide-free ether. Shake the sample and ether until homogeneous. 
Add 20 to 25 milliliters of distilled water and shake well. Allow the 
layers to separate. Remove the distilled water layer and repeat the 
extraction procedure with each of three more 20- to 25-milliliter 
quantities of distilled water. Combine the extractives in a suitable 
volumetric flask and dilute to volume with distilled water. Further 
dilute an aliquot with distilled water to the reference concentration of 
2.5 micrograms of tobramycin per milliliter (estimated).
    (2) Dexamethasone content. Proceed as directed in Sec. 436.216 of 
this chapter, using ambient temperature, an ultraviolet detection system 
operating at a wavelength of 254 nanometers, a column packed with 
microparticulate (3 to 10 micrometers in diameter) reversed phase 
packing material such as octadecyl hydrocarbon bonded silicas, a flow 
rate of 1.5 milliliters per minute, and an injection volume of 20 
microliters. Mobile phase, working standard and sample solutions, system 
suitability requirements, and calculations are as follows:
    (i) Mobile phase. Mix acetonitrile:water (45:55) and adjust if 
necessary by reducing the amount of acetonitrile to increase retention, 
or by increasing the amount of acetonitrile to decrease the retention of 
the solute. Filter the mobile phase through a suitable glass fiber 
filter or equivalent that is capable of removing particulate 
contamination to 1 micron in diameter. Degas the mobile phase just prior 
to its introduction into the chromatograph pumping system.
    (ii) Preparation of working standard and sample solutions and 
resolution test solution--(A) Working standard solution. Accurately 
weigh approximately 20 milligrams of the dexamethasone working standard 
into a 100-milliliter volumetric flask containing methonol:water 
(75:25). Shake until dissolved. Dilute to volume with methanol:water 
(75:25). Transfer 10.0 milliliters of this solution to a separatory 
funnel containing approximately 50 milliliters of hexane. Shake until 
homogeneous. Add 15.0 milliliters of methanol:water (75:25) and shake 
well. Allow the layers to separate. Remove the lower (methanol:water) 
layer and repeat the extraction twice more with 15.0 milliliters of 
methanol:water (75:25). Collect the extractives in a 50-milliliter 
volumetric flask. Dilute to volume with methanol:water (75:25) to obtain 
a solution of known concentration containing approximately 40 micrograms 
of dexamethasone per milliliter.
    (B) Sample solution. Accurately weigh approximately 2.0 grams of the 
sample and place into a separatory funnel containing approximately 50 
milliliters of hexane. Shake until homogeneous. Add 15.0 milliliters of 
methanol:water (75:25) and shake well. Allow the layers to separate. 
Remove the lower (methanol:water) layer and repeat the extraction twice 
more with 15.0 milliliters of methanol:water (75:25). Collect the 
extractives in a 50-milliliter volumetric flask. Dilute to volume with 
methanol:water (75:25) to obtain a solution of known concentration 
containing approximately 40 micrograms of dexamethasone per milliliter 
(estimated).
    (iii) System suitability requirements--(A) Asymmetry. The asymmetry 
(As) is satisfactory if it is not more than 1.6 at 10 percent 
of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 5,500 theoretical plates.
    (C) Resolution. The resolution (Rs) between the peak for 
dexamethasone and its nearest eluting impurity is satisfactory if it is 
not less than 1.1.
    (D) Coefficient of variation. The coefficient of variation (RSD in 
percent) of 5 replicate injections is satisfactory if it

[[Page 744]]

is not more than 2.0 percent. If the system suitability requirements 
have been met, then proceed as described in Sec. 436.216(b) of this 
chapter.
    (iv) Calculations. Calculate the dexamethasone content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.229
    
where:
Au=Area of the dexamethasone peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the dexamethasone peak in the chromatogram of the 
          dexamethasone working standard;
Ps=Dexamethasone content in the dexamethasone working 
          standard solution in micrograms per milliliter;
d=Dilution factor of the sample; and
n=Number of grams of sample assayed.

    (3) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in Sec. 436.20(e)(1).
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) Metal particles. Proceed as directed in Sec. 436.206 of this 
chapter.
    (6) Tobramycin identity. Proceed as directed in Sec. 436.318 of this 
chapter, except prepare the sample for assay as follows: Weigh 
approximately 1 gram of the sample into a test tube. Add 1 to 2 
milliliters of chloroform to the test tube and shake vigorously to 
dissolve the ointment. Centrifuge for approximately 15 minutes to 
clearly separate the layers. Use the top (aqueous) layer in the 
procedure.

    Note: If an oily film remains on the top of the aqueous layer and 
interferes with sampling, the aqueous layer may be transferred to 
another test tube and washed with an additional 1 to 2 milliliters of 
chloroform.

    (7) Dexamethasone identity. The high-performance liquid chromatogram 
of the sample determined as directed in paragraph (b)(2) of this 
section, compares qualitatively to that of the dexamethasone working 
standard.
[55 FR 617, Jan. 8, 1990]



Sec. 444.380e  Tobramycin-fluorometholone acetate ophthalmic suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tobramycin-fluorometholone acetate 
ophthalmic suspension is an aqueous suspension containing, in each 
milliliter, 3.0 milligrams of tobramycin and 1.0 milligram of 
fluorometholone acetate in a suitable and harmless aqueous vehicle. It 
contains one or more suitable and harmless dispersants, preservatives, 
buffers, and tonicity agents. Its tobramycin potency is satisfactory if 
it is not less than 90 percent and not more than 120 percent of the 
number of milligrams of tobramycin that it is represented to contain. 
Its fluorometholone acetate content is satisfactory if it is not less 
than 90 percent and not more than 115 percent of the number of 
milligrams of fluorometholone acetate than it is represented to contain. 
It is sterile. Its pH is not less than 6.0 and not more than 7.0. It 
passes the identity tests for tobramycin and fluorometholone acetate. 
The tobramycin used conforms to the standards prescribed by 
Sec. 440.80(a)(1) of this chapter, except heavy metals. The 
fluorometholone acetate used conforms to the standards prescribed in the 
U.S. Pharmacopeia XXII.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The tobramycin used in making the batch for potency, moisture, 
pH, identity, and residue on ignition.
    (B) The fluorometholone acetate used in making the batch for all 
requirements in U.S. Pharmacopeia XXII.
    (C) The batch for tobramycin potency, fluorometholone acetate 
content, sterility, pH, tobramycin identity, and fluorometholone acetate 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The tobramycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.

[[Page 745]]

    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Tobramycin potency. Proceed as 
directed in Sec. 436.106 of this chapter, preparing the sample for assay 
as follows: Dilute an accurately measured representative portion of the 
sample with sufficient distilled water to obtain a stock solution of 
convenient concentration. Further dilute an aliquot of the stock 
solution with distilled water to the reference concentration of 2.5 
micrograms of tobramycin per milliliter (estimated).
    (2) Fluorometholone acetate content. Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 254 nanometers, a column 
or cartridge packed with microparticulate (3 to 10 micrometers in 
diameter) reversed phase packing material such as octadecyl hydrocarbon 
bonded silicas, a flow rate not to exceed 2.0 milliliters per minute, 
and an injection volume of 10 or 20 microliters. Mobile phase, working 
standard and sample solutions, resolution test solution, system 
suitability requirements, and calculations are as follows:
    (i) Mobile phase. Mix acetonitrile:water (50:50) and adjust if 
necessary by reducing the amount of acetonitrile to increase retention, 
or by increasing the amount of acetonitrile to decrease the retention of 
the solute. Filter the mobile phase through a suitable glass fiber 
filter or equivalent that is capable of removing particulate 
contamination to 1 micron in diameter. Degas the mobile phase just prior 
to its introduction into the chromatograph pumping system.
    (ii) Preparation of working standard and sample solutions, and 
resolution test solution--(A) Working standard solution. Accurately 
weigh approximately 25 milligrams of the fluorometholone acetate working 
standard into a 10-milliliter volumetric flask and add about 5 
milliliters of acetonitrile. Shake until dissolved. Dilute to volume 
with acetonitrile. Further dilute 1.0 milliliter of this solution in a 
volumetric flask to 10 milliliters with acetonitrile to obtain a 
solution of known concentration containing approximately 250 micrograms 
of fluorometholone acetate per milliliter. Mix well.
    (B) Sample solution. Shake vial thoroughly, to homogenize its 
contents, and immediately remove an accurately measured representative 
portion from it. Quantitatively dilute the suspension thus obtained with 
sufficient acetonitrile to obtain a solution containing 250 micrograms 
of fluorometholone acetate per milliliter (estimated). For instance, 
dilute a 1.0 milliliter aliquot of suspension with 3.0 milliliters of 
acetonitrile and filter.
    (C) Resolution test solution. Prepare as directed in paragraph 
(b)(2)(ii)(A) of this section, except use 10 milligrams of 
fluorometholone in addition to the 25 milligrams of fluorometholone 
acetate working standard.
    (iii) System suitability requirements--(A) Asymmetry. The asymmetry 
(AS) is satisfactory if it is not more than 1.35 at 10 
percent of peak height.
    (B) Efficiency of the column. The efficiency of the column 
(hr) is satisfactory if it is not greater than 20, equivalent 
to 1,000 plates for a 10-centimeter column of 5 microns or 2,500 plates 
for a 25-centimeter column of 5 micron size particles.
    (C) Resolution. The resolution (RS) between the peaks of 
fluorometholone acetate and fluorometholone is satisfactory if it is not 
less than 2.0.
    (D) Capacity factor. The capacity factor (k) for fluorometholone 
acetate is satisfactory if it is in the range between 1.0 and 5.0.
     (E) Coefficient of variation. The coefficient of variation (RSD in 
percent) of 5 replicate injections is satisfactory if it is not more 
than 2.0 percent. If the system suitability requirements have been met, 
then proceed as described in Sec. 436.216(b) of this chapter.
    (iv) Calculations. Calculate the fluorometholone acetate content of 
the container as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.230

where:
AU=Area of the fluorometholone acetate peak in the 
          chromatogram of the sample

[[Page 746]]

          (at a retention time equal to that observed for the standard);
AS=Area of the fluorometholone acetate peak in the 
          chromatogram of the fluorometholone acetate working standard;
PS=Fluorometholone acetate content in the fluorometholone 
          acetate working standard solution in micrograms per 
          milliliter; and
d = Dilution factor of the sample.

    (3) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in Sec. 436.20(e)(1).
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted suspension.
    (5) Tobramycin identity. Proceed as directed in Sec. 436.318 of this 
chapter, except prepare the sample for assay as follows: Decant 1.0 
milliliter of the unshaken sample into a test tube. Add 100 milligrams 
of sodium sulfate to the test tube and shake until the sodium sulfate 
has been dispersed. Centrifuge to obtain a clear supernatant. Use the 
supernatant as the sample solution.
    (6) Fluorometholone acetate identity. The high performance liquid 
chromatogram of the sample determined as directed in paragraph (b)(2) of 
this section, compares qualitatively to that of the fluorometholone 
acetate working standard.
[58 FR 26671, May 4, 1993]



                      Subpart E--Otic Dosage Forms



Sec. 444.442  Neomycin sulfate otic dosage forms.



Secs. 444.442a--444.442c  [Reserved]



Sec. 444.442d  Neomycin sulfate ointment; neomycin sulfate- ------------ ointment (the blank being filled in with the established name(s) of certain other 
          active ingredient(s)).

    The requirements for certification and the tests and methods of 
assay for neomycin sulfate ointment and for neomycin sulfate- ----------
-- ointment are described in Sec. 444.542a.



Sec. 444.442e  [Reserved]



Sec. 444.442f  Neomycin sulfate-hydrocortisone-acetic acid otic suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-hydrocortisone-acetic 
acid otic suspension is an aqueous suspension containing in each 
milliliter 5.0 milligrams of neomycin sulfate equivalent to 3.5 
milligrams of neomycin and 10 milligrams of hydrocortisone. It also 
contains 2 percent acetic acid. It may contain one or more suitable and 
harmless buffers, preservatives, and dispersants. Its potency is 
satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of milligrams of neomycin that it is represented 
to contain. It is sterile. Its pH is not less than 4.5 and not more than 
6.0. The neomycin sulfate used conforms to the standards prescribed in 
Sec. 444.42a(a)(1)(i), (v), (vi), and (vii).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (b) The batch for potency, sterility, and pH.
    (ii) Samples required:
    (a) The samples used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 5 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Remove an accurately measured representative portion of the sample and 
dilute with sufficient 0.1M potassium phosphate buffer, pH 8.0 (solution 
3), to give a stock solution of convenient concentration. Further dilute 
with solution 3 to the reference concentration of 1.0 microgram of 
neomycin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
if the steroid prevents solubilization, use 0.25 milliliter in lieu

[[Page 747]]

of 1 milliliter and proceed as directed in paragraph (e)(2) of that 
section.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted suspension.
[39 FR 33668, Sept. 19, 1974, as amended at 46 FR 25608, May 8, 1981]



Sec. 444.442g  Neomycin sulfate-polymyxin B sulfate-hydrocortisone otic suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-polymyxin B sulfate-
hydrocortisone otic suspension contains in each milliliter 3.5 
milligrams neomycin, 10,000 units polymyxin B, and 10 milligrams 
hydrocortisone in a suitable and harmless vehicle. It may also contain 
one or more suitable and harmless buffers, dispersants, and 
preservatives. Its neomycin sulfate content is satisfactory if it is not 
less than 90 percent and not more than 130 percent of the number of 
milligrams of neomycin that it is represented to contain. Its polymyxin 
B sulfate content is satisfactory if it is not less than 90 percent and 
not more than 130 percent of the number of units of polymyxin B that it 
is represented to contain. It is sterile. Its pH is not less than 3.0 
and not more than 7.0. The neomycin sulfate used conforms to the 
standards prescribed by Sec. 444.42(a)(1). The polymyxin B sulfate used 
conforms to the standards prescribed by Sec. 448.30(a)(1) of this 
chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (c) The batch for potency, sterility, and pH.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch:
    (1) For all tests except sterility: A minimum of six immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Dilute an accurately measured 
representative portion with sufficient 0.1M potassium phosphate buffer, 
pH 8.0 (solution 3), to the reference concentration of 1.0 microgram of 
neomycin per milliliter (estimated).
    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, except add to each concentration of the polymyxin B 
standard response line a quantity of neomycin to yield the same 
concentration of neomycin as that present when the sample is diluted to 
contain 10 units of polymyxin B per milliliter. Prepare the sample for 
assay as follows: Dilute an accurately measured representative portion 
with sufficient 10 percent potassium phosphate buffer, pH 6.0 (solution 
6), to the reference concentration of 10 units of polymyxin B per 
milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
if the steroid prevents in lieu of 1 milliliter and proceed as 
disolubilization, use 0.25 milliliter of sample as directed in paragraph 
(e)(2) of that section.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[40 FR 22252, May 22, 1975, as amended at 50 FR 19919, May 13, 1985; 55 
FR 18598, May 3, 1990]



Sec. 444.442h  Neomycin sulfate-polymyxin B sulfate-hydrocortisone otic solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-polymyxin B sulfate-
hydrocortisone otic solution contains in each milliliter 3.5 milligrams 
of neomycin, 10,000 units of

[[Page 748]]

polymyxin B, and 10 milligrams of hydrocortisone in a suitable and 
harmless vehicle. It may also contain one or more suitable and harmless 
buffers, dispersants, and solvents. Its neomycin sulfate content is 
satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of milligrams of neomycin that it is represented 
to contain. Its polymyxin B sulfate content is satisfactory if it is not 
less than 90 percent and not more than 130 percent of the number of 
units of polymyxin B that it is represented to contain. It is sterile. 
The pH is not less than 2.0 and not more than 4.5. The neomycin sulfate 
used conforms to the standards prescribed by Sec. 444.42(a)(1). The 
polymyxin B sulfate used conforms to the standards prescribed by 
Sec. 448.30(a)(1) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (c) The batch for neomycin content, polymyxin B content, sterility, 
and pH.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch:
    (1) For all tests except sterility: A minimum of six immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Dilute an accurately measured 
representative portion of the sample with sufficient 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), to the reference concentration of 
1.0 microgram of neomycin per milliliter (estimated).
    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, except add to each concentration of polymyxin B standard 
response line a quantity of neomycin equal to the amount present when 
the sample is diluted to contain 10 units of polymyxin B per milliliter. 
Prepare the sample for assay as follows: Dilute an accurately measured 
representative portion of the sample with sufficient 10 percent 
potassium phosphate buffer, pH 6.0 (solution 6), to the reference 
concentration of 10 units of polymyxin B per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[41 FR 14186, Apr. 2, 1976; 46 FR 55091, Nov. 6, 1981, as amended at 50 
FR 19919, May 13, 1985]



                  Subpart F--Dermatologic Dosage Forms



Sec. 444.520  Gentamicin sulfate dermatologic dosage forms.



Sec. 444.520a  Gentamicin sulfate ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Gentamicin sulfate ointment is gentamicin 
sulfate with suitable preservatives in a white petrolatum base. Each 
gram contains gentamicin sulfate equivalent to 1.0 milligram of 
gentamicin. Its potency is satisfactory if it is not less than 90 
percent and not more than 135 percent of the number of milligrams of 
gentamicin that it is represented to contain. Its moisture content is 
not more than 1.0 percent. The gentamicin sulfate used conforms to the 
standards prescribed therefor by Sec. 444.20(a)(1).
    (2) Packaging. In addition to the requirements of Sec. 432.1 of this 
chapter, it may be dispensed from a pressurized container wherein it is 
maintained in a compartment separate from the gas used to supply the 
pressure.

[[Page 749]]

    (3) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (4) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The gentamicin sulfate used in making the batch for potency, 
loss on drying, pH, specific rotation, content of gentamicins 
C1, C1a, and C2, and identity.
    (b) The batch for gentamicin potency and moisture.
    (ii) Samples required:
    (a) The gentamicin sulfate used in making the batch: 10 packages, 
each containing not less than 500 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the ointment into 
a separatory funnel containing 50 milliliters of peroxide-free ether. 
Shake the sample and ether until homogeneous, add 20-25 milliliters of 
0.1 M potassium phosphate buffer, pH 8.0 (solution 3), and shake well. 
Allow the layers to separate. Remove the buffer layer and repeat the 
extraction with new portions of the buffer and repeat any additional 
times necessary to insure complete extraction of the antibiotic. Combine 
the extractives and adjust to an appropriate volume to give a stock 
solution of convenient concentration. Further dilute with solution 3 to 
the reference concentration of 0.1 microgram of gentamicin per 
milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985]



Sec. 444.520b  Gentamicin sulfate cream.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Gentamicin sulfate cream is gentamicin 
sulfate with one or more suitable emollients, dispersants, and 
preservatives in a suitable and harmless cream base. Each gram contains 
gentamicin sulfate equivalent to 1.0 milligram of gentamicin. Its 
potency is satisfactory if it is not less than 90 percent and not more 
than 135 percent of the number of milligrams of gentamicin that it is 
represented to contain. The gentamicin sulfate used conforms to the 
standards prescribed therefor by Sec. 444.20(a)(1).
    (2) Packaging. In addition to the requirements of Sec. 432.1 of this 
chapter, it may be dispensed from a pressurized container wherein it is 
maintained in a compartment separate from the gas used to supply the 
pressure.
    (3) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (4) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The gentamicin sulfate used in making the batch for potency, 
loss on drying, pH, specific rotation, gentamicins C1, 
C1a, and C2, and identity.
    (b) The batch for gentamicin potency.
    (ii) Samples required:
    (a) The gentamicin sulfate used in making the batch: 10 packages, 
each containing not less than 500 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay; potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the cream into a 
separatory funnel containing approximately 50 milliliters of peroxide-
free ether. Shake the sample and ether until homogeneous. Add 20 to 25 
milliliters of 0.1M potassium phosphate buffer, ph 8.0 (solution 3), and 
shake gently to avoid gel formation. Allow the layers to separate. 
Remove the buffer layer and repeat the extraction procedure with each of 
three more 20 to 25 milliliter quantities of solution 3. Combine the 
buffer extractives and adjust to an appropriate volume to obtain a stock 
solution of convenient concentration. Further dilute with solution 3 to 
the reference concentration

[[Page 750]]

of 0.1 microgram of gentamicin per milliliter (estimated).
[39 FR 19046, May 30, 1974, as amended at 46 FR 45332, Sept. 11, 1981; 
50 FR 19919, May 13, 1985]



Sec. 444.540  Neomycin palmitate dermatologic dosage forms.



Sec. 444.542  Neomycin sulfate dermatologic dosage forms.



Sec. 444.542a  Neomycin sulfate ointment; neomycin sulfate- ------------ ointment (the blank being filled in with the established name(s) of the other active 
          ingredient(s) present in accordance with paragraph (a)(1) of 
          this section).

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate ointment contains, in 
each gram, 3.5 milligrams of neomycin in a suitable and harmless water-
soluble or oleaginous ointment base, with or without one or more 
suitable and harmless dispersants, emollients, and preservatives. The 
following other drugs may be combined with neomycin sulfate ointment in 
the indicated amounts, per gram:
    (i) If it is for topical use:
    (a) 0.5 milligram of flurandrenolide; or
    (b) 0.25 milligram of fluorometholone; or
    (c) 5.0, 10.0, 15.0, or 25.0 milligrams of hydrocortisone acetate; 
or
    (d) 10.0 or 25.0 milligrams of hydrocortisone; or
    (e) 5.0 milligrams of hydrocortamate hydrochloride; or
    (f) 1.0, 2.5, or 5.0 milligrams of prednisolone acetate; or
    (g) 1.0 milligram of triamcinolone acetonide; or
    (h)-(i) [Reserved]
    (j) 200 milligrams of benzocaine.
    (ii) If it is for ophthalmic use:
    (a) 5.0 milligrams or 15.0 milligrams of hydrocortisone acetate; or
    (b) 2.5 milligrams of sodium prednisolone phosphate; or
    (c) 0.5 milligram of sodium dexamethasone phosphate.
    (d) 1.0 milligram of methyprednisolone; or
    (e) 1.0 milligram of triamcinolone acetonide; or
    (f) 2.5 milligrams or 5.0 milligrams of prednisolone acetate; or
    (g) 15.0 milligrans of cortisone acetate.

If it is an oleaginous base, its moisture content is not more than 1.0 
percent. If it is intended for ophthalmic use, it is sterile. The 
neomycin sulfate used conforms to the standards prescribed by 
Sec. 444.42a(a)(1)(i), (vi), and (vii). Each other substance used, if 
its name is recognized in the U.S.P. or N.F., conforms to the standards 
prescribed therefor by such official compendium.
    (2) Labeling. If it contains a steroid or if it is intended for 
ophthalmic use, it shall be labeled in accordance with the requirements 
prescribed by Sec. 432.5 of this chapter, and its expiration date is 12 
months. If it does not contain a steroid or it is not intended for 
ophthalmic use each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On the label of the immediate container and on the outside 
wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (c) An expiration date that is 12 months after the month during 
which the batch was certified.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, pH, 
and identity.
    (b) The batch for potency and for moisture if the ointment base is 
oleaginous and for sterility if the ointment is intended for ophthalmic 
use.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages each 
containing approximately 300 milligrams.
    (b) The batch:

[[Page 751]]

    (1) For all tests except sterility: A minimum of five immediate 
containers.
    (2) For sterility testing: Twenty immediate containers, collected at 
regular intervals throughout each filling operation.
    (c) In the case of an initial request for certification, each other 
ingredient used in making the batch: One package of each containing 
approximately 5 grams.
    (b) Tests and methods of assay--(1) Potency--(i) Extraction. Proceed 
as directed in Sec. 444.42a(b)(1) of this chapter, except prepare the 
sample by placing an accurately weighed representative portion of the 
ointment into a separatory funnel containing 50 milliliters of peroxide-
free ether. Shake the sample and ether until homogeneous. Add 25 
milliliters of 0.1M potassium phosphate buffer, pH 8.0, and shake well. 
Allow the layers to separate. Remove the buffer layer and repeat the 
extraction with new portions of the buffer at least three times and any 
additional times necessary to insure complete extraction of the 
antibiotic. Combine the extractives and adjust to an appropriate volume 
to give a stock solution of convenient concentration. Further dilute 
with 0.1M potassium phosphate buffer, pH 8.0, to the proper prescribed 
reference concentration.
    (ii) Blending. Proceed as directed in Sec. 444.42a(b)(1), except 
prepare the sample for assay as follows: Transfer an accurately weighed 
sample into a high-speed glass blender, add 1.0 milliliter of 
polysorbate 80 and sufficient 0.1M potassium phosphate buffer, pH 8.0, 
to give a stock solution of convenient concentration. Blend 3 to 5 
minutes. Further dilute with 0.1M potassium phosphate buffer, pH 8.0, to 
the proper prescribed reference concentration. The content of neomycin 
is satisfactory if it is not less than 90 percent and not more than 135 
percent of the number of milligrams of neomycin that it is represented 
to contain.
    (2) Sterility. If the ointment is intended for ophthalmic use, 
proceed as directed in Sec. 436.20 of this chapter, using the method as 
described in paragraph (e)(3) of that section.
    (3) Moisture. If the ointment has an oleaginous base, proceed as 
directed in Sec. 436.201 of this chapter.
[39 FR 19045, May 30, 1974, as amended at 39 FR 33666, Sept. 19, 1974; 
47 FR 23442, May 28, 1982; 49 FR 34351, Aug. 30, 1984; 50 FR 19919, May 
13, 1985; 50 FR 47213, Nov. 15, 1985]



Sec. 444.542b  Neomycin sulfate cream; neomycin sulfate______ cream (the blank being filled in with the established name(s) of the other active ingredient(s) 
          present in accordance with paragraph (a)(1) of this section).

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate cream contains, in each 
gram, 3.5 milligrams of neomycin in a suitable cream base, with or 
without one or more suitable and harmless emollients, perfumes, 
dispersants, and preservatives. The following other drugs may be 
combined with neomycin sulfate cream in the indicated amounts per gram:
    (i) 2 milligrams of betamethasone; or
    (ii) Dexamethasone sodium phosphate equivalent to 1.0 milligram of 
dexamethasone phosphate; or
    (iii) 1 milligram of sodium dexamethasone phosphate; or
    (iv) 2.5 milligrams of dichlorisone acetate; or
    (v) 0.25 milligram of fluocinolone acetonide; or
    (vi) 2.5 milligrams, 5 milligrams, or 10 milligrams of 
methylprednisolone acetate; or
    (vii) 1 milligram of triamcinolone acetonide; or
    (viii) 2.5 milligrams, 5.0 milligrams, or 10.0 milligrams of 
hydrocortisone; or
    (ix) 10.0 milligrams or 25.0 milligrams of hydrocortisone acetate; 
or
    (x) 0.5 milligram of flurandrenolide.

The neomycin sulfate used conforms to the standards prescribed by 
Sec. 444.42a(a)(1) (i), (vi), and (vii). Each other substance used, if 
its name is recognized in the U.S.P. or N.F., conforms to the standards 
prescribed therefor by such official compendium.
    (2) Labeling. If it contains a corticosteroid, it shall be labeled 
in accordance with the requirements prescribed by Sec. 432.5 of this 
chapter, and its expiration date is 12 months. If it does not contain a 
corticosteriod, each

[[Page 752]]

package shall bear on its label or labeling, as hereinafter indicated, 
the following:
    (i) On the label of the immediate container and on the outside 
wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (c) An expiration date that is 12 months after the month during 
which the batch was certified.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, pH, 
and identity.
    (b) The batch for potency.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (c) In case of an initial request for certification, each other 
ingredient used in making the batch: One package of each containing 
approximately 5 grams.
    (b) Tests and methods of assay; potency. Proceed as directed in 
Sec. 444.42a(b)(1), except prepare the sample for assay as follows: 
Transfer an accurately weighed representative portion into a high-speed 
glass blender. Add 1.0 milliliter of polysorbate 80 and sufficient 0.1M 
potassium phosphate buffer, pH 8.0, to give a stock solution of 
convenient concentration and blend 3 to 5 minutes. Further dilute with 
0.1M potassium phosphate buffer, pH 8.0, to the proper prescribed 
reference concentration. Its neomycin content is satisfactory if it is 
not less than 90 percent nor more than 135 percent of the number of 
milligrams of neomycin that it is represented to contain.
[39 FR 19046, May 30, 1974, as amended at 49 FR 34351, Aug. 30, 1984; 53 
FR 18838, May 25, 1988]



Sec. 444.542c  Neomycin sulfate- ------------ lotion (the blank being filled in with the established name(s) of the other active ingredient(s) present in 
          accordance with paragraph (a)(1) of this section).

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. The drug is a suspension containing, in 
each milliliter, 3.5 milligrams of neomycin and the following other 
active ingredients in a suitable and harmless vehicle:
    (i) 10 milligrams of diperodon hydrochloride and 7.5 milligrams of 
aluminum dihydroxy allantoinate; or
    (ii) 5 milligrams or 10 milligrams of hydrocortisone acetate; or
    (iii) 5 milligrams, 10 milligrams, or 20 milligrams of 
hydrocortisone; or
    (iv) 1 milligram, 2.5 milligrams, or 5 milligrams of prednisolone 
acetate; or
    (v) Prednisolone sodium phosphate equivalent to 5.0 milligrams of 
prednisolone phosphate; or
    (vi) 0.5 milligram of flurandrenolide.

It may also contain one or more suitable and harmless dispersants, 
emollients, and preservatives. The neomycin sulfate used conforms to the 
standards prescribed by Sec. 444.42a(a)(1) (i), (vi), and (vii). Each 
other substance used, if its name is recognized in the U.S.P. or N.F., 
conforms to the standards prescribed therefor by such official 
compendium.
    (2) Labeling. If it contains a corticosteroid, it shall be labeled 
in accordance with the requirements prescribed by Sec. 432.5 of this 
chapter and its expiration date is 12 months. If it does not contain a 
corticosteroid, each package shall bear, on its label or labeling, as 
hereinafter indicated, the following:
    (i) On the label of the immediate container and on the outside 
wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.

[[Page 753]]

    (c) An expiration date that is 12 months after the month during 
which the batch was certified.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, pH, 
and identity.
    (b) The batch for potency.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (c) In case of an initial request for certification, each other 
ingredient used in making the batch: One package of each containing 
approximately 5 grams.
    (b) Tests and methods of assay; potency. Proceed as directed in 
Sec. 444.42a(b)(1), except prepare the sample for assay as follows: 
Place an accurately measured representative portion into a high-speed 
glass blender with sufficient 0.1M potassium phosphate buffer, pH 8, to 
give a stock solution of convenient concentration. Blend 3 to 5 minutes. 
Make further dilutions with 0.1M potassium phosphate buffer, pH 8, to 
the proper prescribed reference concentration. Its content of neomycin 
is satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of milligrams of neomycin that it is represented 
to contain.
[39 FR 19046, May 30, 1974, as amended at 49 FR 34351, Aug. 30, 1984]



Sec. 444.542d  [Reserved]



Sec. 444.542e  Neomycin sulfate-polymyxin B sulfate ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-polymyxin B sulfate 
ointment is an ointment containing, in each gram, 3.5 milligrams of 
neomycin and 5,000 units of polymyxin B with suitable and harmless 
emollients, dispersants, and preservatives in a suitable and harmless 
water-miscible base. The neomycin sulfate used conforms to the standards 
prescribed by Sec. 444.42a(a)(1) (i), (vi), and (vii). The polymyxin B 
sulfate used conforms to the standards prescribed by Sec. 448.30a(a)(1) 
(i), (vi), (vii), and (ix) of this chapter. Each other substance used, 
if its name is recognized in the U.S.P. or N.F., conforms to the 
standards prescribed therefor by such official compendium.
    (2) Labeling. Each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On the label of the immediate container and on the outside 
wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (c) An expiration date that is 12 months after the month during 
which the batch was certified.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, pH, 
and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
pH, residue on ignition, and identity.
    (c) The batch for neomycin content and polymyxin content.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch: A minimum of 6 immediate containers.
    (d) In case of an initial request for certification, each other 
ingredient used in making the batch: One package

[[Page 754]]

of each containing approximately 5 grams.
    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 444.542a(b)(1)(ii). Its content of neomycin 
is satisfactory if it is not less than 90 percent and not more than 125 
percent of the number of milligrams of neomycin that it is represented 
to contain.
    (ii) Polymyxin content. Proceed as directed in Sec. 436.105 of this 
chapter, except add to each concentration of the polymyxin standards 
response line a quantity of neomycin to yield the same concentration of 
neomycin as that present when the sample is diluted to contain 10 units 
of polymyxin B per milliliter. Place an accurately weighed 
representative portion of the sample into a separatory funnel containing 
approximately 50 milliliters of peroxide-free ether. Shake the sample 
and ether until homogeneous. Add 20 to 25 milliliters of 10 percent 
potassium phosphate buffer, pH 6.0 (solution 6), and shake well. Remove 
the buffer layer and repeat the extraction procedure with each of three 
more 20 to 25 milliliter quantities of solution 6. Combine the 
extractives in a suitable volumetric flask and fill to volume with 
solution 6. Further dilute an aliquot with solution 6 to the reference 
concentration of 10 units of polymyxin B per milliliter (estimated). Its 
content of polymyxin is satisfactory if it is not less than 90 percent 
and not more than 125 percent of the number of units of polymyxin that 
it is represented to contain.



Sec. 444.542f  Neomycin sulfate-gramicidin topical ointment; neomycin sulfate-gramicidin-triamcinolone acetonide ointment; neomycin sulfate-gramicidin-
          fludrocortisone acetate ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Each gram of neomycin sulfate-gramicidin 
topical ointment contains 2.5 milligrams of neomycin and 0.25 milligram 
of gramicidin. Neomycin sulfate-gramicidin-triamcinolone acetonide 
ointment is an ointment containing, in each gram, 2.5 milligrams of 
neomycin, 0.25 milligram of gramicidin, and 1.0 milligram of 
triamcinolone acetonide. Neomycin sulfate-gramicidin-fludrocortisone 
acetate ointment is an ointment containing, in each gram, 2.5 milligrams 
of neomycin, 0.25 milligram of gramicidin, and 1.0 milligram of 
fludrocortisone acetate. If it is intended for ophthalmic use, it is 
sterile. Their moisture content is not more than 1.0 percent. The 
neomycin sulfate used conforms to the standards prescribed by 
Sec. 444.42a(a)(1) (i), (v), (vi), and (vii), and in addition if it is 
used in the preparation of an ophthalmic ointment, paragraph (a)(1) of 
that section. The gramicidin used conforms to the standards prescribed 
by Sec. 448.25(a)(1) (i), (iii), (iv), (v), and (vi) of this chapter, 
and in addition if it is used in the preparation of an ophthalmic 
ointment, paragraph (a)(1) of that section. Each other substance used, 
if its name is recognized in the U.S.P. or N.F., conforms to the 
standards prescribed therefor by such official compendium.
    (2) Labeling. If it contains a steroid or it is intended for 
ophthalmic use, it shall be labeled in accordance with the requirements 
of Sec. 432.5 of this chapter, and its expiration date is 12 months. If 
it does not contain a steroid or it is not intended for ophthalmic use, 
each package shall bear on its label or labeling, as hereinafter 
indicated, the following:
    (i) On the label of the immediate container and on the outside 
wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (c) An expiration date that is 12 months after the month during 
which the batch was certified.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certifications; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, 
moisture, pH, and identity.

[[Page 755]]

    (b) The gramicidin used in making the batch for potency, moisture, 
residue on ignition, melting point, crystallinity, and identity.
    (c) The batch for neomycin content, gramicidin content, and 
moisture, and for sterility if it is intended for ophthalmic use.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The gramicidin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (c) The batch:
    (1) For all tests except sterility: A minimum of six immediate 
containers.
    (2) For sterility testing: Twenty immediate containers, collected at 
regular intervals throughout each filling operation.
    (d) In case of an initial request for certification, each other 
ingredient used in making the batch: One package of each containing 
approximately 5 grams.
    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 444.542a(b)(1). Its content of neomycin is 
satisfactory if it is not less than 90 percent and not more than 140 
percent of the number of milligrams of neomycin that it is represented 
to contain.
    (ii) Gramicidin content. Proceed as directed in Sec. 448.25(b)(1) of 
this chapter, except prepare the sample for assay by the following 
method: Place an accurately weighed representative portion into a 
separatory funnel. Dissolve the ointment in approximately 50 milliliters 
of petroleum ether. Extract this solution with four 20-milliliter 
portions of 80 percent alcohol prepared from alcohol U.S.P. XX. Combine 
the extractives in a suitable volumetric flask, bring to volume with 
alcohol U.S.P. XX, and mix well. From this stock solution, dilute an 
aliquot with alcohol U.S.P. XX to the reference concentration. Its 
content of gramicidin is satisfactory if it is not less than 90 percent 
and not more that 140 percent of the number of milligrams of gramicidin 
that it is represented to contain.
    (2) Sterility. If the ointment is intended for ophthalmic use, 
proceed as directed in Sec. 436.20 of this chapter, using the method 
described in paragraph (e)(3) of that section. However, if the ointment 
is not soluble in isopropyl myristate proceed as directed in Sec. 436.20 
of this chapter, using the method described in paragraph (e)(2) of that 
section, except use 100 milligrams in lieu of 300 milligrams of solids.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19046, May 30, 1974, as amended at 47 FR 23709, June 1, 1982; 50 
FR 19919, May 13, 1985]



Sec. 444.542g  Neomycin sulfate-gramicidin-triamcinolone acetonide cream.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-gramicidin-triamcinolone 
acetonide cream is a cream containing 2.5 milligrams of neomycin, 0.25 
milligram of gramicidin, and 1.0 milligram of triamcinolone acetonide 
per gram, with one or more suitable and harmless emollients, 
dispersants, and preservatives in a suitable and harmless cream base. 
The neomycin sulfate used conforms to the standards prescribed by 
Sec. 444.42a(a)(1) (i), (vi), and (vii). The gramicidin used conforms to 
the standards prescribed by Sec. 448.25(a)(1) (i), (iv), (v), and (vi) 
of this chapter. Each other substance used, if its name is recognized in 
the U.S.P. or N.F., shall conform to the standards prescribed therefor 
by such official compendium.
    (2) Labeling. It shall be labeled in accordance with the 
requirements prescribed by Sec. 432.5 of this chapter. Its expiration 
date is 12 months.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, pH, 
and identity.
    (b) The gramicidin used in making the batch for potency, residue on 
ignition, melting point, crystallinity and identity.
    (c) The batch for neomycin content and gramicidin content.
    (ii) Samples required:

[[Page 756]]

    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The gramicidin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (c) The batch: A minimum of six immediate containers.
    (d) In case of an initial request for certification, each other 
ingredient used in making the batch: One package of each containing 
approximately 5 grams.
    (b) Tests and methods of assay; potency--(1) Neomycin content. 
Proceed as directed in Sec. 444.542(b). Its neomycin content is 
satisfactory if it is not less than 90 percent and not more than 140 
percent of the number of milligrams of neomycin that it is represented 
to contain.
    (2) Gramicidin content. Proceed as directed in Sec. 448.25(b)(1) of 
this chapter, except to prepare the sample for assay proceed as follows: 
Place an accurately weighed representative portion into a high-speed 
glass blender jar and add sufficient alcohol U.S.P. XX to obtain a stock 
solution of convenient concentration. Blend 3 to 5 minutes. Make proper 
estimated dilutions of an aliquot to the reference concentration with 
alcohol U.S.P. XX. Its content of gramicidin is satisfactory if it is 
not less than 90 percent and not more than 140 percent of the number of 
milligrams of gramicidin that it is represented to contain.
[39 FR 19045, May 30, 1974, as amended at 41 FR 10886, Mar. 15, 1976; 47 
FR 23709, June 1, 1982]



Sec. 444.542h  Neomycin sulfate-gramicidin-triamcinolone acetonide lotion; neomycin sulfate-gramicidin-fludrocortisone acetate lotion.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality and purity. The drug is a lotion containing, in each 
milliliter, 2.5 milligrams of neomycin, 0.25 milligram of gramicidin, 
and either 1 milligram of triamcinolone acetonide or 1 milligram of 
fludrocortisone acetate, with one or more suitable and harmless 
emollients, buffers, dispersants, and preservatives, in a suitable and 
harmless lotion base. The neomycin sulfate used conforms to the 
standards prescribed by Sec. 444.42a(a)(1)(i), (vi), and (vii). The 
gramicidin used conforms to the standards prescribed by 
Sec. 448.25(a)(1)(i), (iv), (v), and (vi) of this chapter. Each other 
substance used, if its name is recognized in the U.S.P. or N.F., 
conforms to the requirements prescribed therefor by such official 
compendium.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. Its expiration date is 12 
months.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, pH, 
and identity.
    (b) The gramicidin used in making the batch for potency, 
crystallinity, residue on ignition, melting point, and identity.
    (c) The batch for neomycin content and gramicidin content.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The gramicidin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (c) The batch: A minimum of 6 immediate containers.
    (d) In case of an initial request for certification, each other 
substance used in making the batch: One package of each containing 
approximately 5 grams.
    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 444.542c(b). Its neomycin content is 
satisfactory if it is not less than 90 percent and not more than 140 
percent of the number of milligrams of neomycin that it is represented 
to contain.
    (ii) Gramicidin content. Proceed as directed in Sec. 448.25(b)(1) of 
this chapter, except prepare the sample by placing an accurately 
measured representative portion into a high-speed glass blender jar with 
sufficient alcohol U.S.P. XX to obtain a stock solution of convenience 
concentration. Blend 3 to 5 minutes.

[[Page 757]]

Make proper estimated dilutions in alcohol U.S.P. XX to the reference 
concentration. Its gramicidin content is satisfactory if it is not less 
than 90 percent and not more than 140 percent of the number of 
milligrams of gramicidin that it is represented to contain.
[39 FR 19045, May 30, 1974, as amended at 41 FR 10886, Mar. 15, 1976; 47 
FR 23709, June 1, 1982]



Sec. 444.542i  [Reserved]



Sec. 444.542j  Neomycin sulfate-polymyxin B sulfate-gramicidin-benzocaine ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-polymyxin B sulfate-
gramicidin-benzocaine ointment is neomycin sulfate, polymyxin B sulfate, 
gramicidin, and benzocaine, with suitable and harmless preservatives, in 
white petrolatum. Each gram contains 3.5 milligrams of neomycin, 2,000 
units of polymyxin B, 0.25 milligram of gramicidin, and 10 milligrams of 
benzocaine. The moisture content is not more than 1.0 percent. The 
neomycin sulfate used conforms to the standards prescribed by 
Sec. 444.42a(a)(1) (i), (v), (vi), and (vii). The polymyxin B sulfate 
used conforms to the standards prescribed by Sec. 448.30a(a)(1) (i), 
(v), (vi), (vii), and (ix) of this chapter. The gramicidin used conforms 
to the standards prescribed by Sec. 448.25(a)(1)(i), (iii), (iv), (v), 
and (vi) of this chapter. Each other ingredient used, if its name is 
recognized in the U.S.P. or N.F., conforms to the standards prescribed 
therefor by such official compendium.
    (2) Labeling. Each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On the label of the immediate container and on the outside 
wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (c) An expiration date that is 12 months after the month during 
which the batch was certified.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, 
moisture, pH, and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
moisture, pH, residue on ignition, and identity.
    (c) The gramicidin used in making the batch for potency, moisture, 
residue on ignition, melting point, crystallinity, and identity.
    (d) The batch for neomycin content, polymyxin B content, gramicidin 
content, and moisture.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The gramicidin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (d) The batch: A minimum of seven immediate containers.
    (e) In case of an initial request for certification, each other 
ingredient used in making the batch: One package of each containing 
approximately 5 grams.
    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 444.542a(b)(1)(i) or (ii). The content of 
neomycin is satisfactory if it is not less than 90 percent and not more 
than 120 percent of the number of milligrams of neomycin that it is 
represented to contain.
    (ii) Polymyxin B content. Proceed as directed in 
Sec. 444.542e(b)(1)(ii) of this chapter. The content of polymyxin B is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of units of polymyxin B that it is represented to 
contain.
    (iii) Gramicidin content. Proceed as directed in Sec. 448.25(b)(1) 
of this chapter, except prepare the sample for assay as follows: Place 
approximately 1 gram of

[[Page 758]]

the ointment, accurately weighed, into a high-speed glass blender. Add 
that quantity of alcohol U.S.P. XX which is sufficient to obtain a stock 
solution of convenient concentration. Blend 3 to 5 minutes. Make proper 
estimated dilutions of an aliquot to the reference concentration with 
alcohol U.S.P. XX. The content of gramicidin is satisfactory if it is 
not less than 90 percent and not more than 120 percent of the number of 
milligrams of gramicidin that it is represented to contain.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19046, May 30, 1974, as amended at 47 FR 23710, June 1, 1982]



Sec. 444.542k  Neomycin sulfate-polymyxin B sulfate-hydrocortisone acetate cream.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-polymyxin B sulfate-
hydrocortisone acetate cream contains, in each gram, neomycin sulfate 
equivalent to 3.5 milligrams of neomycin, polymyxin B sulfate equivalent 
to 10,000 units of polymyxin B, and 5.0 milligrams of hydrocortisone 
acetate in a suitable and harmless vehicle. Its neomycin sulfate content 
is satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of milligrams of neomycin that it is represented 
to contain. Its polymyxin B sulfate content is satisfactory if it is not 
less than 90 percent and not more than 130 percent of the number of 
units of polymyxin B that it is represented to contain. The neomycin 
sulfate used conforms to the standards prescribed by Sec. 444.42(a)(1). 
The polymyxin B sulfate used conforms to the standards prescribed by 
Sec. 448.30(a)(1) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identify.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (c) The batch for neomycin content and polymyxin B content.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay; potency--(1) Neomycin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Transfer an accurately weighed 
representative portion of the sample into a high-speed glass blender jar 
containing 1.0 milliliter polysorbate 80 and sufficient 0.1 M potassium 
phosphate buffer, pH 8.0 (solution 3), to obtain a stock solution of 
convenient concentration. Blend for 3 to 5 minutes. Dilute an aliquot of 
the stock solution with solution 3 to the reference concentration of 1.0 
microgram of neomycin per milliliter (estimated).
    (2) Polymyxin B content. Proceed as directed in Sec. 436.105 of this 
chapter, except add to each concentration of the polymyxin B standard 
response line a quantity of neomycin to yield the same concentration of 
neomycin as that present when the sample is diluted to contain 10 units 
of polymyxin B per milliliter. Prepare the sample for assay as follows: 
Transfer an accurately weighed representative portion of the sample into 
a high-speed glass blender jar containing 1.0 milliliter polysorbate 80 
and sufficient 10 percent potassium phosphate buffer, pH 6.0 (solution 
6), to obtain a stock solution of convenient concentration. Blend for 3 
to 5 minutes. Dilute an aliquot of the stock solution with solution 6 to 
the reference concentration of 10 units of polymyxin B per milliliter 
(estimated).
[50 FR 15108, Apr. 17, 1985, as amended at 55 FR 11584, Mar. 29, 1990]

[[Page 759]]



Sec. 444.542l  Neomycin sulfate-polymyxin B sulfate cream.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate-polymyxin B sulfate 
cream is a cream containing, in each gram, neomycin sulfate equivalent 
to 3.5 milligrams of neomycin and polymyxin B sulfate equivalent to 
10,000 units of polymyxin B in a suitable and harmless vehicle. It may 
contain a suitable local anesthetic. Its neomycin sulfate content is 
satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of milligrams of neomycin that it is represented 
to contain. Its polymyxin B sulfate content is satisfactory if it is not 
less than 90 percent and not more than 130 percent of the number of 
units of polymyxin B that it is represented to contain. The neomycin 
sulfate used conforms to the standards prescribed by Sec. 444.42(a)(1). 
The polymyxin B sulfate used conforms to the standards prescribed by 
Sec. 448.30(a)(1) of this chapter.
    (2) Labeling--(i) On the label of the immediate container and on the 
outside wrapper or container, if any:
    (a) The batch mark;
    (b) The name and quantity of each active ingredient contained in the 
drug; and
    (c) An expiration date that conforms to the requirements prescribed 
by Sec. 432.5(a)(3) of this chapter.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (c) The batch for neomycin content and polymyxin B content.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay; potency--(1) Neomycin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Transfer an accurately weighed 
representative portion of the sample into a high-speed glass blender jar 
containing 1.0 milliliter polysorbate 80 and sufficient 0.1 M potassium 
phosphate buffer, pH 8.0 (solution 3), to obtain a stock solution of 
convenient concentration. Blend for 3 to 5 minutes. Dilute an aliquot of 
the stock solution with solution 3 to the reference concentration of 1.0 
microgram of neomycin per milliliter (estimated).
    (2) Polymyxin B content. Proceed as directed in Sec. 436.105 of this 
chapter, except add to each concentration of the polymyxin B standard 
response line a quantity of neomycin to yield the same concentration of 
neomycin as that present when the sample is diluted to contain 10 units 
of polymyxin B per milliliter. Prepare the sample for assay as follows: 
Transfer an accurately weighed portion of the sample into a high-speed 
glass blender jar containing 1.0 milliliter polysorbate 80 and 
sufficient 10 percent potassium phosphate buffer, pH 6.0 (solution 6), 
to obtain a stock solution of convenient concentration. Blend for 3 to 5 
minutes. Dilute an aliquot of the stock solution with solution 6 to the 
reference concentration of 10 units of polymyxin B per milliliter 
(estimated).
[50 FR 15109, Apr. 17, 1985, as amended at 55 FR 11584, Mar. 29, 1990; 
55 FR 50173, Dec. 5, 1990]

[[Page 760]]



                        Subparts G-I  [Reserved]



                  Subpart J--Certain Other Dosage Forms



Sec. 444.942  Neomycin sulfate in certain other dosage forms.



Sec. 444.942a  Neomycin sulfate for compounding oral products.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Neomycin sulfate for compounding oral 
products is the sulfate salt of a kind of neomycin or a mixture of two 
or more such salts. It is so purified and dried that:
    (i) It has a potency of not less than 600 micrograms of neomycin per 
milligram.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 8 percent.
    (iv) Its pH is an aqueous solution containing 33 milligrams per 
milliliter is not less than 5.0 nor more than 7.5.
    (v) It gives a positive identity test for neomycin.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. Each such container shall contain not less than 10 
grams and not more than 100 grams of neomycin sulfate.
    (3) Labeling. It shall be labeled in accordance with the 
requirements prescribed by Sec. 432.5(a) of this chapter. Its expiration 
date is 12 months.
    (4) Requests for certification; samples. (i) In addition to 
complying with the conditions of Sec. 431.1 of this chapter, a person 
who requests certification of a batch of neomycin sulfate for 
compounding oral products shall submit with the request a statement 
showing the batch mark, the number of packages of each size in the 
batch, and the date on which the latest assay of the drug comprising 
such batch was completed. Such request shall be accompanied or followed 
by results of tests and assays made on the batch for potency, moisture, 
pH, and identity.
    (ii) Such person shall submit with his request a sample consisting 
of a 0.5 gram portion for each 5,000 packages in the batch, but in no 
case less than 10 such portions. Each such portion shall be collected at 
such intervals throughout the entire time of packaging the batch that 
the quantities packaged during the intervals are approximately equal.
    (b) Tests and methods of assay; potency, moisture, pH, and identity. 
Proceed as directed in Sec. 444.42a(b) (1), (5), (6), and (7).
[39 FR 19046, May 30, 1974, as amended at 50 FR 19919, May 13, 1985; 53 
FR 12658, Apr. 15, 1988; 53 FR 31837, Aug. 22, 1988]



Sec. 444.942b  Sterile neomycin sulfate and polymyxin B sulfate solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile neomycin sulfate and polymyxin B 
sulfate solution is an aqueous solution containing in each milliliter 40 
milligrams of neomycin and 200,000 units of polymyxin B. If packaged in 
a multiple-dose container, it shall contain a suitable and harmless 
preservative. It is sterile. Its pH is not less than 4.5 and not more 
than 6.0, except that for issuance of a certificate it is not less than 
5.0. The neomycin sulfate used conforms to the standards prescribed by 
Sec. 444.42a(a)(1) (i), (vi), and (vii). The polymyxin B sulfate used 
conforms to the standards prescribed by Sec. 448.30a(a)(1) (i), (vi), 
(vii), and (ix) of this chapter. Each other substance used, if its name 
is recognized in the U.S.P. or the N.F., conforms to the standards 
prescribed therefor by such official compendium.
    (2) Labeling. In addition to being labeled in accordance with the 
requirements of Sec. 432.5 of this chapter, the labeling shall include a 
statement to the effect that the drug is to be diluted for use as a 
urinary bladder irrigant and is not for injection. Its expiration date 
is 12 months.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The neomycin sulfate used in making the batch for potency, pH, 
and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
pH, residue on ignition, and identity.

[[Page 761]]

    (c) The batch for neomycin content, polymyxin B content, pH, and 
sterility.
    (ii) Samples required:
    (a) The neomycin sulfate used in making the batch: Ten packages, 
each containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: Ten packages, 
each containing approximately 300 milligrams
    (c) The batch:
    (1) For all tests except sterility: A minimum of six immediate 
containers.
    (2) For sterility testing: Twenty immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Neomycin content. 
Proceed as directed in Sec. 444.42a(b)(1), except prepare the sample as 
follows: Remove an accurately measured portion and dilute with 0.1M 
potassium phosphate buffer, pH 8.0, to the proper prescribed reference 
concentration. The neomycin content is satisfactory if it is not less 
than 90 percent nor more than 130 percent of the number of milligrams of 
neomycin that it is represented to contain.
    (ii) Polymyxin B content. Remove an accurately measured portion and 
dilute with 10-percent potassium phosphate buffer, pH 6.0, to a 
reference concentration of 10 units of polymyxin B per milliliter. 
Proceed as directed in Sec. 448.30a(b)(1) of this chapter, except add to 
each concentration of the polymyxin B standard curve a quantity of 
neomycin to yield the same concentration of neomycin as that present 
when the sample is diluted to contain 10 units of polymyxin B per 
milliliter. The polymyxin B content is satisfactory if it is not less 
than 90 percent nor more than 130 percent of the number of units of 
polymyxin B that it is represented to contain.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) pH. Proceed as directed in Sec. 440.80a(b)(5)(ii) of this 
chapter, using the undiluted sample.
[39 FR 19045, May 30, 1974, as amended at 41 FR 56307, Dec. 28, 1976; 42 
FR 18059, Apr. 5, 1977; 50 FR 19919, May 13, 1985]



PART 446--TETRACYCLINE ANTIBIOTIC DRUGS--Table of Contents




                          Subpart A--Bulk Drugs

Sec.
446.10  Chlortetracycline hydrochloride.
446.10a  Sterile chlortetracycline hydrochloride.
446.15  Demeclocycline.
446.16  Demeclocycline hydrochloride.
446.20  Doxycycline hyclate.
446.20a  Sterile doxycycline hyclate.
446.21  Doxycycline monohydrate.
446.42  Meclocycline sulfosalicylate.
446.50  Methacycline hydrochloride.
446.60  Minocycline hydrochloride.
446.65  Oxytetracycline.
446.65a  Sterile oxytetracycline.
446.66  Oxytetracycline calcium.
446.67  Oxytetracycline hydrochloride.
446.67a  Sterile oxytetracycline hydrochloride.
446.75a  Sterile rolitetracycline.
446.76a  Sterile rolitetracycline nitrate.
446.80  Tetracycline.
446.81  Tetracycline hydrochloride.
446.81a  Sterile tetracycline hydrochloride.
446.82  Tetracycline phosphate complex.

                      Subpart B--Oral Dosage Forms

446.110  Chlortetracycline hydrochloride capsules.
446.115  Demeclocycline oral dosage forms.
446.115a  Demeclocycline oral suspension.
446.115b  Demeclocycline for oral suspension.
446.116  Demeclocyline hydrochloride oral dosage forms.
446.116a  Demeclocycline hydrochloride tablets.
446.116b  [Reserved]
446.116c  Demeclocycline hydrochloride capsules.
446.120  Doxycycline hyclate oral dosage forms.
446.120a  Doxycycline hyclate capsules.
446.120b  Doxycycline calcium oral suspension.
446.120c  Doxycycline hyclate tablets.
446.120d  Doxycycline hyclate pellet-filled capsules.
446.121  Doxycycline monohydrate oral dosage forms.
446.121a  Doxycycline monohydrate for oral suspension.
446.121b  Doxycycline monohydrate capsules.
446.150  Methacycline hydrochloride oral dosage forms.
446.150a  Methacycline hydrochloride capsules.
446.150b  Methacycline hydrochloride oral suspension.
446.160  Minocycline hydrochloride oral dosage forms.
446.160a  Minocycline hydrochloride tablets.

[[Page 762]]

446.160b  Minocycline hydrochloride capsules.
446.160c  Minocycline hydrochloride oral suspension.
446.165  Oxytetracycline oral dosage forms.
446.165a  Oxytetracycline tablets.
446.165b--446.165c  [Reserved]
446.165d  Oxytetracycline for oral suspension.
446.166  Oxytetracycline calcium oral suspension.
446.167  Oxytetracycline hydrochloride capsules.
446.180  Tetracycline oral dosage forms.
446.180a--446.180b  [Reserved]
446.180c  Tetracycline oral suspension.
446.181  Tetracycline hydrochloride oral dosage forms.
446.181a--446.181c  [Reserved]
446.181d  Tetracycline hydrochloride tablets.
446.181e  Tetracycline hydrochloride capsules.
446.182  Tetracycline phosphate complex capsules.

                   Subpart C--Injectable Dosage Forms

446.220  Doxycycline hyclate for injection.
446.260  Sterile minocycline hydrochloride.
446.265  Oxytetracycline injection.
446.267  Oxytetracycline hydrochloride for injection.
446.275  Rolitetracycline injectable dosage forms.
446.275a  Rolitetracycline for intravenous use.
446.275b  Rolitetracycline for intramuscular use.
446.276  Rolitetracycline nitrate injectable dosage forms.
446.276a  Rolitetracycline nitrate for intravenous use.
446.276b  Rolitetracycline nitrate for intramuscular use.
446.281  Tetracycline hydrochloride injectable dosage forms.
446.281a  Sterile tetracycline hydrochloride.
446.281c  Tetracycline hydrochloride for intramuscular use.
446.281d  Tetracycline hydrochloride for intravenous use.
446.282  Tetracycline phosphate complex for injection.

                   Subpart D--Ophthalmic Dosage Forms

446.310  Chlortetracycline hydrochloride ophthalmic ointment.
446.367  Oxytetracycline hydrochloride ophthalmic dosage forms.
446.367c  Oxytetracycline hydrochloride-hydrocortisone acetate 
          ophthalmic suspension.
446.367e  Oxytetracycline hydrochloride-polymyxin B sulfate ophthalmic 
          ointment.
446.381  Tetracycline hydrochloride ophthalmic dosage forms.
446.381a  Tetracycline hydrochloride ophthalmic ointment.
446.381b  Tetracycline hydrochloride ophthalmic suspension.

                      Subpart E--Otic Dosage Forms

446.467  Oxytetracycline hydrochloride-polymyxin B sulfate otic 
          ointment.

                  Subpart F--Dermatologic Dosage Forms

446.510  Chlortetracycline hydrochloride ointment.
446.542  Meclocycline sulfosalicylate cream.
446.567  Oxytetracycline hydrochloride dermatologic dosage forms.
446.567a  [Reserved]
446.567b  Oxytetracycline hydrochloride-polymyxin B sulfate topical 
          ointment.
446.567c  Oxytetracycline hydrochloride-polymyxin B sulfate topical 
          powder.
446.581  Tetracycline hydrochloride dermatologic dosage forms.
446.581a--446.581b  [Reserved]
446.581c  Tetracycline hydrochloride for topical solution.
446.581d  Tetracycline hydrochloride ointment.

                     Subpart G--Vaginal Dosage Forms

446.667  Oxytetracycline hydrochloride-polymyxin B sulfate vaginal 
          tablets.

               Subpart H--Rectal Dosage Forms  [Reserved]

                          Subpart I  [Reserved]

            Subpart J--Certain Other Dosage Forms  [Reserved]

    Authority:  21 U.S.C. 357.

    Source:  39 FR 19076, May 30, 1974, unless otherwise noted.



                          Subpart A--Bulk Drugs



Sec. 446.10  Chlortetracycline hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chlortetracycline hydrochloride is [4S - 
(4,4a,5a,6, 12a] - 7 - 
chloro - 4 - (dimethylamino) - 1,4, 4a,5,5a,6,11,12a - octahydro - 
3,6,10,12, 12a - pentahydroxy - 6 - methyl - 1,11 - dioxo - 2 - 
naphthacenecarboxamidemonohydro-chloride. Chlortetracycline is produced 
by the growth of Streptomyces

[[Page 763]]

aureofaciens. It is a yellow powder. It is so purified and dried that:
    (i) Its potency is not less than 900 micrograms per milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 2.0 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 2.3 and not more than 3.3.
    (v) It is crystalline.
    (vi) It meets the identity test for chlortetracycline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, crystallinity, and identity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.01N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of chlortetracycline 
hydrochloride per milliliter (estimated). Further dilute an aliquot of 
the stock solution with sterile distilled water to the reference 
concentration of 0.06 microgram of chlortetracycline hydrochloride per 
milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as di- rected in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (6) Identity. To 1 milligram of sample, add 2.0 milliliters of 
concentrated sulfuric acid. In the presence of chlortetracycline, a deep 
blue color is produced that becomes dark green.
[43 FR 11154, Mar. 17, 1978; 43 FR 34456, Aug. 4, 1978, as amended at 50 
FR 19920, May 13, 1985]



Sec. 446.10a  Sterile chlortetracycline hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chlortetracycline hydrochloride is [4S - 
(4,4a,5a,6,12a)] - 7 - 
chloro - 4 - (dimethylamino) - 1,4,4a,5,5a,6,11,12a - octahydro - 
3,6,10,12,12a - pentahydroxy - 6 - methyl - 1,11 - dioxo - 2 - 
naphthacenecarboxamide monohydrochloride. Chlortetracycline is produced 
by the growth of Streptomyces aureofaciens. It is a yellow powder. It is 
so purified and dried that:
    (i) Its potency is not less than 900 micrograms per milligram.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) It contains no depressor substances.
    (vi) Its loss on drying is not more than 2.0 percent.
    (vii) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 2.3 and not more than 3.3.
    (viii) It is crystalline.
    (ix) It meets the identity test for chlortetracycline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, depressor substances, loss on drying, pH, crystallinity, and 
identity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.01N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of chlortetracycline 
hydrochloride per milliliter (estimated). Further dilute an aliquot of 
the stock solution with

[[Page 764]]

sterile distilled water to the reference concentration of 0.06 microgram 
of chlortetracycline hydrochloride per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 5 milligrams of chlortetracycline 
hydrochloride per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (9) Identity. To 1.0 milligram of sample, add 2.0 milliliters of 
concentrated sulfuric acid. In the presence of chlortetracycline, a deep 
blue color is produced that becomes dark green.
[43 FR 11154, Mar. 17, 1978; 43 FR 34456, Aug. 4, 1978, as amended at 46 
FR 60568, Dec. 11, 1981; 50 FR 19920, May 13, 1985]



Sec. 446.15  Demeclocycline.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Demeclocycline is [4S - 
(4,4a,5a,6,12a)] - 7 - 
chloro - 4 - (dimethylamino) - 1,4,4a,5,5a,6,11, 12a - octahydro - 
3,6,10,12,12a - pentahydroxy - 1, 11 - dioxo - 2 - 
naphthacenecarboxamide. It is so purified and dried that:
    (i) Its potency is not less than 970 micrograms of demeclocycline 
hydrochloride equivalent per milligram on the anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 4.3 percent and not more 
than 6.7 percent.
    (iv) Its pH is an aqueous solution containing 10 milligrams per 
milliliter is not less than 4 and not more than 5.5.
    (v) When calculated on the anhydrous basis, its absorptivity at 380 
nanometers relative to that of the demeclocycline hydrochloride working 
standard is 107.4plus-minus3.88.
    (vi) It is crystalline.
    (vii) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, absorptivity, crystallinity, and identity.
    (ii) Samples required: 10 packages, each containing approximately 
250 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of demeclocycline 
hydrochloride per milliliter (estimated). Further dilute an aliquot of 
the stock solution with sterile distilled water to the reference 
concentration of 0.100 microgram of demeclocycline hydrochloride per 
milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Absorptivity. Determine the percent absorptivity of the sample 
relative to that of the standard in the following manner: Dissolve an 
accurately weighed portion of approximately 40 milligrams of the sample 
in 2 milliliters of 0.1N HCl, dilute to exactly 250 milliliters with 
distilled water, and mix thoroughly. Transfer a 10-milliliter aliquot of 
this solution to a 100-milliliter volumetric flask. Add about 75 
milliliters of distilled water and 5 milliliters of 5N NaOH, dilute to 
volume with distilled water, and mix thoroughly. Exactly 6 minutes after 
the addition of the NaOH, determine the absorbance of the solution at a 
wavelength of 380 nanometers, using a suitable spectrophotometer and 
distilled water as the blank. Treat a portion of the demeclocycline 
hydrochloride

[[Page 765]]

working standard in the same manner. Determine the percent relative 
absorptivity of the sample using the following calculation:
[GRAPHIC] [TIFF OMITTED] TR01JA93.231

    where: m=percent moisture in the sample.

    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (7) Identity. Proceed as directed in Sec. 446.16(b)(7). The value 
yielded by calculation ranges between 0.97 and 1.17.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11155, Mar. 17, 1978; 43 
FR 34456, Aug. 4, 1978; 46 FR 16683, Mar. 13, 1981; 50 FR 19920, May 13, 
1985]



Sec. 446.16  Demeclocycline hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Demeclocycline hydrochloride is [4S - 
(4 4a,5a,6,12a)] - 7 - 
chloro - 4 - (dimethylamino) - 1,4,4a,5,5a,6,11,12a - octahydro-
3,6,10,12,12a - pentahydroxy - 1,11 - dioxo - 2 - naphthacenecarboxamide 
monohydrochloride. It is so purified and dried that:
    (i) Its potency is not less than 900 micrograms per milligram on the 
anhydrous basis.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 2 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 2 and not more than 3.
    (v) When calculated on the anhydrous basis, its absorptivity at 380 
nanometers relative to that of the demeclocycline hydrochloride standard 
is 100plus-minus4.2 percent.
    (vi) It is crystalline.
    (vii) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, absorptivity, crystallinity, and identity.
    (ii) Samples required: 10 packages, each containing approximately 
250 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of demeclocycline 
hydrochloride per milliliter (estimated). Further dilute an aliquot of 
the stock solution with sterile distilled water to the reference 
concentration of 0.100 microgram of demeclocycline hydrochloride per 
milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Absorptivity. Determine the percent absorptivity of the sample 
relative to that of the standard in the following manner: Dissolve an 
accurately weighed portion of approximately 40 milligrams of the sample 
in 2 milliliters of 0.1N HCl, dilute to exactly 250 milliliters with 
distilled water, and mix thoroughly. Transfer a 10 milliliter aliquot of 
this solution to a 100-milliliter volumetric flask. Add about 75 
milliliters of distilled water and 5 milliliters of 5N NaOH, dilute to 
volume with distilled water, and mix thoroughly. Exactly 6 minutes after 
the addition of the NaOH, determine the absorbance of the solution at a 
wavelength of 380 nanometers, using a suitable spectrophotometer and 
distilled water as the blank. Treat a portion of

[[Page 766]]

the working standard in the same manner. Determine the percent relative 
absorptivity of the sample using the following calculation:
[GRAPHIC] [TIFF OMITTED] TR01JA93.232

where: m=percent moisture in the sample.

    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (7) Identity. Accurately weigh 40 milligrams of the sample and place 
into a 200-milliliter volumetric flask. Add 100 milliliters of 0.1N HCl 
and place on a shaker until the sample is dissolved. Dilute to volume 
with 0.1N HCl and mix well. Transfer a 5-milliliter aliquot of the 
solution to each of two 50-milliliter volumetric flasks. To one flask 
add 10 milliliters of 6N HCl and to the other add 10 milliliters of 3N 
HCl. Place the acid-treated flasks into a boiling water batch for 20 
minutes. Remove the flasks and place in a cold water bath. When cool, 
dilute to volume with water and mix well. Treat a portion of the 
standard in the same manner. Using a suitable spectrophotometer, place 
the 6N HCl-treated sample into the reference cell and read against the 
3N HCl-treated sample at a wavelength of 368 nanometers. Reverse the 
order of the cells in the cell holder and read at a wavelength of 430 
nanometers.
[GRAPHIC] [TIFF OMITTED] TR01JA93.233

where: m=percent moisture in the sample.

[39 FR 19076, May 30, 1974, as amended at 43 FR 11155, Mar. 17, 1978; 43 
FR 34456, Aug. 4, 1978; 50 FR 19920, May 13, 1985]



Sec. 446.20  Doxycycline hyclate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Doxycycline hyclate is [4S - 
4a,4a,5a,5a,6,12a]-
 4 - dimethylamino) -1,4,4a,5,5a,6,11,12a - octahydro- 3,5,10,12,12a-
pentahydroxy - 6 -methyl-1,11- dioxo -2 - naphthacenecarboxamide 
hydrochloridehemiethanolate hemihydrate. It is so purified and dried 
that:
    (i) Its potency is not less than 800 nor more than 920 micrograms of 
doxycycline per milligram on an ``as is'' basis.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 1.4 nor more than 2.75 
percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 2.0 nor more than 3.0.
    (v) It contains not less than 82 nor more than 90 percent 
doxycycline on an ``as is'' basis.
    (vi) It gives a positive identity test for doxycycline hyclate.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, doxycycline content, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.

[[Page 767]]

    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of doxycycline per 
milliliter (estimated). Further dilute with sterile distilled water to 
the reference concentration of 0.100 microgram of doxycycline per 
milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing the equivalent of 10 milligrams of 
doxycycline per milliliter.
    (5) Doxycycline content--(i) Equipment--(a) Sheet (chromatographic). 
Whatman No. 4 filter paper for chromatography, 15  x  57 centimeters.
    (b) Chamber (chromatographic). Square glass chromatography jar, 30 
x  30  x  60 centimeters, equipped with 25-centimeter troughs for 
descending chromatography.
    (ii) Preparation of solutions--(a) 0.05N Methanolic hydrochloric 
acid. Dilute 4.2 milliliters of concentrated hydrochloric acid to 1 
liter with methanol.
    (b) pH 4.2 buffer. Mix 5.86 volumes of 0.1M citric acid with 4.14 
volumes of 0.2M disodium phosphate.
    (c) Chromatographic system. Mix toluene, pyridine, and pH 4.2 buffer 
in volumetric proportions of 20:3:10, respectively. Allow the phases to 
separate. Place the upper phase in the troughs near the top of the 
chamber. Place the lower phase in the bottom of the chamber. Saturate 
the atmosphere of the tightly sealed chamber for 24 hours before use by 
placing white blotters on two opposite sides of the chamber so that 
their ends are immersed in the lower phase in the bottom of the chamber. 
Replace the solvent in troughs before the chromatograms are to be 
developed.
    (iii) Preparation of the doxycycline standard solution. Accurately 
weigh about 50 milligrams of the doxycycline working standard into a 5-
milliliter volumetric flask and bring to volume with 0.05N methanolic 
hydrochloric acid. Store in the refrigerator and use within 7 days.
    (iv) Preparation of sample. Accurately weight about 50 milligrams of 
the sample into a 5-milliliter volumetric flask and bring to volume with 
0.05N methanolic hydrochloric acid.
    (v) Preparation of the chromatogram. Dip the chromatographic sheets 
into pH 4.2 buffer and lightly blot each sheet between clean 
nonfluorescing, white blotters. Use separate sheets for the doxycycline 
standard solution, for each doxycycline sample solution, and for blanks 
without standard or sample application. Care must be taken so that the 
moist sheets do not become too dry; a period of 5 to 10 minutes between 
impregnating the paper and placing it in the chromatographic chamber is 
usually satisfactory. Evenly apply a 0.100-milliliter aliquot of a 
doxycycline solution to the origin line of a sheet as a 14-centimeter-
long streak. Place the sheets in the chamber and develop them in a 
descending manner for 2 hours. The doxycycline band should move 
approximately 12.5 centimeters from the origin line. Remove the sheets 
from the chamber and air-dry for about 10 minutes.
    (vi) Processing the chromatogram. Examine each sheet under 366-
nanometer ultraviolet light. Outline the fluorescent bands with a 
pencil. The main marked area should be approximately 10  x  15 
centimeters in size. Outline areas on the blank sheet approximately 
equal in size and in the same locations as those outlined on the 
standard sheet. Exposure of the sheets to ammonia or other alkaline 
vapors must be avoided. Cut the marked areas from the sheets and then 
cut them into approximately 2-centimeter squares. For each sheet, place 
the squares from each of the following areas into separate 125-
milliliter Erlenmeyer flasks: The main doxycycline band of the sample, 
the main doxycycline band of the standard, all the other bands of the 
standard, the area of the blank sheet corresponding to the main band of 
the standard, the other area of the blank sheet corresponding to the 
other bands of the standard. The time between removing the sheets from 
the chamber and placing the squares into the Erlenmeyer

[[Page 768]]

flasks should be minimal, since excessive drying of the paper can lead 
to erratic elutions.
    (vii) Elution. To each flask add 50 milliliters of 0.05N methanolic 
hydrochloric acid and agitate on a reciprocating shaker for 1 hour. 
Decant the contents of each flask into another flask by pouring through 
a small funnel fitted with a glass wool plug.
    (viii) Doxycycline standard solution for direct measurement of 
absorbance. Pipette a 0.100-milliliter aliquot of the doxycycline 
standard solution into each of three 125-milliliter Erlenmeyer flasks. 
Add 50 milliliters of 0.05N methanolic hydrochloric acid to each of 
these flasks.
    (ix) Absorbance measurement. Using a suitable spectrophotometer and 
0.05N methanolic hydrochloric acid as the reference solvent, determine 
the absorbance of each eluate and of each doxycycline standard solution 
at the absorption maximum at about 349 nanometers.
    (x) Calculation of percent doxycycline in samples. Calculate as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.234

where:
Au=Absorbance of the eluate from the main doxycycline band of 
the sample sheet.
As=Absorbance of the eluate from the main doxycycline band on 
the standard sheet.
Ab=Absorbance of the eluate from the area of the blank sheet 
corresponding to the area of the doxycycline band of the standard sheet.
Wu=Weight in milligrams of sample.
Ws=Weight in milligrams of doxycycline working standard.

    (xi) Recovery of the doxycycline standard from the chromatogram. As 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.235

where:
Ap=Absorbance of the doxycycline standard solution described 
in paragraph (b)(5)(viii) of this section.
F=The fractional purity of doxycycline standard solution described in 
paragraph (b)(5)(xii) of this section.


If the recovery of the doxycycline standard from the chromatogram is 
less than 95 percent, repeat the chromatogram.
    (xii) Determination of the fractional purity of the doxycycline 
working standard. Determine F by means of the following equation:
[GRAPHIC] [TIFF OMITTED] TR01JA93.236

where:
Ac=Absorbance of the eluate from sections of the standard 
chromatogram containing nondoxycycline 349 nanometers-absorbing 
contaminants.
Acb=Absorbance of the eluates from the sections of the blank 
sheets corresponding to those sections of the nondoxycycline-absorbing 
contaminants of the standard sheets.

    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 0.25 potassium bromide mixture described in paragraph (b)(1) 
of that section.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11155, Mar. 17, 1978; 50 
FR 19920, May 13, 1985]



Sec. 446.20a  Sterile doxycycline hyclate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, equality, and purity. Sterile doxycycline hyclate is [4S - 
(4,4a,5,5a,6 12a)] 
- 4 - (dimethylamino) - 1,4,4a,5,5a,6,11,12a - octahydro - 3,5,10,12,12a 
- pentahydroxy - 6 - methyl - 1,11 - dioxo - 2 - naphthacenecarboxamide 
hydrochloride hemiethanolate hemihydrate. It is so purified and dried 
that:
    (i) Its potency is not less than 800 nor more than 920 micrograms of 
doxycycline per milligram on an ``as is'' basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]

[[Page 769]]

    (v) It contains no depressor substances.
    (vi) Its moisture content is not less than 1.4 nor more than 2.75 
percent.
    (vii) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 2.0 nor more than 3.0.
    (viii) It contains not less than 82 nor more than 90 percent 
doxycycline on an ``as is'' basis.
    (ix) It gives a positive identity test for doxycycline hyclate.
    (x) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this subchapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this subchapter, each such 
request shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, depressor substances, moisture, pH, doxycycline content, 
identity, and crystallinity.
    (ii) Samples required:
    (a) For all tests except sterility: 12 packages, each containing 
approximately 300 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of doxycycline per 
milliliter (estimated). Further dilute with sterile distilled water to 
the reference concentration of 0.100 microgram of doxycycline per 
milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this 
subchapter, using the method described in paragraph (e)(1) of that 
section, except use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this 
subchapter, using a solution containing 7.5 milligrams of doxycycline 
per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
subchapter.
    (6) Moisture. Proceed as directed in Sec. 436.201 of this 
subchapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using an aqueous solution containing the equivalent of 10 milligrams of 
doxycycline per milliliter.
    (8) Doxycycline content. Proceed as directed in Sec. 446.20(b)(5).
    (9) Identity. Proceed as directed in Sec. 436.211 of this 
subchapter, using the 0.25 potassium bromide mixture described in 
paragraph (b)(1) of that section.
    (10) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
subchapter.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11155, Mar. 17, 1978; 46 
FR 60568, Dec. 11, 1981; 50 FR 19920, May 13, 1985]



Sec. 446.21  Doxycycline monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Doxycycline monohydrate is [4S - 
(4,4a,5,5a,6,12a)] 
- 4 - (dimethylamino) - 1,4,4a,5,5a,6,11, - 12a - octahydro - 
3,5,10,12,12a - pentahydroxy - 6 - methyl - 1,11 - dioxo - 2 - naphtha - 
cenecarboxamide monohydrate. It is so purified and dried that:
    (i) Its potency is not less than 880 micrograms nor more than 980 
micrograms of doxycycline per milligram on an ``as is'' basis.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 3.6 percent nor more 
than 4.6 percent.
    (iv) Its pH in an aqueous suspension containing the equivalent of 10 
milligrams of doxycycline per milliliter is not less than 5.0 nor more 
than 6.5.
    (v) It contains not less than 90 percent nor more than 98 percent 
doxycycline on an ``as is'' basis.
    (vi) It gives a positive identity test for doxycycline monohydrate.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, doxycycline content, identity, and crystallinity.

[[Page 770]]

    (ii) Samples of the batch: 10 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of doxycycline per 
milliliter (estimated). Further dilute with sterile distilled water to 
the reference concentration of 0.100 microgram of doxycycline per 
milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous suspension containing the equivalent of 10 milligrams of 
doxycycline per milliliter.
    (5) Doxycycline content. Proceed as directed in Sec. 446.20(b)(5).
    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 0.25 potassium bromide mixture described in paragraph (b)(1) 
of that section.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11155, Mar. 17, 1978; 45 
FR 16476, Mar. 14, 1980; 50 FR 19920, May 13, 1985]



Sec. 446.42  Meclocycline sulfosalicylate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Meclocycline sulfosalicylate is the 
sulfosalicylate salt of 7-chloro-4-(dimethylamino)-1,4,4a,5,5a,6,11,12a-
octahydro-3,5,10,12,12a-pentahydroxy-6-methylene-1,11-dioxo-2-
naphthacenecarboxamide. It is so purified and dried that:
    (i) Its potency is not less than 620 micrograms of meclocycline per 
milligram on an ``as is'' basis.
    (ii) Its moisture content is not more than 4.0 percent.
    (iii) Its pH is in an aqueous suspension containing 10 milligrams 
per milliliter is not less than 2.5 and not more than 3.5.
    (iv) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on potency, moisture, pH, and 
crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the high-pressure 
liquid chromatography method shall be conclusive.
    (i) High-pressure liquid chromatography. Proceed as directed in 
Sec. 436.329 of this chapter.
    (ii) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 0.01N 
methanolic hydrochloric acid (solution 13) to obtain a stock solution of 
convenient concentration. Further dilute an aliquot of the stock 
solution with distilled water to the reference concentration of 0.06 
microgram of meclocycline per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous suspension containing 10 milligrams of meclocycline per 
milliliter.
    (4) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[46 FR 3836, Jan. 16, 1981]



Sec. 446.50  Methacycline hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Methacycline hydrochloride is [4S - 
(4,4a,5,5a, - 12a)] - 4 - 
(dimethylamino) - 1,4,4a,5,5a,6,11,12a - octahydro - 3,5,10,12,12a - 
pentahydroxy - 6 - methylene - 1,11 - dioxo - 2 - naphthacenecar - 
boxamide monohydrochloride. It is so purified and dried that:
    (i) Its potency is not less than 832 micrograms of methacycline per 
milligram on an ``as is'' basis.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 2 percent.

[[Page 771]]

    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 2.0 nor more than 3.0.
    (v) Its absorptivity at the absorption maximum of 345 nanometers 
relative to that of the methacycline working standard similarly treated 
is 92.4plus-minus4 percent.
    (vi) It gives a positive result to the identity test for 
methacycline hydrochloride.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, absorptivity, identity, and crystallinity.
    (ii) Samples of the batch: 10 packages, each containing 300 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient sterile distilled 
water to obtain a stock solution of convenient concentration. Further 
dilute an aliquot of the stock solution with sterile distilled water to 
the reference concentration of 0.06 microgram of methacycline per 
milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams of methacycline per 
milliliter.
    (5) Absorptivity. Determine the absorbance of the sample and 
standard solutions in the following manner: Dissolve approximately 50 
milligrams each of the sample and standard in 100 milliliters of 0.01N 
methanolic hydrochloric acid. Transfer a 10-milliliter aliquot to a 250-
milliliter volumetric flask and dilute to volume with 0.01N methanolic 
hydrochloric acid. Using a suitable spectrphotometer and 0.01N 
methanolic hydrochloric acid as the blank, scan the absorption spectrum 
between the wavelengths of 250 and 400 nanometers. Determine the 
absorbance of each solution at the maxima, ca. 345 nanometers. Determine 
the percent absorptivity of the sample relative to the absorptivity of 
the standard using the following calculations:

Percent relative absorptivity=(Absorbance of sample x weight in 
          milligrams of standards x potency of standard in micrograms 
          per milligram)/(Absorbance of standard x weight in milligrams 
          of sample x 10)

    (6) Identity. The absorption spectrum between the wavelength of 250 
and 400 nanometers, determined as directed in paragraph (b)(5) of this 
section, compares qualitatively with that of the methacycline standard.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11155, Mar. 17, 1978; 50 
FR 19920, May 13, 1985]



Sec. 446.60  Minocycline hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Minocycline hydrochloride is [4S-
(4,4a,5a,12a)]-4,7-bis 
(dimethylamino)-1,4,4a,5,5a,6,11, - 12a-octahydro-3,10,12, - 12a-
tetrahydroxy-1,11-dioxo-2-naphthacenecarboxamide monohydrochloride. It 
is so purified and dried that:
    (i) Its potency is not less than 890 micrograms per milligram and 
not more than 950 micrograms per milligram on the anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 4.3 percent and not more 
than 8.0 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams of 
minocycline per milliliter is not less than 3.5 and not more than 4.5.
    (v) Its epi-minocycline content is not more than 1.2 percent.
    (vi) It gives a positive identity test for minocycline 
hydrochloride.
    (vii) It is crystalline.
    (viii) Its residue on ignition is not more than 0.15 percent.
    (ix) The absorptivity at 560 nanometers of an aqueous solution 
containing 10 milligrams of minocycline hydrochloride per milliliter is 
not more than 0.006.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 772]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, epi-minocycline content, identity, crystallinity, residue on 
ignition, and absorptivity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Minocycline potency. Proceed as 
directed in Sec. 436.216 of this chapter, using ambient temperature, an 
ultraviolet detection system operating at a wavelength of 280 
nanometers, a 4.6-millimeter  x  3-centimeter guard column containing 
10-micrometer diameter RP-8 Lichrosorb, a 4.6-millimeter  x  15-
centimeter analytical column packed with octyl silane chemically bonded 
to porous microsilica particles, 5 micrometers in diameter, a flow rate 
of 2.9 milliliters per minute, and a known injection volume of 10 
microliters. Reagents, working standard and sample solutions, system 
suitability requirements, and calculations are as follows:
    (i) Reagents--(a) 0.1 M Disodium ethylenediamine-tetraacetate 
(EDTA). Accurately weigh 37.22 grams of disodium 
ethylenediaminetetraacetate into a 1,000-milliliter volumetric flask. 
Dissolve in and dilute to mark with deionized water.
    (b) 0.2 M Ammonium oxalate. Accurately weigh 28.42 grams of ammonium 
oxalate into a 1,000-milliliter volumetric flask. Dissolve in and dilute 
to mark with deionized water.
    (c) Mobile phase. Mix 250 milliliters of dimethylformamide, 200 
milliliters of 0.1M disodium ethylenediaminetetraacetate and 550 
milliliters of 0.2M ammonium oxalate. (5:4:11). Allow the solution to 
cool to room temperature and then adjust the pH to 6.2 to 6.3 with 0.4M 
tetrabutylammonium hydroxide. Filter and degas the mobile phase just 
prior to its introduction into the chomatographic pumping system.
    (ii) Preparations of working standard, sample and resolution testing 
solutions--(a) Working standard solution. Dissolve an accurately weighed 
portion of the minocycline hydrocholoride working standard with 
sufficient mobile phase (prepared as described in paragraph (b)(1)(i)(c) 
of this section) to obtain a solution containing 500 micrograms of 
minocycline activity per milliliter. Use this standard solution within 3 
hours of preparation.
    (b) Sample solution. Dissolve an accurately weighed sample in 
sufficient mobile phase to obtain a solution containing 500 micrograms 
of minocycline activity per milliliter (estimated). Use this solution 
within 3 hours of preparations.
    (iii) System suitability requirements--(a) Asymmetry factor. 
Calculate the asymmetry factor (As), measured at a point 5 
percent of the peak height from the baseline, as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.237

where:

a=Horizontal distance from point of ascent to point of maximum peak 
          height; and
b=Horizontal distance from the point of maximum peak height to point of 
          descent.

The asymmetry factor (As)is satisfactory if it is not less 
than 0.9 and not more than 1.35.

    (b) Efficiency of the column. From the number of theoretical plates 
(n) calculated as described in Sec. 436.216(c)(2) calculate the reduced 
plate height (hr) as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.238

where:

L=Length of the column in centimeters;
n=number of theoretical plates; and
dp=Average diameter of the particles in analytical column 
          packing in micrometers.

The absolute efficiency (hr) is satisfactory if it is not 
more than 50 for the minocycline peak.
    (c) Resolution. Dissolve 50 milligrams of minocycline hydrochloride 
in 25 milliliters of deionized water. Pipet 5 milliliters of this 
solution into a 25-milliliter volumetric flask and heat on a steam bath 
for 60 minutes. Transfer the contents of the flask to a small beaker and 
evaporate to dryness. Dissolve the residue in mobile phase, transfer to 
a 25-milliliter volumetric flask, dilute to mark with mobile phase, mix, 
and filter through Whatman No. 1 filter

[[Page 773]]

paper. Use this solution to determine the resolution factor. The 
resolution (R) between the peaks for minocycline and epi-minocycline is 
satisfactory if it is not less than 2.0.
    (d) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent) of 5 replicate 
injections is satisfactory if it is not more than 2.0 percent.
    (e) Capacity factor (k'). Calculate the capacity factor (k') for 
minocycline as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.239

where:

tr=Retention time of minocycline in minutes; and
to=Column dead time in minutes, which is estimated from the 
          following equation:
          [GRAPHIC] [TIFF OMITTED] TR01JA93.240
          
where:

D=Column diameter in centimeters;
L=Column length in centimeters;
0.75=Average total column porosity; and
F=Flow rate in milliliters per minute.


The capacity factor (k') for minocycline is satisfactory if it is not 
less than 6.2 and not more than 11.5.
    If the system suitability requirements have been met, then proceed 
as described in Sec. 436.216(b) of this chapter. Alternate 
chromatographic conditions are acceptable provided reproducibility and 
resolution are comparable to the system. However, the sample preparation 
described in paragraph (b)(1)(ii)(b) of this section should not be 
changed.
    (iv) Calculations--Calculate the micrograms of minocycline per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.241

where:
Au=Area of the minocycline peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the minocycline peak in the chromatogram of the 
          minocycline working standard;
Ps=Minocycline activity in the minocycline working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of minocycline sample per milliliter of sample 
          solution; and
m=Percent moisture content of the sample.

    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams of minocycline per 
milliliter.
    (5) Epi-minocycline content. Proceed as directed in paragraph (b)(1) 
of this section. Calculate the epi-minocycline content as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.242

where:
Aepi=Area of the epi-minocycline peak in the chromatogram of 
          the sample; and
Atotal=The sum of the areas of all the peaks eluting after 
          the solvent front.

    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 0.5 percent potassium bromide disc prepared as described in 
paragraph (b)(1) of that section.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (8) Residue on ignition. Proceed as directed in Sec. 436.207(b) of 
this chapter.
    (9) Absorptivity. Accurately weigh about 1 gram of sample into a 
100-milliliter volumetric flask, dissolve, and dilute to mark with 
deionized water. Determine the absorbance of this solution on a suitable 
spectrophotometer at 560 nanometers (nm) using 5-centimeter cells with 
water in the reference cell. Calculate the absorptivity as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.243

[39 FR 19076, May 30, 1974, as amended at 43 FR 11156, Mar. 17, 1978; 43 
FR 34456, Aug. 4, 1978; 44 FR 22058, Apr. 13, 1979; 50 FR 19920, May 13, 
1985; 53 FR 32607, Aug. 26, 1988; 53 FR 39839, Oct. 12, 1988; 54 FR 
47205, Nov. 13, 1989]



Sec. 446.65  Oxytetracycline.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline is [4S-
(4,4a,5,5a,6,12a)]-
4-(dimeth-

[[Page 774]]

ylamino)-1,4,4a,5,5a,6,11,12a-octa - hydro- 3,5,6,10,12,12a-hexahydroxy-
6-methyl- 1,11-dioxo-2-naphthacenecar - boxamide dihydrate. 
Oxytetracycline is produced by the growth of Streptomyces rimosus. It is 
so purified and dried that:
    (i) Its potency is not less than 832 micrograms of oxytetracycline 
per milligram on an ``as is'' basis.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 6 percent and not more 
than 9 percent.
    (iv) Its pH in an aqueous suspension containing 10 milligrams per 
milliliter is not less than 4.5 and not more than 7.0.
    (v) When calculated on an anhydrous basis its absorptivity at 353 
nanometers relative to that of the oxytetracycline working standard 
similarly treated is 100plus-minus4 percent.
    (vi) It gives a positive result to an identity test for 
oxytetracycline.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, absorptivity, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
either of the following methods; however, the results obtained from the 
microbiological turbidimetric assay shall be conclusive.
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of oxytetracycline 
per milliliter (estimated). Further dilute an aliquot of the stock 
solution with sterile distilled water to the reference concentration of 
0.24 microgram of oxytetracycline per milliliter (estimated).
    (ii) Chemical assay. Proceed as directed in Sec. 436.320 of this 
chapter.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous suspension containing 10 milligrams per milliliter.
    (5) Absorptivity. Determine the absorbance of the sample and 
standard solutions in the following manner: Dissolve approximately 50 
milligrams each of the sample and standard in 250 milliliters of 0.1N 
hydrochloric acid. Transfer a 10-milliliter aliquot to a 100-milliliter 
volumetric flask and dilute to volume with 0.1N hydrochloric acid. Using 
a suitable spectrophotometer and 0.1N hydrochloric acid as the blank, 
determine the absorbance of each solution at 353 nanometers. Determine 
the percent absorptivity of the sample relative to the absorptivity of 
the standard using the following calculations:
[GRAPHIC] [TIFF OMITTED] TR01JA93.244

where: m = Percent moisture in the sample.

    (6) Identity. To about 1 milligram of sample, add 2 milliliters of 
sulfuric acid; a light-red color is produced when oxytetracycline is 
present.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[43 FR 11156, Mar. 17, 1978, as amended at 50 FR 19920, May 13, 1985]



Sec. 446.65a  Sterile oxytetracycline.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality,

[[Page 775]]

and purity. Sterile oxytetracycline is [4S - 
(4,4a,5,5a,6,12a)] 
- 4 - (dimethylamino) - 1,4,4a,5,5a,6,11, 12a - octahydro - 
3,5,6,10,12,12a - hexahydroxy - 6 - methyl - 1,11 - dioxo - 2 - 
naphthacenecarboxamide dihydrate. Oxytetracycline is produced by the 
growth of Streptomyces rimosus. It is so purified and dried that:
    (i) Its potency is not less than 832 micrograms of oxytetracycline 
per milligram on an ``as is'' basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) It contains no depressor substances.
    (vi) Its moisture content is not less than 6 percent and not more 
than 9 percent.
    (vii) Its pH in an aqueous suspension containing 10 milligrams per 
milliliter is not less than 4.5 and not more than 7.0.
    (viii) When calculated on an anhydrous basis, its absorptivity at 
353 nanometers relative to that of the oxytet- racycline working 
standard similarly treated, is 100plus-minus4 percent.
    (ix) It gives a positive result to an identity test for 
oxytetracycline.
    (x) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, depressor substances, moisture, pH, absorptivity, identity, 
and crystallinity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
either of the following methods; however, the results obtained from the 
microbiological turbidimetric assay shall be conclusive.
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of oxytetracycline 
per milliliter (estimated). Further dilute an aliquot of the stock 
solution with sterile distilled water to the reference concentration of 
0.24 microgram of oxytetracycline per milli- liter (estimated).
    (ii) Chemical assay. Proceed as directed in Sec. 436.320 of this 
chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 5.0 milligrams of oxytetracycline per 
milliliter prepared by dissolving 40 milligrams in 2.0 milliliters of 
0.1N hydrochloric acid and diluting with the required amount of sterile, 
pyrogen-free distilled water.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter, preparing the sample by dissolving 40 milligrams in 2.0 
milliliters of 0.1N hydrochloric acid and diluting with the required 
amount of sterile distilled water.
    (6) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous suspension containing 10 milligrams per milliliter.
    (8) Absorptivity. Determine the absorbance of the sample and 
standard solutions in the following manner: Dissolve approximately 50 
milligrams each of the sample and standard in 250 milliliters of 0.1N 
hydrochloric acid. Transfer a 10-milliliter aliquot to a 100-milliliter 
volumetric flask, and dilute to volume with 0.1N hydrochloric acid. 
Using a suitable spectrophotometer and 0.1N hydrochloric acid as the 
blank, determine the absorbance of each solution at 353 nanometers. 
Determine the percent absorptivity of the sample relative to the 
absorptivity of the standard using the following calculations:

[[Page 776]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.245


where: m =Percent moisture in the sample.

    (9) Identity. To about 1 milligram of sample, add 2 milliliters of 
sulfuric acid; a light-red color is produced when oxytetracycline is 
present.
    (10) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[43 FR 11156, Mar. 17, 1978; 43 FR 34456, Aug. 4, 1978, as amended at 46 
FR 60568, Dec. 11, 1981; 50 FR 19920, May 13, 1985]



Sec. 446.66  Oxytetracycline calcium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline calcium is [4S-
(4,4a,5,5a,6,12
)]-4-(dimethylamino)-1,4,4a,5,5a,6,11, 12a- octahydro-3,5,6,10,12,12a-
hexa hydroxy-6-methyl-1,11-dioxo- 2-naphthacenecarboxamide calcium salt. 
Oxytetracycline is produced by the growth of Streptomyces rimosus. It is 
so purified and dried that:
    (i) Its potency is equivalent to not less than 865 micrograms of 
oxytetracycline per milligram on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 8 percent and not more 
than 14 percent.
    (iv) Its pH in an aqueous suspension containing 25 milligrams per 
milliliter is not less than 6.0 and not more than 8.0
    (v) Its calcium content as the sulfated ash is not less than 3.85 
percent and not more than 4.35 percent on an anhydrous basis.
    (vi) It gives a positive identity test.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, calcium content, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
either of the following methods; however, the results obtained from the 
microbiological turbidimetric assay shall be conclusive.
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of oxytetracycline 
per milliliter (estimated). Further dilute an aliquot of the stock 
solution with sterile distilled water to the  reference  concentration  
of  0.24 microgram of oxytetracycline per milliliter (estimated).
    (ii) Chemical assay. Proceed as directed in Sec. 436.320 of this 
chapter.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
saturated aqueous suspension containing 25 milligrams per milliliter.
    (5) Calcium content. Proceed as directed in Sec. 436.207(b) of this 
chapter, except from the weight of residue obtained calculate the 
calcium content as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.246


[[Page 777]]


where: m = Percent moisture in the sample.

    (6) Identity. To about 1 milligram of sample, add 2 milliliters of 
sulfuric acid; a light-red color is produced when oxytetracycline is 
present.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[43 FR 11157, Mar. 17, 1978; 43 FR 34456, Aug. 4, 1978, as amended at 50 
FR 19920, May 13, 1985]



Sec. 446.67  Oxytetracycline hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline hydrochloride is [4S-
(4,4a,5,5a, 
6,12a,)] - 4 - (dimethylamino) - 1,4,4a,5, 5a,6, 11, 
12a - octahydro - 3,5,6,10,12,12a - hexahydroxy - 6 - methyl - 1,11 - 
dioxo - 2 - naphthacenecarboxamide monohydrochloride. Oxytetracycline is 
produced by the growth of Streptomyces rimosus. It is so purified and 
dried that:
    (i) Its potency is not less than 835 micrograms of oxytetracycline 
per milligram on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 2 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 2.0 and not more than 3.0.
    (v) When calculated on an anhydrous basis, its absorptivity at 353 
nanometers relative to that of the oxytetracycline standard similarly 
treated is 92.5plus-minus4.3 percent.
    (vi) It gives a positive result to an identity test for 
oxytetracycline.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, absorptivity, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
either of the following methods; however, the results obtained from the 
microbiological turbidimetric assay shall be conclusive.
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of oxytetracycline 
per milliliter (estimated). Further dilute an aliquot of the stock 
solution with sterile distilled water to the reference concentration of 
0.24 microgram of oxytetracycline per milli- liter (estimated).
    (ii) Chemical assay. Proceed as directed in Sec. 436.320 of this 
chapter.
    (2) [Reserved]
    (3) Loss on drying. Proceed as di- rected in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Absorptivity. Determine the absorbance of the sample and 
standard solutions in the following manner: Dissolve approximately 50 
milligrams each of the sample and standard in 250 milliliters of 0.1N 
hydrochloric acid. Transfer a 10-milliliter aliquot to a 100-milliliter 
volumetric flask and dilute to volume with 0.1N hydrochloric acid. Using 
a suitable spectrophotometer and 0.1N hydrochloric acid as the blank, 
determine the absorbance of each solution at 353 nanometers. Determine 
the percent absorptivity of the sample relative to the absorptivity of 
the standard, using the following calculations:
[GRAPHIC] [TIFF OMITTED] TR01JA93.247


[[Page 778]]


where: m = Percent moisture in the sample.

    (6) Identity. To about 1 milligram of sample, add 2 milliliters of 
sulfuric acid; a light-red color is produced when oxytetracycline is 
present.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[43 FR 11157, Mar. 17, 1978; 43 FR 34456, Aug. 4, 1978, as amended at 50 
FR 19920, May 13, 1985]



Sec. 446.67a  Sterile oxytetracycline hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile oxytetracycline hydrochloride is 
[4S - 
(4,4a,5,5a,6,12)] 
- 4 - (dimethylamino) - 1,4,4a,5,5a,6,11,12a - octahydro - 
3,5,6,10,12,12a - hexahydroxy - 6 - methyl - 1,11 - dioxo - 2 - 
naphthacenecarboxamide monohydrochloride. It is produced by the growth 
of Streptomyces rimosus. It is so purified and dried that:
    (i) Its potency is not less than 835 micrograms of oxytetracycline 
per milligram on an anhydrous basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) It contains no depressor substances.
    (vi) Its loss on drying is not more than 2.0 percent.
    (vii) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 2.0 and not more than 3.0.
    (viii) When calculated on an anhydrous basis, its absorptivity at 
353 nanometers relative to that of the oxytetracycline working standard 
similarly treated is 92.5plus-minus4.3 percent.
    (ix) It gives a positive result to an identity test for 
oxytetracycline.
    (x) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, depressor substances, loss on drying, pH, absorptivity, 
identity, and crystallinity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Assay for potency by 
either of the following methods; however, the results obtained from the 
microbiological turbidimetric assay shall be conclusive.
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of oxytetracycline 
per milliliter (estimated). Further dilute an aliquot of the stock 
solution with sterile distilled water to the reference concentration of 
0.24 microgram of oxytetracycline per milliliter (estimated).
    (ii) Chemical assay. Proceed as directed in Sec. 436.320 of this 
chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 5 milligrams of oxytetracycline per 
milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) Loss on drying. Proceed as di- rected in Sec. 436.200(b) of this 
chapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (8) Absorptivity. Determine the absorbance of the sample and 
standard solutions in the following manner: Dissolve approximately 50 
milligrams each of the sample and standard in 250 milliliters of 0.1N 
hydrochloric acid. Transfer a 10-milliliter aliquot to a 100-milliliter 
volumetric flask and dilute to volume with 0.1N hydrochloric acid. Using 
a suitable spectrophotometer and 0.1N hydrochloric acid as the blank, 
determine the absorbance of each solution

[[Page 779]]

at 353 nanometers. Determine the percent absorptivity of the sample 
relative to the absorptivity of the standard using the following 
calculations:
[GRAPHIC] [TIFF OMITTED] TR01JA93.248

where: m = Percent moisture in the sample.

    (9) Identity. To about 1 milligram of sample, add 2 milliliters of 
sulfuric acid; a light-red color is produced when oxytetracycline is 
present.
    (10) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[43 FR 11158, Mar. 17, 1978; 43 FR 34456, Aug. 4, 1978, as amended at 46 
FR 60568, Dec. 11, 1981; 50 FR 19920, May 13, 1985]



Sec. 446.75a  Sterile rolitetracycline.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile rolitetracycline is [4S-
(4,4a,5a,6,12a)] - 4 - 
(dimethylamino) - 1,4,4a,5,5a,6,11,12a - octahydro - 3,6,10,12,12a - 
pentahydroxy - 6 - methyl - 1,11 - dioxo - N - (1 - pyrrolidinylmethyl) 
- 2 - naphthacenecarboxamide. It is so purified and dried that:
    (i) Its potency is not less than 900 micrograms per milligram on the 
anhydrous basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) It contains no depressor substances.
    (vi) Its moisture content is not more than 3.0 percent.
    (vii) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 7 and not more than 9, and such solution is 
substantially clear.
    (viii) It is crystalline.
    (ix) When calculated on an anhydrous basis, its absorptivity at 380 
nanometers relative to that of the rolitetracycline standard similarly 
treated is 100plus-minus4.4 percent.
    (x) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this subchapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this subchapter, each such 
request shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, depressor substances, moisture, pH, crystallinity, 
absorptivity, and identity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 
methyl alcohol to give a solution containing 1 milligram of 
rolitetracycline per milliliter (estimated). Further dilute an aliquot 
of the stock solution with sterile distilled water to the reference 
concentration of 0.24 microgram of rolitetracycline per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this 
subchapter, using the method described in paragraph (e)(1) of that 
section, except use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this 
subchapter, using a solution containing 5.0 milligrams of 
rolitetracycline per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
subchapter.
    (6) Moisture. Proceed as directed in Sec. 436.201 of this 
subchapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using an aqueous solution containing 10 milligrams per milliliter.

[[Page 780]]

    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
subchapter.
    (9) Absorptivity. Determine the absorbance of the sample and 
standard solutions in the following manner: Dissolve an accurately 
weighed portion of approximately 40 milligrams each of the sample and 
standard in approximately 150 milliliters of distilled water and mix 
thoroughly. Dilute each to exactly 250 milliliters with distilled water 
and mix thoroughly. Transfer a 10.0-milliliter aliquot of each of these 
solutions to separate 100-milliliter volumetric flasks. Add 
approximately 75 milliliters of distilled water and 5.0 milliliters of 
5N NaOH to each flask, and then dilute to volume with water and mix 
thoroughly. Exactly 6 minutes after the addition of the NaOH, determine 
the absorbance of each solution at 380 nanometers, using a suitable 
spectrophotometer and distilled water as the blank. Determine the 
percent absorptivity of the sample relative to the absorptivity of the 
standard using the following calculations:
[GRAPHIC] [TIFF OMITTED] TR01JA93.249

where m=percent moisture in the sample.

    (10) Identity. Place approximately 100 milligrams of the sample to 
be tested in a test tube, and 5 milliliters of 1N NaOH, and heat gently 
to boiling for about 15 seconds. (The musty, aminelike odor of 
pyrrolidine is detectable.) Allow to cool to room temperature. A deep 
burgundy-red color of the clear solution indicates the presence of 
rolitetracycline.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11158, Mar. 17, 1978; 43 
FR 34456, Aug. 4, 1978; 46 FR 60568, Dec. 11, 1981; 50 FR 19920, May 13, 
1985]



Sec. 446.76a  Sterile rolitetracycline nitrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile rolitetracycline nitrate is [4S-
(4,4a,5a,6, 12a)] - 4 - 
(dimethylamino) - 1,4,4a,5,5a,6,11,12a - octahydro - 3,6,10,12,12a - 
pentahydroxy - 6 - methyl - 1,11 - dioxo - N - (1 - pyrrolidinylmethyl) 
- 2 - naphthacenecarboxamide mononitrate sesquihydrate. It is so 
purified and dried that:
    (i) It contains not less than 765 micrograms of rolitetracycline per 
milligram on an ``as is'' basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) It contains no depressor substances.
    (vi) Its moisture content is not more than 5.0 percent.
    (vii) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 3.5 and not more than 5.5.
    (viii) It is crystalline.
    (ix) When calculated on an anhydrous basis, its absorptivity at 380 
nanometers relative to that of the rolitetracycline standard treated is 
89.2plus-minus4.0 percent.
    (x) It gives a positive result to the identity tests for 
rolitetracycline nitrate.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this subchapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, depressor substances, moisture, pH, crystallinity, 
absorptivity, and identity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.

[[Page 781]]

    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient sterile distilled 
water to obtain a stock solution of convenient concentration. Further 
dilute an aliquot of the stock solution with sterile distilled water to 
the reference concentration of 0.24 microgram of rolitetracycline per 
milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this 
subchapter, using the method described in paragraph (e)(1) of that 
section, except use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this 
subchapter, using a solution containing 5.0 milligrams of 
rolitetracycline per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
subchapter.
    (6) Moisture. Proceed as directed in Sec. 436.201 of this 
subchapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using an aqueous solution containing 10 milligrams per milliliter.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
subchapter.
    (9) Absorptivity. Determine the absorbance of the sample and 
standard solutions in the following manner: Dissolve an accurately 
weighed portion of approximately 40 milligrams each of the sample and 
standard in approximately 150 milliliters of distilled water and mix 
thoroughly. Dilute each to exactly 250 milliliters with distilled water 
and mix thoroughly. Transfer a 10.0-milliliter aliquot of each of these 
solutions to representative 100-milliliter volumetric flasks. Add about 
75 milliliters of distilled water and 5.0 milliliters of 5N NaOH to each 
and then dilute to volume with water and mix thoroughly. Exactly 6 
minutes after the addition of the NaOH, determine the absorbance of each 
solution at 380 nanometers, using a suitable spectrophotometer and 
distilled water as the blank. Determine the percent absorptivity of the 
sample relative to the absorptivity of the standard using the following 
calculations:
[GRAPHIC] [TIFF OMITTED] TR01JA93.250

where: m=percent moisture in the sample.

    (10) Identity--(i) Rolitetracycline. Place approximately 100 
milligrams of the sample to be used in a test tube, add 5 milliliters of 
1N NaOH, and heat gently to boiling for about 15 seconds. (The musty, 
amine-like odor of pyrrolidine is detectable.) Allow to cool to room 
temperature. A deep burgundy-red color of the clear solution indicates 
the presence of rolitetracycline.
    (ii) Nitrate identity. Transfer approximately 1 gram of sample to a 
250-milliliter beaker, add 100 milliliters of water, and acidify with 1 
milliliter of acetic acid. Heat to boiling and, with constant stirring, 
add 10 milliliters of a 10-percent solution of nitron (1,4-diphenyl-3,5-
endo-anilino-4,5-dihydro-1,2,4-triazole) 
C20H16N4 7 in 1N acetic 
acid. Allow to cool. A heavy precipitate indicates the presence of 
nitrate.
---------------------------------------------------------------------------

    7 Nitron is available from J. T. Baker Laboratory 
Chemicals, North Phillipsburg, N.J. 08865.
---------------------------------------------------------------------------

[39 FR 19076, May 30, 1974, as amended at 43 FR 11159, Mar. 17, 1978; 46 
FR 60568, Dec. 11, 1981; 50 FR 19920, May 13, 1985]



Sec. 446.80  Tetracycline.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline is [4S -

[[Page 782]]

(4,4a,5a,6,12a)] - 4 - 
(dimethylamino) - 1,4,4a,5,5a,6,11,12a - octahydro - 3,6,10,12,12a - 
pentahydroxy - 6 - methyl - 1,11 - dioxo - 2 - naphthacenecarboxamide. 
It is so purified and dried that:
    (i) Its potency is not less than 975 micrograms per milligram on the 
anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 13 percent.
    (iv) Its pH in an aqueous suspension containing 10 milligrams per 
milliliter is not less than 3.0 and not more than 7.0.
    (v) When calculated on the anhydrous basis, its absorptivity at 380 
nanometers relative to that of the tetracycline hydrochloride working 
standard similarly treated is 108.2plus-minus3.75 percent.
    (vi) Its 4-epianhydrotetracycline content is not more than 2.0 
percent.
    (vii) It is crystalline.
    (viii) It passes the identity test for tetracycline.
    (2) Labeling. In addition to the requirements of Sec. 432.5 of this 
chapter, each package shall bear on its label or labeling the statement 
``For use only in the manufacture of nonparenteral drugs.''
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, absorptivity, 4-epianhydrotetra- cycline content, crystallinity, and 
identity.
    (ii) Samples required: 10 packages, each containing approximately 60 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of tetracycline 
hydrochloride per milliliter (estimated). Further dilute an aliquot of 
the stock solution with sterile distilled water to the reference 
concentration of 0.24 microgram of tetracycline hydrochloride per 
milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous suspension containing 10 milligrams per milliliter.
    (5) Absorptivity. Dissolve approximately 40 milligrams of the sample 
(as the anhydrous compound), accurately weighed, in 2.0 milliliters of 
0.1N hydrochloric acid and dilute with distilled water to 250 
milliliters. Transfer a 10.0-milliliter aliquot of this solution to a 
100-milliliter volumetric flask, add approximately 75 milliliters of 
distilled water and 5.0 milliliters of 5N NaOH, dilute to volume with 
water and mix thoroughly. Treat a sample of the tetracycline 
hydrochloride working standard in the same manner. Exactly 6 minutes 
after the addition of the NaOH, determine the absorbance of each 
solution at 380 nanometers, using a suitable spectrophotometer and 
distilled water as the blank. Determine the percent absorptivity of the 
sample relative to the absorptivity of the standard using the following 
calculations:
[GRAPHIC] [TIFF OMITTED] TC01AP94.061

where: m = Percent moisture in the sample.

    (6) 4-Epianhydrotetracycline. Proceed as directed in Sec. 436.309 of 
this chapter.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (8) Identity. Proceed as directed in Sec. 436.308 of this chapter.
[43 FR 11159, Mar. 17, 1978; 43 FR 34456, Aug. 4, 1978, as amended at 50 
FR 19920, May 13, 1985]

[[Page 783]]



Sec. 446.81  Tetracycline hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline hydrochloride is [4S-
(4,4a,5a,6, 12a)] - 4 - 
(dimethylamino) - 1,4,4a,5,5a,6,11,12a - octahydro - 3,6,10,12,12a - 
pentahydroxy - 6 - methyl - 1,11 - dioxo - 2 - naphthacenecarboxamide 
monohydrochloride. It is so purified and dried that:
    (i) Its potency is not less than 900 micrograms per milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 2 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 1.8 and not more than 2.8.
    (v) When calculated on the anhydrous basis, its absorptivity at 380 
nanometers relative to that of the tetracycline hydrochloride working 
standard similarly treated is 100plus-minus4 percent.
    (vi) Its 4-epianhydrotetracycline content is not more than 2.0 
percent.
    (vii) It is crystalline.
    (viii) It passes the identity test for tetracycline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, absorptivity, 4-epianhydrotetracycline content, 
crystallinity, and identity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of tetracycline 
hydrochloride per milliliter (estimated). Further dilute an aliquot of 
the stock solution with sterile distilled water to the reference 
concentration of 0.24 microgram of tetracycline hydrochloride per 
milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Absorptivity. Dissolve approximately 40 milligrams of the 
sample, accurately weighed, in approximately 150 milliliters of 
distilled water by mixing thoroughly. Dilute to 250 milliliters with 
distilled water and mix thoroughly. Transfer a 10.0 milliliter aliquot 
of this solution to a 100-milliliter volumetric flask, add about 75 
milliliters of distilled water and 5.0 milliliters of 5N NaOH, dilute to 
volume with water, and mix thoroughly. Treat a sample of the 
tetracycline hydrochloride working standard in the same manner. Exactly 
6 minutes after the addition of the NaOH, determine the absorbance of 
each solution at 380 nanometers, using a suitable spectrophotometer and 
distilled water as the blank. Determine the percent absorptivity of the 
sample relative to the absorptivity of the standard using the following 
calculations:
[GRAPHIC] [TIFF OMITTED] TC01AP94.062

where: m = Percent moisture in the sample.

    (6) 4-Epianhydrotetracycline. Proceed as directed in Sec. 436.309 of 
this chapter.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (8) Identity. Proceed as directed in Sec. 436.308 of this chapter.
[43 FR 11159, Mar. 17, 1978, as amended at 50 FR 19920, May 13, 1985]

[[Page 784]]



Sec. 446.81a  Sterile tetracycline hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline hydrochloride is [4S - 
(4,4a,5a,6, 12a)] - 4 - 
dimethylamino) - 1,4,4a,5,5a,6,11,12a - octahydro - 3,6,10,12,12a - 
pentahydroxy - 6 - methyl - 1,11 - dioxo - 2 - naphthacene - carboxamide 
monohydrochloride. It is so purified and dried that:
    (i) Its potency is not less than 900 micrograms of tetracycline 
hydrochloride per milligram. If it is packaged for dispensing, its 
content is satisfactory if it is not less than 90 percent and not more 
than 115 percent of the number of milligrams of tetracycline 
hydrochloride that it is represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) It contains no depressor substances.
    (vi) Its loss on drying is not more than 2 percent.
    (vii) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 1.8 and not more than 2.8.
    (viii) When calculated on the anhydrous basis, its absorptivity at 
380 nanometers relative to that of the tetracycline hydrochloride 
working standard similarly treated is 100plus-minus4 percent.
    (ix) Its 4-epianhydrotetracycline content is not more than 2.0 
percent.
    (x) It is crystalline.
    (xi) It passes the identity test for tetracycline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, depressor substances, loss on drying, pH, absorptivity, 4-
epianhydrotetracycline content, crystallinity, and identity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If the batch is packaged for dispensing.
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a stock solution containing 1,000 micrograms of 
tetracycline hydrochloride per milliliter (estimated); also, if it is 
packaged for dispensing, reconstitute as directed in the labeling. Then 
using a suitable hypodermic needle and syringe, remove all of the 
withdrawable contents if it is represented as a single dose container; 
or, if the labeling specifies the amount of potency in a given volume of 
the resultant preparation, remove an accurately measured representative 
portion from each container. Dilute the sample thus obtained with 
sufficient 0.1N hydrochloric acid to obtain a stock solution of 
convenient concentration containing not less than 150 micrograms of 
tetracycline hydrochloride per milliliter (estimated). Further dilute an 
aliquot of the stock solution with sterile distilled water to the 
reference concentration of 0.24 microgram of tetracycline hydrochloride 
per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a [so-]lution containing 5.0 milligrams of te-[tracycline] 
hydrochloride per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) Loss on drying. Proceed as di- rected in Sec. 436.200(b) of this 
chapter.

[[Page 785]]

    (7) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (8) Absorptivity. Dissolve approximately 40 milligrams of the 
sample, accurately weighed, in approximately 150 milliliters of 
distilled water by mixing thoroughly. Dilute to 250 milliliters with 
distilled water and mix thoroughly. Transfer a 10.0-milliliter aliquot 
of this solution to a 100-milliliter volumetric flask, add approximately 
75 milliliters of distilled water and 5.0 milliliters of 5N NaOH, dilute 
to volume with water, and mix thoroughly. Treat a sample of the 
tetracycline hydrochloride working standard in the same manner. Exactly 
6 minutes after the addition of the NaOH, determine the absorbance of 
each solution at 380 nanometers, using a suitable spectrophotometer and 
distilled water as the blank. Determine the percent absorptivity of the 
sample relative to the absorptivity of the standard using the following 
calculation:
[GRAPHIC] [TIFF OMITTED] TC01AP94.063

where: m = Percent moisture in the sample.

    (9) 4-Epianhydrotetracycline. Proceed as directed in Sec. 436.309 of 
this chapter.
    (10) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (11) Identity. Proceed as directed in Sec. 436.308 of this chapter.
[43 FR 11160, Mar. 17, 1978; 43 FR 34456, Aug. 4, 1978, as amended at 44 
FR 31636, June 1, 1979; 46 FR 60568, Dec. 11, 1981; 50 FR 19920, May 13, 
1985]



Sec. 446.82  Tetracycline phosphate complex.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline phosphate complex is [4S-
(4,4a,5a,6, 12a)] - 4 - 
(dimethylamino) - 1,4,4a,5,5a,6,11,12a - octahydro - 3,6,10,12,12a - 
pentahydroxy - 6 - methyl - 1,11 - dioxo - 2 - naphthacenecarboxamide 
phosphate complex. It is so purified and dried that:
    (i) Its potency is not less than 750 micrograms per milligram on the 
anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 9 percent.
    (iv) Its pH in an aqueous suspension containing 10 milligrams per 
milliliter is not less than 2.0 and not more than 4.0.
    (v) When calculated on the anhydrous basis, its absorptivity at 380 
nanometers relative to that of the tetracycline hydrochloride working 
standard similarly treated is 82.0plus-minus4.9 percent.
    (vi) Its 4-epianhydrotetracycline content is not more than 2.0 
percent.
    (vii) It passes the identity test, showing a presence of phosphate, 
a content of not more than 0.2 percent chloride, and a content of not 
more than 1 percent tetracycline base.
    (viii) It is crystalline.
    (2) Labeling. In addition to the requirements of Sec. 432.5 of this 
chapter, each such package shall bear on its label or labeling the 
statement ``For use only in the manufacture of nonparenteral drugs''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, absorptivity, 4-epianhydro tetracycline content, identity, and 
crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 60 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1N hydrochloric 
acid to obtain a concentration of 1,000 micrograms of tetracycline 
hydrochloride per milliliter (estimated). Further dilute an aliquot of

[[Page 786]]

the stock solution with sterile distilled water to the reference 
concentration of 0.24 microgram of tetracycline hydrochloride per 
milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
suspension containing 10 milligrams of the sample per milliliter.
    (5) Absorptivity. Dissolve approximately 40 milligrams of the 
sample, accurately weighed, in 2.0 milliliters of 0.1N HCl and dilute to 
250 milliliters with distilled water. Transfer a 10.0 milliliter aliquot 
of this solution to a 100-milliliter volumetric flask, add about 75 
milliliters of distilled water and 5.0 milliliters of 5N NaOH, dilute to 
volume with water, and mix thoroughly. Treat a sample of the 
tetracycline hydrochloride working standard in the same manner. Exactly 
6 minutes after the addition of NaOH, determine the absorbance of each 
solution at 380 nanometers, using a suitable spectrophotometer and 
distilled water as the blank. Determine the percent absorptivity of the 
sample relative to the absorptivity of the standard using the following 
calculations:
[GRAPHIC] [TIFF OMITTED] TC01AP94.064

where: m = Percent moisture in the sample.

    (6) 4-Epianhydrotetracycline. Proceed as directed in Sec. 436.309 of 
this chapter.
    (7) Identity--(i) Presence of phosphate. Prepare a filtrate as 
follows: Suspend 100 milligrams of the sample in 10 milliliters of 
distilled water and filter a small portion by gravity. Transfer 1.0 
milliliter of the filtrate to a 100-milliliter glass-stoppered cylinder, 
add 10.0 milliliters of distilled water, 2.0 milliliters of ammonium 
molybdate test solution, 1.0 milliliter of stannous chloride test 
solution, and 10.0 milliliters of isobutyl alcohol-benzene mixture (1:1 
ratio), all in the order named. Shake vigorously for 1 minute, allow the 
layers to separate, and examine the top organic layer. In the presence 
of phosphate, the top layer turns blue.
    (ii) Chloride content. To 1.0 milliliter of the filtrate prepared as 
directed in the first sentence of paragraph (b)(7)(i) of this section, 
add 1 drop of silver nitrate test solution and 1 drop of nitric acid. 
Any turbidity produced is not greater than that obtained by similarly 
treating 1.0 milliliter of 0.057N hydrochloric acid.
    (iii) Determination of percent tetracycline base. This test is used 
to determine the quantity of tetracycline present as base in mixtures 
with phosphate salts.
    (a) Reagents--(1) 1,4-Dioxane.
    (2) Purified dioxane: Pass the dioxane through a column of Amberlite 
IRA 400 (OH-) resin or equivalent.
    (3) Perchloric acid, 0.01N: Dilute 0.84 milliliter of 70 percent 
perchloric acid to 1,000 milliliters with purified dioxane; standardize 
at least once every 2 days, as follows: Weigh accurately about 70 
milligrams of diphenylguanidine, and dissolve in 50 milliliters of ethyl 
alcohol in a 250-milliliter flask. Add two drops of methyl red, and 
titrate with the perchloric acid solution until the yellow color changes 
to orange. Deduct the volume of the perchloric acid consumed by 50 
milliliters of the ethyl alcohol, and calculate the normality. Each 
2.113 milligrams of diphenylguanidine is equivalent to 1 milliliter of 
0.01N perchloric acid.
    (4) Methyl red indicator: Dissolve 100 milligrams of methyl red in 
100 milliliters of methyl alcohol.
    (b) Procedure. Place an accurately weighed 1-gram sample into a 50-
milliliter Erlenmeyer flask, add 10.0 milliliters of purified dioxane 
and shake the mixture manually for about 2 minutes. Allow to settle, 
decant all the supernatant liquid into a 50-milliliter polyethylene 
centrifuge tube, cover with Parafilm (or equivalent), and centrifuge 
until clear (about 3 minutes).

[[Page 787]]

Pipette 5.0 milliliters of the clear, supernatant solution into a 50-
milliliter beaker, stir magnetically, and titrate with 0.01N perchloric 
acid, using methyl red as the indicator. The endpoint is the last color 
change to orange when a drop of titrant is added. Calculate the percent 
tetracycline base as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.065

    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[43 FR 11161, Mar. 17, 1978; 43 FR 34456, Aug. 4, 1978, as amended at 50 
FR 19920, May 13, 1985]



                      Subpart B--Oral Dosage Forms



Sec. 446.110  Chlortetracycline hydrochloride capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chlortetracycline hydrochloride capsules 
are composed of chlortetracycline hydrochloride and one or more suitable 
and harmless diluents, lubricants, and fillers. Each capsule contains 
50, 100, or 250 milligrams of chlortetracycline hydrochloride. The 
potency is satisfactory if it is not less than 90 percent and not more 
than 120 percent of the number of milligrams of chlortetracycline 
hydrochloride that it is represented to contain. The loss on drying is 
not more than 1 percent. The chlortetracycline hydrochloride used 
conforms to the standards prescribed by Sec. 446.10(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The chlortetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, crystallinity, and identity.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:
    (a) The chlortetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 36 capsules.
    (b) Test and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 0.01N hydrochloric acid to give a 
stock solution of convenient concentration. Blend for 3 to 5 minutes. 
Remove an aliquot of the stock solution and further dilute with sterile 
distilled water to the reference concentration of 0.06 microgram of 
chlortetracycline hydrochloride per milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[43 FR 11162, Mar. 17, 1978; 43 FR 34456, Aug. 4, 1978, as amended at 50 
FR 19920, May 13, 1985]



Sec. 446.115  Demeclocycline oral dosage forms.



Sec. 446.115a  Demeclocycline oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Demeclocycline oral suspension is 
composed of demeclocycline with or without one or more suitable and 
harmless buffer substances, suspending and stabilizing agents, and 
preservatives suspended in a suitable and harmless vehicle. Each 
milliliter contains demeclocycline equivalent to 15 milligrams of 
demeclocycline hydrochloride. Its potency is satisfactory if it is not 
less than 90 percent and not more than 125 percent of the number of 
milligrams of demeclocycline hydrochloride equivalent that it is 
represented to contain. The pH is not less than 4 and not more than 5.8. 
The demeclocycline used conforms to the standards prescribed by 
Sec. 446.15(a)(1).

[[Page 788]]

    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The demeclocycline used in making the batch for potency, 
moisture, pH, absorptivity, crystallinity, and identity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The demeclocycline used in making the batch: 10 packages, each 
containing approximately 250 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Transfer an accurately measured representative portion of the well-
shaken suspension to an appropriate-sized volumetric flask and dilute to 
volume with 0.1N hydrochloric acid to obtain a stock solution of 
convenient concentration containing not less than 150 micrograms of 
demeclocycline hydrochloride per milliliter (estimated). Mix well. 
Further dilute an aliquot of the stock solution with sterile distilled 
water to the reference concentration of 0.100 microgram of 
demeclocycline hydrochloride per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11162, Mar. 17, 1978; 43 
FR 50677, Oct. 31, 1978; 50 FR 19920, May 13, 1985]



Sec. 446.115b  Demeclocycline for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Demeclocycline for oral suspension is 
composed of demeclocycline with or without one or more suitable and 
harmless buffer substances, preservatives, diluents, colorings, and 
flavorings. When reconstituted as directed in the labeling, each 
milliliter contains demeclocycline equivalent to 15 milligrams of 
demeclocycline hydrochloride. Its potency is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of demeclocycline hydrochloride equivalent that it is 
represented to contain. Its moisture content is not more than 5 percent. 
The demeclocycline used conforms to the standards prescribed by 
Sec. 446.15(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The demeclocycline used in making the batch for potency, 
moisture, pH, absorptivity, crystallinity, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The demeclocycline used in making the batch: 10 packages, each 
containing approximately 250 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Transfer an accurately 
measured representative portion of the well-shaken suspension to an 
appropriate-sized volumetric flask and dilute to volume with 0.1N 
hydrochloric acid to obtain a stock solution of convenient concentration 
containing not less than 150 micrograms of demeclocycline per milliliter 
(estimated). Further dilute an aliquot of the stock solution with 
sterile distilled water to the reference concentration of 0.100 
microgram of demeclocycline hydrochloride per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11162, Mar. 17, 1978; 50 
FR 19920, May 13, 1985]



Sec. 446.116  Demeclocycline hydrochloride oral dosage forms.



Sec. 446.116a  Demeclocycline hydrochloride tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality,

[[Page 789]]

and purity. Demeclocycline hydrochloride tablets are composed of 
demeclocycline hydrochloride with one or more suitable and harmless 
diluents, lubricants, binders, and flavorings. Each tablet contains 75 
milligrams, 150 milligrams, or 300 milligrams of demeclocycline 
hydrochloride. Its potency is satisfactory if it is not less than 90 
percent and not more than 125 percent of the number of milligrams of 
demeclocycline hydrochloride that it is represented to contain. Its loss 
on drying is not more than 2 percent. It shall disintegrate within 30 
minutes. The demeclocycline hydrochloride used conforms to the standards 
prescribed by Sec. 446.16(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The demeclocycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, crystallinity, and identity.
    (b) The batch for potency, loss on drying, and disintegration time.
    (ii) Samples required:
    (a) The demeclocycline hydrochloride used in making the batch: 10 
packages, each containing approximately 250 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient 0.1N hydrochloric acid to give a stock 
solution of convenient concentration containing not less than 150 
micrograms of demeclocycline hydrochloride per milliliter (estimated). 
Blend for 3 to 5 minutes. Remove an aliquot of the stock solution and 
further dilute with sterile distilled water to the reference 
concentration of 0.100 microgram of demeclocycline hydrochloride per 
milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter.
[39 FR 19076 , May 30, 1974, as amended at 43 FR 11162, Mar. 17, 1978; 
50 FR 19920, May 13, 1985]



Sec. 446.116b  [Reserved]



Sec. 446.116c  Demeclocycline hydrochloride capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Demeclocycline hydrochloride capsules are 
composed of demeclocycline hydrochloride, with one or more suitable and 
harmless diluents and lubricants, enclosed in a gelatin capsule. Each 
capsule contains 75 milligrams, 150 milligrams, or 300 milligrams of 
demeclocycline hydrochloride. Its potency is satisfactory if it is not 
less than 90 percent and not more than 125 percent of the number of 
milligrams of demeclocycline hydrochloride that it is represented to 
contain. Its loss on drying is not more than 2 percent, except that if 
starch is used as a diluent the loss on drying is not more than 8 
percent. The demeclocycline hydrochloride used conforms to the standards 
prescribed by Sec. 446.16(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The demeclocycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, crystallinity, and identity.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:
    (a) The demeclocycline hydrochloride used in making the batch: 10 
packages, each containing approximately 250 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient

[[Page 790]]

0.1N hydrochloric acid to give a stock solution of convenient 
concentration containing not less than 150 micrograms of demeclocycline 
hydrochloride per milliliter (estimated). Blend for 3 to 5 minutes. 
Remove an aliquot of the stock solution and further dilute with sterile 
distilled water to the reference concentration of 0.100 microgram of 
demeclocycline hydrochloride per milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11162, Mar. 17, 1978; 50 
FR 19920, May 13, 1985]



Sec. 446.120  Doxycycline hyclate oral dosage forms.



Sec. 446.120a  Doxycycline hyclate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Doxycycline hyclate capsules are composed 
of doxycycline hyclate and one or more suitable and harmless lubricants 
and diluents enclosed in a gelatin capsule. Each capsule contains 
doxycycline hyclate equivalent to either 50, 100, or 300 milligrams of 
doxycycline. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
doxycycline that it is represented to contain. The moisture content is 
not more than 5.0 percent. It passes the identity test for the presence 
of the doxycycline moiety. The doxycycline hyclate used conforms to the 
standards prescribed by Sec. 446.20.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The doxycycline hyclate used in making the batch for potency, 
moisture, pH, doxycycline content, identity, and crystallinity.
    (b) The batch for potency, moisture, and identity.
    (ii) Samples required:
    (a) The doxycycline hyclate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch: A minimum of 36 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Blend a representative number of capsules in a high-speed glass blender 
jar containing 0.1N hydrochloric acid to obtain a stock solution of 
convenient concentration containing not less than 150 micrograms of 
doxycycline per milliliter (estimated). Blend for 3 to 5 minutes. Remove 
an aliquot of the stock solution and further dilute with sterile 
distilled water to the reference concentration of 0.100 microgram of 
doxycycline per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Identity. Proceed as directed in Sec. 436.308 of this chapter, 
except prepare the standard and sample solutions as follows: Dissolve 
precise amounts of the doxycycline capsule contents and of the 
doxycycline working standard in methanol and further dilute each 
solution to a concentration of 1 milligram of doxycycline per 
milliliter. Prepare the sample-standard mixed solution by mixing equal 
volumes of the final standard and sample solutions. The standard and 
sample must each produce a major, yellow fluorescent spot with the same 
Rf value, and the standard-sample mixed solution must show no 
separation of major spots.
[39 FR 19076, May 30, 1974. Redesignated at 39 FR 41250, Nov. 26, 1974, 
and amended at 43 FR 11162, Mar. 17, 1978; 44 FR 20667, Apr. 6, 1979; 50 
FR 19920, May 13, 1985]



Sec. 446.120b  Doxycycline calcium oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Doxycycline calcium oral suspension is 
prepared from doxycycline hyclate and contains one or more suitable and 
harmless buffer substances, preservatives, diluents, solvents, 
colorings, and flavorings. Its potency is satisfactory if it is not less 
than 90 percent and not more than 125 percent of the number of 
milligrams of doxycycline that it is represented to contain. Its pH is 
not less than 6.5 and

[[Page 791]]

not more than 8.0. It passes the identity test for the presence of the 
doxycycline moiety. The doxycycline hyclate used conforms to the 
standards prescribed by Sec. 446.20(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The doxycycline hyclate used in making the batch for potency, 
moisture, pH, doxycycline content, identity, and crystallinity.
    (b) The batch for potency, pH, and identity.
    (ii) Samples required:
    (a) The doxycycline hyclate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Transfer an appropriate aliquot of the suspension to a volumetric flask 
and dissolve with sufficient 0.1N hydrochloric acid to give a stock 
solution of convenient concentration (containing not less than 150 
micrograms of doxycycline per milliliter in acid). Further dilute an 
aliquot of the stock solution with sterile distilled water to the 
reference concentration of 0.100 microgram of doxycycline per milliliter 
(estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
    (3) Identity. Proceed as directed in Sec. 436.308 of this chapter, 
except prepare the standard and sample solutions as follows: Dissolve 
precise amounts of the doxycycline calcium oral suspension and of the 
doxycycline working standard in methanol and further dilute each 
solution with methanol to a concentration of 1 milligram of doxycycline 
per milliliter. Prepare the sample-standard mixed solution by mixing 
equal volumes of the final concentration of the sample and standard 
solutions. The sample and standard must each produce a major, yellow 
fluorescent spot with the same Rf value, and the sample-
standard mixed solution must show no separation of major spots.
[39 FR 41250, Nov. 11, 1974, as amended at 45 FR 16476, Mar. 14, 1980; 
50 FR 19920, May 13, 1985]



Sec. 446.120c  Doxycycline hyclate tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Doxycycline hyclate tablets contain 
doxycycline hyclate with or without one or more disintegrants, 
lubricants, colorings, and coating substances. Each tablet contains 
doxycycline hyclate equivalent to 50 or 100 milligrams of doxycycline. 
Its potency is satisfactory if it is not less than 90 percent and not 
more than 120 percent of the number of milligrams of doxycycline that it 
is represented to contain. Its moisture content is not more than 5.0 
percent. It passes the dissolution test. It passes the identity test. 
The doxycycline hyclate conforms to the standards prescribed by 
Sec. 446.20(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The doxycycline hyclate used in making the batch for potency, 
moisture, pH, doxycycline content, identity, and crystallinity.
    (b) The batch for potency, moisture, dissolution, and identity.
    (ii) Samples required:
    (a) The doxycycline hyclate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch: A minimum of 100 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar containing 0.1N hydrochloric acid to obtain a stock solution of 
convenient concentration containing not less than 150 micrograms of 
doxycycline per milliliter (estimated).

[[Page 792]]

Blend for 3 to 5 minutes. Remove an aliquot of the stock solution and 
further dilute with sterile distilled water to the reference 
concentration of 0.100 microgram of doxycycline per milliliter 
(estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter, except:
    (i) In lieu of paragraph (a) of that section, a distance of 
4.50.5 centimeters should be maintained between the lower 
edge of the stirring blade and the lowest inner surface of the vessel 
during the test; and
    (ii) In lieu of paragraph (d) of that section, use the 
interpretation described in the United States Pharmacopeia XX 
dissolution test. The quantity, Q (the amount of doxycycline dissolved) 
is 55 percent at 60 minutes and 85 percent at 90 minutes.
    (4) Identity. Proceed as directed in Sec. 436.308 of this chapter, 
except prepare the sample and standard solutions as follows: Grind 
tablet to a powder. Dissolve precise amount of the doxycycline tablet 
and of the doxycycline working standard in methanol and further dilute 
each solution to a concentration of 1 milligram of doxycycline per 
milliliter. Prepare the sample-standard mixed solution by mixing equal 
volumes of the final standard and sample solutions. The standard and 
sample must each produce a major, yellow fluorescent spot with the same 
Rf value and the standard-sample mixed solution must show no 
separation of major spots.
[46 FR 7273, Jan. 23, 1981, as amended at 48 FR 23813, May 27, 1983; 48 
FR 51293, Nov. 8, 1983; 50 FR 19920, May 13, 1985]



Sec. 446.120d  Doxycycline hyclate pellet-filled capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Doxycycline hyclate pellet-filled 
capsules contain pellets which are composed of doxycycline hyclate and 
suitable and harmless diluents, binders, and lubricants. Each capsule 
contains doxycycline hyclate equivalent to 100 milligrams of 
doxycycline. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
doxycycline that it is represented to contain. The moisture content is 
not more than 5.0 percent. It passes the acid resistance test. It passes 
the dissolution test. The doxycycline hyclate conforms to the standards 
prescribed by Sec. 446.20(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The doxycycline hyclate used in making the batch for potency, 
safety, moisture, pH, doxycycline content, identity, and crystallinity.
    (b) The batch for potency, moisture, acid resistance, and 
dissolution.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The doxycycline hyclate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch: A minimum of 100 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing 0.1N hydrochloric acid to obtain a stock solution 
of convenient concentration containing not less than 150 micrograms of 
doxycycline per milliliter (estimated). Blend for 3 to 5 minutes. Remove 
an aliquot of the stock solution and further dilute with sterile 
distilled water to the reference concentration of 0.100 microgram of 
doxycycline per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Acid resistance. Proceed as directed in Sec. 436.543 of this 
chapter.
    (4) Dissolution. Empty the contents of one pellet-filled capsule 
into the basket and proceed as directed in Sec. 436.544 of this chapter. 
The quantity Q (the amount of doxycycline dissolved) is 85 percent at 30 
minutes.
[50 FR 41679, Oct. 15, 1985, as amended at 55 FR 11584, Mar. 29, 1990]

[[Page 793]]



Sec. 446.121  Doxycycline monohydrate oral dosage forms.



Sec. 446.121a  Doxycycline monohydrate for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Doxycycline monohydrate for oral 
suspension is doxycycline monohydrate with one or more suitable and 
harmless buffer substances, preservatives, diluents, colorings, and 
flavorings. Its moisture content is not more than 3 percent. It passes 
the identity test for the presence of the doxycycline moiety. When 
prepared as directed in the labeling, each milliliter contains the 
equivalent of 5 milligrams of doxycycline and its pH is not less than 
5.0 and not more than 6.5. Its potency is satisfactory if it is not less 
than 90 percent and not more than 125 percent of the number of 
milligrams of doxycycline that it is represented to contain. The 
doxycycline monohydrate used conforms to the standards prescribed by 
Sec. 446.21(a)(1).
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, this drug shall be labeled ``doxycycline for oral 
suspension''.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The doxycycline monohydrate used in making the batch for 
potency, moisture, pH, doxycycline content, identity, and crystallinity.
    (b) The batch for potency, moisture, pH, and identity.
    (ii) Samples required:
    (a) The doxycycline monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute the sample as directed in the labeling. Transfer an 
accurately measured representative portion of the well-shaken suspension 
to an appropriate-sized volumetric flask and dilute to volume with 0.1N 
hydrochloric acid to obtain a stock solution of convenient concentration 
containing not less than 150 micrograms of doxycycline per milliliter 
(estimated). Further dilute an aliquot of the stock solution with 
sterile distilled water to the reference concentration of 0.100 
microgram of doxycycline per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Reconstitute as directed in the labeling and proceed as 
directed in Sec. 436.202 of this chapter, using the undiluted sample.
    (4) Identity. Proceed as directed in Sec. 436.308 of this chapter, 
except prepare the standard and sample solutions as follows: Dissolve 
precise amounts of the doxycycline monohydrate for oral suspension and 
of the doxycycline working standard in methanol and further dilute each 
solution to a concentration of 1 milligram of doxycycline per 
milliliter. Prepare the sample-standard mixed solution by mixing equal 
volumes of the final concentration of the sample and standard solutions. 
The sample and standard must each produce a major, yellow fluorescent 
spot with the same Rf value and the sample-standard mixed 
solution must show no separation of major spots.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11163, Mar. 17, 1978; 50 
FR 19920, May 13, 1985. Redesignated at 55 FR 6637, Feb. 26, 1990]



Sec. 446.121b  Doxycycline monohydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Doxycycline monohydrate capsules are 
composed of doxycycline monohydrate and one or more suitable and 
harmless lubricants and diluents enclosed in a gelatin capsule. Each 
capsule contains doxycycline monohydrate equivalent to 100 milligrams of 
doxycycline. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
doxycycline that it is represented to contain. The moisture content is 
not more than 5.5 percent. It passes the dissolution test. It passes the 
identity test. The doxycycline monohydrate used conforms to the 
standards prescribed by Sec. 446.21.

[[Page 794]]

    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The doxycycline monohydrate used in making the batch for 
potency, moisture, pH, doxycycline content, identity, and crystallinity.
    (B) The batch for potency, moisture, dissolution, and identity.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research:
    (A) The doxycycline monohydrate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (B) The batch: A minimum of 100 capsules.
    (b) Tests and methods of assay--(1) Doxycycline potency. Proceed as 
directed in Sec. 436.216 of this chapter, using ambient temperature, an 
ultraviolet detection system operating at a wavelength of 280 
nanometers, a 4.6-millimeter X 3-centimeter guard column containing 5- 
to 10-micrometer diameter octyl silane chemically bonded to totally 
porous microsilica particles, a 3.9-millimeter X 30-centimeter 
analytical column packed with octadecyl silane chemically bonded to 
porous silica or ceramic microparticles, 5 to 10 micrometers in 
diameter, a flow rate of 1.5 milliliters per minute, and a 10-microliter 
loop injector. Reagents, working standard and sample solutions, system 
suitability requirements, and calculations are as follows:
    (i) Reagents--(A) 0.1M sodium phosphate buffer. Prepare a solution 
containing 13.8 grams of monobasic sodium phosphate per liter of 
distilled water.
    (B) Mobile phase. Mix 450 milliliters of 0.1M monobasic sodium 
phosphate and 550 milliliters of methanol. Add 3 milliliters of N,N-
dimethyl-n-octylamine. Adjust the pH to 8.0 with 5N sodium hydroxide. 
Filter the mobile phase through a suitable glass filter or equivalent 
that is capable of removing particulate contamination to 1 micron in 
diameter. Degas the mobile phase just prior to its introduction into the 
chromatograph pumping system.
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(A) Working standard solution. Dissolve an accurately weighed 
portion of the doxycycline hyclate working standard in sufficient 0.1N 
hydrochloric acid to obtain a known concentration of about 1,000 
micrograms of doxycycline per milliliter. Further dilute with distilled 
water to a concentration of 40 micrograms of doxycycline activity per 
milliliter. Filter through a membrane filter of 0.5 micron or finer 
porosity.
    (B) Sample solution. Remove, as completely as possible, the contents 
of a representative number of capsules. Mix the combined contents and 
transfer an accurately weighed portion of the powder, equivalent to 
about 100 milligrams of doxycycline, to a 100-milliliter volumetric 
flask. Add 20 milliliters of 0.1N hydrochloric acid and sonicate for 5 
minutes. Dilute to mark with 0.1N hydrochloric acid. Further 
quantitatively dilute an aliquot of this solution with distilled water 
to a concentration of 40 micrograms of doxycycline activity per 
milliliter (estimated). Filter through a membrane filter of 0.5 micron 
or finer porosity. Content uniformity analyses may be obtained from 
sample solutions prepared as above except that the contents of one 
capsule are quantitatively transferred to the 100-milliliter volumetric 
flask.
    (C) Resolution test solution. Dissolve 50 milligrams of doxycycline 
in 25 milliliters of distilled water. Pipet 5 milliliters of this 
solution into a 25-milliliter volumetric flask and heat on a steam bath 
for 60 minutes. Transfer the contents of the flask to a small beaker and 
evaporate to dryness. Dissolve the residue in distilled water, transfer 
to a 25-milliliter volumetric flask, dilute to mark with distilled 
water, mix, and filter through Whatman No. 1 filter paper. Use this 
solution to determine the resolution factor.
    (iii) System suitability requirements--(A) Asymmetry factor. 
Calculate the asymmetry factor (As), measured at a point 5 
percent of the peak height from the baseline, as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.251

where:


[[Page 795]]


a=Horizontal distance from point of ascent to a point of a maximum peak 
          height; and
b=Horizontal distance from the point of maximum peak height to point of 
          descent.


The asymmetry factor (As) is satisfactory if it is not less 
than 1.4 and not more than 2.0
    (B) Efficiency of the column. From the number of theoretical plates 
(n) calculated as described in Sec. 436.216(c)(2) of this chapter 
calculate the reduced plate height (hr) as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.252

where:

L=Length of the column in centimeters;
n=number of theoretical plates; and
dp=Average diameter of the particles in analytical column 
          packing in micrometers.


The absolute efficiency (hr) is satisfactory if it is not 
more than 37.5 for the doxycycline peak.
    (C) The resolution (R) between peaks for doxycycline and epi-
doxycycline is satisfactory if it is not less than 1.5.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent) of 5 replicate 
injections is satisfactory if it is not more than 2.0 percent.
     (E) Capacity factor (k'). Calculate the capacity factor (k') for 
doxycycline as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.253

where:

tr=Retention time of doxycycline in minutes; and
to=Column dead time in minutes, which is estimated from the 
          following equation:
          [GRAPHIC] [TIFF OMITTED] TR01JA93.254
          
where:

D=Column diameter in centimeters;
L=Column length in centimeters;
0.75=Average total column porosity; and
F=Flow rate in milliliters per minute.


The capacity factor (k') for doxycycline is satisfactory if it is not 
less than 1.5 and not more than 2.5. If the system suitability 
requirements have been met, then proceed as described in Sec. 436.216(b) 
of this chapter. Alternate chromatographic conditions are acceptable 
provided reproducibility and resolution are comparable to the system 
described. However, the sample preparation described in paragraph 
(b)(1)(ii)(B) of this section should not be changed.
    (iv) Calculations. Calculate the doxycycline content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.255
    
where:

Au=Area of the doxycycline peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the doxycycline peak in the chromatogram of the 
          working standard;
Ps=Doxycycline activity in the doxycycline working standard 
          solution in micrograms per milliliter;
d=Dilution factor of the sample; and
n=Number of capsules in the sample assayed.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q (the amount of doxycycline dissolved) is 85 
percent at 60 minutes.
    (4) Identity. The high-pressure liquid chromatogram of the sample 
determined in paragraph (b)(1) of this section compares qualitatively to 
that of the doxycycline working standard.
[55 FR 6637, Feb. 26, 1990]



Sec. 446.150  Methacycline hydrochloride oral dosage forms.



Sec. 446.150a  Methacycline hydrochloride capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Methacycline hydrochloride capsules are 
composed of methacycline hydrochloride and one or more suitable and 
harmless lubricants and diluents enclosed in a gelatin capsule. Each 
capsule contains methacycline hydrochloride equivalent to either 70 
milligrams of methacycline, 140 milligrams of methacycline, or 280 
milligrams of methacycline. Its potency is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of methacycline that

[[Page 796]]

it is represented to contain. The moisture content is not more than 7.5 
percent. The methacycline hydrochloride used conforms to the standards 
prescribed by Sec. 446.50(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The methacycline hydrochloride used in making the batch for 
potency, moisture, pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The methacycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Blend a representative number of capsules in a high-speed glass blender 
jar containing sufficient sterile distilled water to give a stock 
solution of convenient concentration. Further dilute an aliquot of the 
stock solution with sterile distilled water to the reference 
concentration of 0.06 microgram of methacycline per milliliter 
(estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11163, Mar. 17, 1978; 46 
FR 46313, Sept. 18, 1981; 50 FR 19920, May 13, 1985]



Sec. 446.150b  Methacycline hydrochloride oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Methacycline hydrochloride oral 
suspension contains methacycline hydrochloride and one or more suitable 
and harmless buffers, dispersants, diluents, colorings, flavorings, and 
preservatives. It contains methacycline hydrochloride equivalent to 14 
milligrams of methacycline per milliliter. Its potency is satisfactory 
if it is not less than 90 percent and not more than 125 percent of the 
number of milligrams of methacycline that it is represented to contain. 
Its pH is not less than 6.5 nor more than 8.0. The methacycline 
hydrochloride used conforms to the standards prescribed by 
Sec. 446.50(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The methacycline hydrochloride used in making the batch for 
potency, moisture, pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency and pH.
    (ii) Samples required.
    (a) The methacycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Transfer an accurately measured representative portion of the well-
shaken suspension to an appropriate-sized volumetric flask, and dilute 
to volume with sterile distilled water. Mix well. Remove an aliquot of 
the stock solution and further dilute with sterile distilled water to 
the reference concentration of 0.06 microgram of methacycline per 
milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter using 
the undiluted sample.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11163, Mar. 17, 1978; 50 
FR 19920, May 13, 1985]



Sec. 446.160  Minocycline hydrochloride oral dosage forms.



Sec. 446.160a  Minocycline hydrochloride tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Minocycline hydrochloride tablets are 
composed of minocycline hydrochloride and one or more suitable

[[Page 797]]

and harmless diluents, binders, lubricants, coloring, and coating 
substances. Each tablet contains minocycline hydrochloride equivalent to 
100 milligrams of minocycline. Its potency is satisfactory if it 
contains not less than 90 percent and not more than 115 percent of the 
number of milligrams of minocycline that it is represented to contain. 
Its moisture content is not more than 12 percent. The tablets 
disintegrate within 30 minutes. The minocycline hydrochloride used 
conforms to the standards prescribed by Sec. 446.60(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The minocycline hydrochloride used in making the batch for 
potency, moisture, pH, epi-minocycline content, identity, crystallinity, 
residue on ignition, and absorptivity.
    (b) The batch for potency, moisture, and disintegration time.
    (ii) Samples required:
    (a) The minocycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 446.60(b)(1) of this part, except prepare the sample solution and 
calculate the minocycline potency as follows:
    (i) Sample solution. Grind a representative number of tablets in a 
mortar and pestle. Wash the ground tablets into a volumetric flask 
containing mobile phase (described in Sec. 446.60(b)(1)(i)(c) of this 
part) and shake to dissolve. Dilute with mobile phase to give a stock 
solution of convenient concentration. Filter the stock solution. Further 
dilute using mobile phase to obtain a solution containing 500 micrograms 
of minocycline activity per milliliter (estimated). Use this solution 
within 3 hours of preparation.
    (ii) Calculations. Calculate the minocycline content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.256
    
where:
Au=Area of the minocycline peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the minocycline peak in the chromatogram of the 
          minocycline working standard;
Ps=Minocycline activity in the minocycline working standard 
          solution in micrograms per milliliter;
d = Dilution factor of the sample; and
n = Number of tablets in the sample assayed.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure described in paragraph (e)(1) of that 
section.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11163, Mar. 17, 1978; 44 
FR 22058, Apr. 13, 1979; 50 FR 19920, May 13, 1985; 53 FR 32609, Aug. 
26, 1988]



Sec. 446.160b  Minocycline hydrochloride capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Minocycline hydrochloride capsules are 
composed of minocycline hydrochloride and one or more suitable and 
harmless lubricants and diluents enclosed in a gelatin capsule. Each 
capsule contains minocycline hydrochloride equivalent to 50 or 100 
milligrams of minocycline. Its potency is satisfactory if it is not less 
than 90 percent and not more than 115 percent of the number of 
milligrams of minocycline that it is represented to contain. Its 
moisture content is not more than 12 percent. The minocycline 
hydrochloride used conforms to the standards prescribed by 
Sec. 446.60(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The minocycline hydrochloride used in making the batch for 
potency, moisture, pH, epi-minocycline content,

[[Page 798]]

identity, crystallinity, residue on ignition, and absorptivity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The minocycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 446.60(b)(1) of this part, except prepare the sample solution and 
calculate the minocycline potency as follows:
    (i) Sample solution. Open a representative number of capsules and 
empty the contents into a volumetric flask containing mobile phase 
(described in Sec. 446.60(b)(1)(i)(c) of this part) and shake to 
dissolve. Dilute with mobile phase to give a stock solution of 
convenient concentration. Filter the stock solution. Remove an aliquot 
of the stock solution and further dilute with mobile phase to obtain a 
solution containing 500 micrograms of minocycline activity per 
milliliter (estimated). Use this solution within 3 hours of preparation.
    (ii) Calculations. Calculate the minocycline content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.257
    
where:
Au= Area of the minocycline peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As= Area of the minocycline peak in the chromatogram of the 
          minocycline working standard:
Ps= Minocycline activity in the minocycline working standard 
          solution in micrograms per milliliter;
d = Dilution factor of the sample; and
n = Number of capsules in the sample assayed.
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11163, Mar. 17, 1978; 44 
FR 22058, Apr. 13, 1979; 50 FR 19920, May 13, 1985; 53 FR 32609, Aug. 
26, 1988]



Sec. 446.160c  Minocycline hydrochloride oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Minocycline hydrochloride oral suspension 
is minocycline hydrochloride with one or more suitable flavorings, 
wetting agents, preservatives, and diluents in an aqueous vehicle. Each 
milliliter contains minocycline hydrochloride equivalent to 10 
milligrams of minocycline. Its potency is satisfactory if it is not less 
than 90 percent and not more than 130 percent of the number of 
milligrams of minocycline that it is represented to contain. Its pH is 
not less than 7.0 and not more than 9.0. The minocycline hydrochloride 
used conforms to the standards prescribed by Sec. 446.60(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this subchapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The minocycline hydrochloride used in making the batch for 
potency, moisture, pH, epi-minocycline content, identity, crystallinity, 
residue on ignition, and absorptivity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The minocycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 446.60(b)(1) of this part, except prepare the sample solution and 
calculate the minocycline potency as follows:
    (i) Sample solution. Transfer an accurately measured 5-milliliter 
portion of the well-shaken suspension to a 100-milliliter volumetric 
flask. Dilute to mark with mobile phase (described in 
Sec. 446.60(b)(1)(i)(c) of this part) and mix well. Filter this solution 
and use within 3 hours of its preparation.
    (ii) Calculations. Calculate the minocycline content as follows:

[[Page 799]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.258


where:
Au= Area of the minocycline peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As= Area of the minocycline peak in the chromatogram of the 
          minocycline working standard:
Ps= Minocycline activity in the minocycline working standard 
          solution in micrograms per milliliter; and
d = Dilution factor of the sample.
    (2) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using the undiluted sample.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11163, Mar. 17, 1978; 44 
FR 22058, Apr. 13, 1979; 50 FR 19920, May 13, 1985; 53 FR 32609, Aug. 
26, 1988]



Sec. 446.165  Oxytetracycline oral dosage forms.



Sec. 446.165a  Oxytetracycline tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline tablets are tablets 
composed of oxytetracycline and one or more suitable and harmless, 
diluents, binders, lubricants, colorings, and coating substances. The 
potency of each tablet is 250 milligrams of oxytetracycline. Its potency 
is satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of oxytetracycline that it is 
represented to contain. The moisture content is not more than 7.5 
percent. They shall disintegrate within 1 hour. The oxytetracycline used 
conforms to the standards prescribed by Sec. 446.65(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oxytetracycline used in making the batch for potency, 
moisture, pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency, moisture, and disintegration time.
    (ii) Samples required:
    (a) The oxytetracycline used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient 0.1N hydrochloric acid to obtain a stock 
solution of convenient concentration containing not less than 150 
micrograms of oxytetracycline per milliliter (estimated). Blend for 3 to 
5 minutes. Remove an aliquot of the stock solution and further dilute 
with sterile distilled water to the reference concentration of 0.24 
microgram of oxytetracycline per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the method described in paragraph (e)(1) of that section.
[43 FR 11163, Mar. 17, 1978; 43 FR 34456, Aug. 4, 1978, as amended at 50 
FR 19920, May 13, 1985]



Secs. 446.165b--446.165c  [Reserved]



Sec. 446.165d  Oxytetracycline for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline for oral suspension is 
oxytetracycline with one or more suitable and harmless buffer 
substances, preservatives, diluents, colorings, and flavorings. When 
prepared as directed in the labeling, each milliliter contains 50 
milligrams of oxytetracycline. Its potency is satisfactory if it is not 
less than 90 percent and not more than 115 percent of the number of 
milligrams of oxytetracycline that it is represented to contain. Its 
loss on drying is not more than 2 percent. When reconstituted as 
directed in the labeling, its pH is not less than 5.5 and not more than 
7.5. The oxytetracycline used conforms to the standards prescribed by 
Sec. 446.65(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 800]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oxytetracycline used in making the batch for potency, 
moisture, pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency, loss on drying, and pH.
    (ii) Samples required:
    (a) The oxytetracycline used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Transfer an accurately 
measured representative portion of the well-shaken suspension to an 
appropriate-sized volumetric flask and dilute to volume with 0.1N 
hydrochloric acid to obtain a stock solution of convenient concentration 
containing not less than 150 micrograms of oxytetracycline per 
milliliter (estimated). Mix well. Further dilute an aliquot of the stock 
solution with sterile distilled water to the reference concentration of 
0.24 microgram of oxytetracycline per milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) pH. Reconstitute as directed in the labeling and proceed as 
directed in Sec. 436.202 of this chapter.
[43 FR 11164, Mar. 17, 1978, as amended at 48 FR 51293, Nov. 8, 1983; 50 
FR 19920, May 13, 1985]



Sec. 446.166  Oxytetracycline calcium oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline calcium oral suspension 
contains oxytetracycline calcium with one or more suitable and harmless 
buffer substances, suspending and stabilizing agents, flavorings, 
colorings, solvents, and preservatives suspended in a suitable and 
harmless vehicle. It may contain N-acetyl glucosamine. Each milliliter 
contains a quantity of oxytetracycline calcium equivalent to 25 
milligrams of oxytetracycline. Its potency is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of oxytetracycline that it is represented to contain. Its pH 
is not less than 5.0 and not more than 8.0. The oxytetracycline calcium 
used conforms to the standards prescribed by Sec. 446.66(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oxytetracycline calcium used in making the batch for 
potency, moisture, pH, calcium content, identity, and crystallinity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The oxytetracycline calcium used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Transfer an accurately measured representative portion of the sample to 
an appropriate-sized volumetric flask and dilute to volume with 0.1N 
hydrochloric acid to give a stock solution of convenient concentration 
containing not less than 150 micrograms of oxytetracycline per 
milliliter (estimated). Mix well. Remove an aliquot of the stock 
solution and further dilute with sterile distilled water to the 
reference concentration of 0.24 microgram of oxytetracycline per 
milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[43 FR 11164, Mar. 17, 1978 as amended at 43 FR 50677, Oct. 31, 1978; 45 
FR 16476, Mar. 14, 1980; 50 FR 19920, May 13, 1985]



Sec. 446.167  Oxytetracycline hydrochloride capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline hydrochloride capsules 
are gelatin capsules

[[Page 801]]

containing oxytetracycline hydrochloride with or without one or more 
suitable and harmless buffers, preservatives, diluents, binders, and 
lubricants. They may contain glucosamine hydrochloride. Each capsule 
contains 50 milligrams, 100 milligrams, 125 milligrams, or 250 
milligrams of oxytetracycline. Its potency is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of oxytetracycline that it is represented to contain. The 
loss on drying is not more than 5.0 percent. It passes the dissolution 
test. The oxytetracycline hydrochloride used conforms to the standards 
prescribed by Sec. 446.67(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The oxytetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency, loss on drying, and dissolution.
    (ii) Samples required:
    (a) The oxytetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 0.1N hydrochloric acid to give a stock 
solution of convenient concentration containing not less than 150 
micrograms of oxytetracycline per milliliter (estimated). Blend for 3 to 
5 minutes. Remove an aliquot of the stock solution and further dilute 
with sterile distilled water to the reference concentration of 0.24 
microgram of oxytetracycline per milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter, except in lieu of paragraph (a) of that section, a distance of 
4.50.5 centimeters should be maintained between the lower 
edge of the stirring blade and the lowest inner surface of the vessel 
during the test. The quantity Q (the amount of oxytetracycline 
dissolved) is 60 percent within 30 minutes and 85 percent within 60 
minutes.
[43 FR 11164, Mar. 17, 1978, as amended at 44 FR 48189, Aug. 17, 1979; 
47 FR 32938, July 30, 1982; 48 FR 51293, Nov. 3, 1983; 49 FR 37058, 
Sept. 21, 1984; 50 FR 19920, May 13, 1985]



Sec. 446.180  Tetracycline oral dosage forms.



Secs. 446.180a--446.180b  [Reserved]



Sec. 446.180c  Tetracycline oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline oral suspension is composed 
of tetracycline with or without one or more suitable and harmless buffer 
substances, suspending and stabilizing agents, and preservatives, 
suspended in a suitable and harmless vehicle. Each milliliter contains 
tetracycline equivalent to 25 milligrams of tetracycline hydrochloride. 
Its potency is satisfactory if it contains the equivalent of not less 
than 90 percent and not more than 125 percent of the number of 
milligrams of tetracycline hydrochloride that it is represented to 
contain. Its pH is not less than 3.5 and not more than 6.0. Its 4-
epianhydrotetracycline content is not more than 5.0 percent. The 
tetracycline used conforms to the standards prescribed by 
Sec. 446.80(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tetracycline used in making the batch for potency, moisture, 
pH, absorptivity, 4-epianhydrotetracycline content, crystallinity, and 
identity.
    (b) The batch for potency, pH, and 4-epianhydrotetracycline content.
    (ii) Samples required:

[[Page 802]]

    (a) The tetracycline used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Transfer an accurately measured representative portion of the well-
shaken suspension to an appropriate-sized volumetric flask and dilute to 
volume with 0.1N hydrochloric acid to give a stock solution of 
convenient concentration containing not less than 150 micrograms of 
tetracycline hydrochloride per milliliter (estimated). Mix well. Remove 
an aliquot of the stock solution and further dilute with sterile 
distilled water to the reference concentration of 0.24 microgram of 
tetracycline hydrochloride per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted suspension.
    (3) 4-Epianhydrotetracycline. Proceed as directed in Sec. 436.309(b) 
of this chapter.
[43 FR 11164, Mar. 17, 1978; 43 FR 34456, Aug. 4, 1978, as amended at 45 
FR 16472, 16476, Mar. 14, 1980; 50 FR 19920, May 13, 1985]



Sec. 446.181  Tetracycline hydrochloride oral dosage forms.



Secs. 446.181a--446.181c  [Reserved]



Sec. 446.181d  Tetracycline hydrochloride tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline hydrochloride tablets 
contain tetracycline hydrochloride with or without one or more buffer 
substances, preservatives, diluents, binders, lubricants, colorings, and 
flavorings. Each tablet contains 250 milligrams or 500 milligrams of 
tetracycline hydrochloride. Its potency is satisfactory if it contains 
not less than 90 percent and not more than 125 percent of the number of 
milligrams of tetracycline hydrochloride that it is represented to 
contain. Its loss on drying is not more than 3.0 percent. It passes the 
dissolution test. Its 4-epianhydrotetracycline content is not more than 
3.0 percent. The tetracycline hydrochloride used conforms to the 
standards prescribed by Sec. 446.81(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, 4-epianhydrotetracycline 
content, crystallinity, and identity.
    (b) The batch for potency, loss on drying, dissolution, and 4-
epianhydrotetracycline content.
    (ii) Samples required:
    (a) The tetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient 0.1N hydrochloric acid to obtain a stock 
solution of convenient concentration containing not less than 150 
micrograms of tetracycline hydrochloride per milliliter (estimated). 
Blend for 3 to 5 minutes. Remove an aliquot of the stock solution and 
further dilute with sterile distilled water to the reference 
concentration of 0.24 microgram of tetracycline hydrochloride per 
milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter, except in lieu of paragraph (a) of that section, a distance of 
4.5plus-minus0.5 centimeters should be maintained between the 
lower edge of the stirring blade and the lowest inner surface of the 
vessel during the test. The quantity Q (the amount of tetracycline 
hydrochloride dissolved) is 60 percent within 30 minutes and 85 percent 
within 60 minutes.

[[Page 803]]

    (4) 4-Epianhydrotetracycline. Proceed as directed in Sec. 436.309 of 
this chapter.
[43 FR 11165, Mar. 17, 1978, as amended at 44 FR 48189, Aug. 17, 1979; 
47 FR 32938, July 30, 1982; 48 FR 51293, Nov. 8, 1983; 49 FR 37058, 
Sept. 21, 1984; 50 FR 19920, May 13, 1985]



Sec. 446.181e  Tetracycline hydrochloride capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline hydrochloride capsules are 
composed of tetracycline hydrochloride with or without one or more 
suitable and harmless buffer substances, preservatives, diluents, 
binders, lubricants, colorings, and flavorings enclosed in a gelatin 
capsule. Each capsule contains 50, 100, 125, 250, or 500 milligrams of 
tetracycline hydrochloride. Its potency is satisfactory if it is not 
less than 90 percent and not more than 125 percent of the number of 
milligrams of tetracycline hydrochloride that it is represented to 
contain. Its loss on drying is not more than 4 percent. Its 4-
epianhydrotetracycline content is not more than 3.0 percent. It passes 
the dissolution test. The tetracycline hydrochloride used conforms to 
the standards prescribed by Sec. 446.81(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, 4-epianhydrotetracycline 
content, crystallinity, and identity.
    (b) The batch for potency, loss on drying, 4-epianhydrotetracycline 
content, and dissolution.
    (ii) Samples required:
    (a) The tetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 0.1N hydrochloric acid to obtain a 
stock solution of convenient concentration containing not less than 150 
micrograms of tetracycline hydrochloride per milliliter (estimated). 
Blend for 3 to 5 minutes. Remove an aliquot of the stock solution and 
further dilute with sterile distilled water to the reference 
concentration of 0.24 microgram of tetracycline hydrochloride per 
milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) 4-Epianhydrotetracycline. Proceed as directed in Sec. 436.309 of 
this chapter.
    (4) Dissolution. Proceed as directed in Sec. 436.215 of this chapter 
except in lieu of paragraph (a) of that section, a distance of 
4.5plus-minus0.5 centimeters should be maintained between the 
lower edge of the stirring blade and the lowest inner surface of the 
vessel during the test. The quantity Q (the amount of tetracycline 
hydrochloride dissolved), except for the 500-milligram capsule, is 60 
percent within 30 minutes and 85 percent within 60 minutes. For the 500-
milligram capsule, the quantity Q is 50 percent within 30 minutes, 70 
percent within 60 minutes, and 85 percent within 90 minutes.
[43 FR 11166, Mar. 17, 1978, as amended at 44 FR 48189, Aug. 17, 1979; 
47 FR 32938, July 30, 1982; 48 FR 51293, Nov. 8, 1983; 49 FR 37058, 
Sept. 21, 1984; 50 FR 19920, May 13, 1985]



Sec. 446.182  Tetracycline phosphate complex capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline phosphate complex capsules 
contain tetracycline phosphate complex with or without one or more 
buffer substances, preservatives, diluents, binders, lubricants, 
colorings, and flavorings enclosed in a gelatin capsule. Each capsule 
contains tetracycline phosphate complex equivalent to 50, 100, 125, 250, 
or 500 milligrams of tetracycline hydrochloride. Its potency is 
satisfactory if it contains the equivalent of not less than 90 percent 
and not more than 125 percent of the number of milligrams of 
tetracycline hydrochloride that it is represented to contain. Its loss 
on drying is not more than 9.0 percent. Its 4-

[[Page 804]]

epianhydrotetracycline content is not more than 3.0 percent. The 
tetracycline phosphate complex used conforms to the standards prescribed 
by Sec. 446.82 (a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification, samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tetracycline phosphate complex used in making the batch for 
potency, moisture, pH, absorptivity, 4-epianhydrotetracycline content, 
identity, and crystallinity.
    (b) The batch for potency, loss on drying, and 4-
epianhydrotetracycline content.
    (ii) Samples required:
    (a) The tetracycline phosphate complex used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 0.1N hydrochloric acid to obtain a 
stock solution of convenient concentration containing not less than 150 
micrograms of tetracycline hydrochloride per milliliter (estimated). 
Blend for 3 to 5 minutes. Remove an aliquot of the stock solution and 
further dilute with sterile distilled water to the reference 
concentration of 0.24 microgram of tetracycline per milliliter 
(estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) 4-Epianhydrotetracycline. Proceed as directed in Sec. 436.309 of 
this chapter.
[43 FR 11166, Mar. 17, 1978, as amended at 50 FR 19920, May 13, 1985]



                   Subpart C--Injectable Dosage Forms



Sec. 446.220  Doxycycline hyclate for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Doxycycline hyclate for injection is a 
dry mixture of doxycycline hyclate and a buffer substance. Its potency 
is satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of doxycycline that it is 
represented to contain. It is sterile. It is nonpyrogenic. It contains 
no depressor substances. Its loss on drying is not more than 2.0 
percent. Its pH when reconstituted as directed in the labeling is not 
less than 1.8 and not more than 3.3. It passes the identity test for the 
presence of the doxycycline moiety. The doxycycline hyclate used 
conforms to the standards prescribed by Sec. 446.20a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this subchapter.
    (3) Requests for certification: samples. In addition to complying 
with the requirements of Sec. 431.1 of this subchapter, each such 
request shall contain:
    (i) Results of tests and assays on:
    (a) The doxycycline hyclate used in making the batch for potency, 
moisture, pH, doxycycline content, identity, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, depressor 
substances, loss on drying, pH, and identity.
    (ii) Samples required:
    (a) The doxycycline hyclate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this subchapter, preparing the sample for assay as 
follows: Reconstitute as directed in the labeling. Using a suitable 
hypodermic needle and syringe, remove all of the withdrawable contents 
from each container if it is represented as a single-dose container; or 
if the labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute the solution thus obtained with 
sufficient 0.1N hydrochloric

[[Page 805]]

acid to give a stock solution of convenient concentration (containing 
not less than 150 micrograms of doxycycline in acid). Further dilute an 
aliquot of the stock solution with sterile distilled water to the 
reference concentration of 0.100 microgram of doxycycline per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this 
subchapter, using the method described in paragraph (e)(1) of that 
section, except use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this 
subchapter, using a solution containing 7.5 milligrams of doxycycline 
per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
subchapter.
    (6) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
subchapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using the drug reconstituted as directed in the labeling.
    (8) Identity. Proceed as directed in Sec. 436.308 of this 
subchapter, except prepare the standard and sample solutions as follows: 
Dissolve precise amounts of the doxycycline hyclate for injection and of 
the doxycycline working standard in methanol and further dilute each 
solution to a concentration of 1 milligram of doxycycline per 
milliliter. Prepare the sample-standard mixed solution by mixing equal 
volumes of the final concentration of the sample and standard solutions. 
The sample and standard must each produce a major, yellow fluorescent 
spot with the same Rf value and the sample-standard mixed 
solution must show no separation of major spots.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11166, Mar. 17, 1978; 43 
FR 34457, Aug. 4, 1978; 46 FR 60568, Dec. 11, 1981; 50 FR 19920, May 13, 
1985]



Sec. 446.260  Sterile minocycline hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile minocycline hydrochloride is a 
lyophilized powder of minocycline hydrochloride. Its potency is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of minocycline that it is 
represented to contain. It is sterile. It is nonpyrogenic. It contains 
no depressor substance. Its moisture content is not more than 3.0 
percent. Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 2.0 and not more than 3.5. The minocycline 
hydrochloride used conforms to the standards prescribed by 
Sec. 446.60(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The minocycline hydrochloride used in making the batch for 
potency, moisture, pH, epi-minocycline content, identity, crystallinity, 
residue on ignition, and absorptivity.
    (b) The batch for potency, sterility, pyrogens, depressor 
substances, moisture, and pH.
    (ii) Samples required:
    (a) The minocycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 446.60(b)(1) of this part, except prepare the sample solution and 
calculate the minocycline potency as follows:
    (i) Sample solution. Reconstitute as directed in the labeling. Using 
a suitable hypodermic needle and syringe, remove the withdrawable 
contents from each container represented as a single-dose container; or 
if the labeling specifies the amount of potency in a given volume of the 
resultant preparation, withdraw an accurately measured representation 
portion from each container. Dilute the sample thus obtained with 
sufficient mobile phase (described in Sec. 446.60(b)(1)(i)(c) of this 
part) to give a stock solution of convenient concentration. Filter the 
stock solution. Further dilute an aliquot of this stock solution with 
mobile phase to obtain a solution containing 500 micrograms of

[[Page 806]]

minocycline activity per milliliter (estimated). Use this solution 
within 3 hours of preparation.
    (ii) Calculations--(a) Calculate the minocycline content of the 
single-dose vial as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.259

where:
Au= Area of the minocycline peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As= Area of the minocycline peak in the chromatogram of the 
          minocycline working standard:
Ps= Minocycline activity in the minocycline working standard 
          solution in micrograms per milliliter; and
d = Dilution factor of the sample.

    (b) Calculate the minocycline content of the multiple-dose vial as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.260

where:
Au= Area of the minocycline peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As= Area of the minocycline peak in the chromatogram of the 
          minocycline working standard:
Ps= Minocycline activity in the minocycline working standard 
          solution in micrograms per milliliter;
d = Dilution factor of the sample.
n = Volume of sample solution assayed.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 5 milligrams per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the sample preparation described in paragraph (d)(4) of that 
section.
    (7) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams of minocycline per 
milliliter.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11166, Mar. 17, 1978; 43 
FR 34457, Aug. 4, 1978; 44 FR 22058, Apr. 13, 1979; 46 FR 60568, Dec. 
11, 1981; 50 FR 19920, May 13, 1985; 53 FR 32609, Aug. 26, 1988; 54 FR 
47205, Nov. 13, 1989]



Sec. 446.265  Oxytetracycline injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline injection is a solution 
of oxytetracycline with or without one or more suitable and harmless 
buffer substances, anesthetics, preservatives, antioxidants, complexing 
agents, and solvents. Each milliliter contains 50 milligrams or 125 
milligrams of oxytetracycline. Its potency is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of oxytetracycline that it is represented to contain. It is 
sterile. It is nonpyrogenic. It contains no depressor substances. Its pH 
is not less than 8.0 and not more than 9.0. The oxytetracycline used 
conforms to the standards prescribed by Sec. 446.65a(a)(1), except 
sterility, pyrogens, and depressor substances.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oxytetracycline used in making the batch for potency, 
moisture, pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, depressor 
substances, and pH.
    (ii) Samples required:
    (a) The oxytetracycline used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for

[[Page 807]]

assay as follows: Transfer an accurately measured representative 
quantity of the sample to an appropriate-sized volumetric flask. Dilute 
to volume with 0.1N hydrochloric acid to obtain a stock solution of 
convenient concentration containing not less than 150 micrograms of 
oxytetracycline per milliliter (estimated). Further dilute an aliquot of 
the stock solution with sterile distilled water to the reference 
concentration of 0.24 microgram of oxytetracycline per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 5.0 milligrams of oxytetracycline per 
milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[43 FR 11166, Mar. 17, 1978, as amended at 46 FR 60568, Dec. 11, 1981; 
48 FR 51293, Nov. 8, 1983; 50 FR 19920, May 13, 1985]



Sec. 446.267  Oxytetracycline hydrochloride for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline hydrochloride for 
injection is a dry mixture of oxytetracycline hydrochloride and a 
suitable buffer substance. Its potency is satisfactory if it is not less 
than 90 percent and not more than 115 percent of the number of 
milligrams of oxytetracycline that it is represented to contain. It is 
sterile. It is nonpyrogenic. It contains no depressor substances. Its 
loss on drying is not more than 3.0 percent. Its pH in an aqueous 
solution containing 25 milligrams per milliliter is not less than 1.8 
and not more than 2.8. The oxytetracycline hydrochloride used conforms 
to the standards prescribed by Sec. 446.67a(a)(1), except sterility, 
pyrogens, and depressor substances.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oxytetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, depressor 
substances, loss on drying, and pH.
    (ii) Samples required:
    (a) The oxytetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Then, using a suitable 
hypodermic needle and syringe, promptly remove all the withdrawable 
contents if it is represented as a single dose container; or, if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from the container. Dilute the sample thus obtained with 
sufficient 0.1N hydrochloric acid to obtain a stock solution of 
convenient concentration containing not less than 150 micrograms of 
oxytetracycline per milliliter (estimated). Further dilute an aliquot of 
the stock solution with sterile distilled water to the reference 
concentration of 0.24 microgram of oxytetracycline per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in 436.32(b) of this chapter, 
using a solution containing 5.0 milligrams per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter,

[[Page 808]]

using the diluent recommended by the manufacturer in the labeling for 
the drug.
    (6) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 25 milligrams per milliliter.
[43 FR 11167, Mar. 17, 1978, as amended at 46 FR 60568, Dec. 11, 1981; 
50 FR 19920, May 13, 1985]



Sec. 446.275  Rolitetracycline injectable dosage forms.



Sec. 446.275a  Rolitetracycline for intravenous use.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Rolitetracycline for intravenous use is a 
dry mixture of rolitetracycline and one or more suitable buffer 
substances. Its potency is satisfactory if it is not less than 90 
percent and not more than 115 percent of the number of milligrams of 
rolitetracycline that it is represented to contain. It is sterile. It is 
nonpyrogenic. It contains no depressor substances. Its loss on drying is 
not more than 5 percent. When reconstituted as directed in the labeling, 
its pH is not less than 3.0 and not more than 4.5. The rolitetracycline 
used conforms to the standards prescribed by Sec. 446.75a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this subchapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this subchapter, each such 
request shall contain:
    (i) Results of tests and assays on:
    (a) The rolitetracycline used in making the batch for potency, 
moisture, pH, crystallinity, absorptivity, and identity.
    (b) The batch for potency, sterility, pyrogens, depressor 
substances, loss on drying, and pH.
    (ii) Samples required:
    (a) The rolitetracycline used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this subchapter, preparing the sample for assay as 
follows: Reconstitute the sample as directed in the labeling. Using a 
suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is represented as a single dose container; or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute the sample thus obtained with 
sufficient distilled water to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of the stock solution with 
distilled water to the reference concentration of 0.24 microgram of 
rolitetracycline per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this 
subchapter, using the method described in paragraph (e)(1) of that 
section, except use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this 
subchapter, using a solution containing 5.0 milligrams of 
rolitetracycline per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
subchapter.
    (6) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using a solution prepared as directed in the labeling.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11167, Mar. 17, 1978; 46 
FR 46313, Sept. 18, 1981; 46 FR 60568, Dec. 11, 1981; 50 FR 19920, May 
13, 1985]



Sec. 446.275b  Rolitetracycline for intramuscular use.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Rolitetracycline for intramuscular use is 
a dry mixture of rolitetracycline and one or more suitable buffer 
substances and anesthetic agents. Its potency is satisfactory if it is 
not less than 90 percent and not more than 115 percent of the number of 
milligrams of rolitetracycline that it is

[[Page 809]]

represented to contain. It is sterile. It is nonpyrogenic. Its loss on 
drying is not more than 5 percent. When reconstituted as directed in the 
labeling, its pH is not less than 3.0 and not more than 4.5. The 
rolitetracycline used conforms to the standards prescribed by 
Sec. 446.75a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this subchapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this subchapter, each such 
request shall contain:
    (i) Results of tests and assays on:
    (a) The rolitetracycline used in making the batch for potency, 
depressor substances, moisture, pH, crystallinity, absorptivity, and 
identity.
    (b) The batch for potency, sterility, pyrogens, loss on drying, and 
pH.
    (ii) Samples required:
    (a) The rolitetracycline used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this subchapter, preparing the sample for assay as 
follows: Reconstitute the sample as directed in the labeling. Then using 
a suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is represented as a single dose container; or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute the sample thus obtained with 
sufficient distilled water to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of the stock solution with 
distilled water to the reference concentration of 0.24 microgram of 
rolitetracycline per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this 
subchapter, using the method described in paragraph (e)(1) of that 
section, except use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this 
subchapter, using a solution containing 5.0 milligrams of 
rolitetracycline per milliliter.
    (4) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
subchapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using a solution prepared as directed in the labeling.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11167, Mar. 17, 1978; 46 
FR 46313, Sept. 18, 1981; 46 FR 60568, Dec. 11, 1981; 50 FR 19920, May 
13, 1985]



Sec. 446.276  Rolitetracycline nitrate injectable dosage forms.



Sec. 446.276a  Rolitetracycline nitrate for intravenous use.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Rolitetracycline nitrate for intravenous 
use is a dry mixture of rolitetracycline nitrate and one or more 
suitable buffer substances. Its potency is satisfactory if it contains 
not less than 90 percent and not more than 115 percent of the number of 
milligrams of rolitetracycline that it is represented to contain. It is 
sterile. It is nonpyrogenic. It contains no depressor substances. Its 
loss on drying is not more than 5 percent. When reconstituted as 
directed in the labeling, its pH is not less than 2.5 nor more than 4.0. 
The rolitetracycline nitrate used conforms to the standards prescribed 
by Sec. 446.76a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this subchapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this subchapter, each such 
request shall contain:
    (i) Results of tests and assays on:
    (a) The rolitetracycline nitrate used in making the batch for 
potency, moisture, pH, crystallinity, absorptivity, and identity.
    (b) The batch for potency, sterility, pyrogens, depressor 
substances, loss on drying, and pH.
    (ii) Samples required:
    (a) The rolitetracycline nitrate used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (b) The batch:

[[Page 810]]

    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this subchapter, preparing the sample for assay as 
follows: Reconstitute the sample as directed in the labeling. Using a 
suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is [repre-]sented as a single dose container; or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute the sample thus obtained with 
sufficient distilled water to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of the stock solution with 
distilled water to the reference concentration of 0.24 microgram of 
rolitetracycline per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this 
subchapter, using the method described in paragraph (e)(1) of that 
section, except use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this 
subchapter, using a solution containing 5.0 milligrams of 
rolitetracycline per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
subchapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using a solution prepared as directed in the labeling.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11168, Mar. 17, 1978; 43 
FR 34457, Aug. 4, 1978; 46 FR 46313, Sept. 18, 1981; 46 FR 60568, Dec. 
11, 1981; 50 FR 19920, May 13, 1985]



Sec. 446.276b  Rolitetracycline nitrate for intramuscular use.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Rolitetracycline nitrate for 
intramuscular use is a dry mixture of rolitetracycline nitrate, one or 
more suitable buffer substances, and lidocaine hydrochloride. Its 
potency is satisfactory if it is not less than 90 percent and not more 
than 115 percent of the number of milligrams of rolitetracycline that it 
is represented to contain. It is sterile. It is nonpyrogenic. Its loss 
on drying is not more than 5 percent. When reconstituted as directed in 
the labeling, its pH is not less than 2.5 nor more than 4.0. The 
rolitetracycline nitrate used conforms to the standards prescribed by 
Sec. 446.76a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this subchapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this subchapter, each such 
request shall contain:
    (i) Results of tests and assays on:
    (a) The rolitetracycline nitrate used in making the batch for 
potency, depressor substances, moisture, pH, crystallinity, 
absorptivity, and identity.
    (b) The batch for potency, sterility, pyrogens, loss on drying, and 
pH.
    (ii) Samples required:
    (a) The rolitetracycline nitrate used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this subchapter, preparing the sample for assay as 
follows: Reconstitute the sample as directed in the labeling. Then using 
a suitable hypodermic needle and syringe, remove all of the withdrawable 
contents if it is represented as a single-dose container; or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute the sample thus obtained with 
sufficient distilled water to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of the stock solution with 
distilled water to the reference concentration of 0.24 microgram of 
rolitetracycline per milliliter (estimated).

[[Page 811]]

    (2) Sterility. Proceed as directed in Sec. 436.20 of this 
subchapter, using the method described in paragraph (e)(1) of that 
section, except use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this 
subchapter, using a solution containing 5.0 milligrams of 
rolitetracycline per milliliter.
    (4) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
subchapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using a solution prepared as directed in the labeling.
[39 FR 19076, May 30, 1974, as amended at 43 FR 11168, Mar. 30, 1978; 46 
FR 46313, Sept. 18, 1981; 46 FR 60568, Dec. 11, 1981; 50 FR 19920, May 
13, 1985]



Sec. 446.281  Tetracycline hydrochloride injectable dosage forms.



Sec. 446.281a  Sterile tetracycline hydrochloride.

    The requirements for certification and the tests and methods of 
assay for sterile tetracycline hydrochloride packaged for dispensing are 
described in Sec. 446.81a.



Sec. 446.281c  Tetracycline hydrochloride for intramuscular use.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline hydrochloride for 
intramuscular use is a dry mixture of tetracycline hydrochloride, 
magnesium chloride, or magnesium ascorbate and one or more suitable 
buffer substances, with or without one or more suitable preservatives 
and anesthetic agents, and with or without one or more suitable 
solubilizers and stabilizers. Its potency is satisfactory if it is not 
less than 90 percent and not more than 115 percent of the number of 
milligrams of tetracycline hydrochloride that it is represented to 
contain. It is sterile. It is nonpyrogenic. Its loss on drying is not 
more than 5.0 percent. Its pH in an aqueous solution containing 10 
milligrams per milliliter is not less than 2.0 and not more than 3.0. 
Its 4-epianhydrotetracycline content is not more than 3.0 percent. The 
tetracycline hydrochloride used conforms to the standards prescribed by 
Sec. 446.81a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tetracycline hydrochloride used in making the batch for 
potency, depressor substances, loss on drying, pH, absorptivity, 4-
epianhydrotetracycline content, crystallinity, and identity.
    (b) The batch for potency, sterility, pyrogens, loss on drying, pH, 
and 4-epianhydrotetracycline content.
    (ii) Samples required:
    (a) The tetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 10 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute the sample as directed in the labeling. Then, using a 
suitable hypodermic needle and syringe, remove all the withdrawable 
contents if it is represented as a single dose container; or, if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute the sample thus obtained with 
sufficient 0.1N hydrochloric acid to obtain a stock solution of 
convenient concentration containing not less than 150 micrograms of 
tetracycline hydrochloride per milliliter (estimated). Further dilute an 
aliquot of the stock solution with sterile distilled water to the 
reference concentration of 0.24 microgram of tetracycline hydrochloride 
per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 5.0 milligrams of tetracycline hydrochloride 
per milliliter.
    (4) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.

[[Page 812]]

    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (6) 4-Epianhydrotetracycline. Proceed as directed in Sec. 436.309 of 
this chapter.
[44 FR 31636, June 1, 1979, as amended at 46 FR 60568, Dec. 11, 1981; 47 
FR 13326, Mar. 30, 1982; 50 FR 19920, May 13, 1985]



Sec. 446.281d  Tetracycline hydrochloride for intravenous use.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline hydrochloride for 
intravenous use is a dry mixture of tetracycline hydrochloride with one 
or more suitable and harmless stabilizing agents. Its potency is 
satisfactory if it contains not less than 90 percent and not more than 
115 percent of the number of milligrams of tetracycline hydrochloride 
that it is represented to contain. It is sterile. It is nonpyrogenic. It 
contains no depressor substances. Its loss on drying is not more than 
5.0 percent. Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 2.0 and not more than 3.0. Its 4-
epianhydrotetracycline content is not more than 3.0 percent. The 
tetracycline hydrochloride used conforms to the standards prescribed by 
Sec. 446.81a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, 4-epianhydrotetracycline 
content, cystallinity, and identity.
    (b) The batch for potency, sterility, pyrogens, depressor 
substances, loss on drying, pH, and 4-epianhydrotetracycline content.
    (ii) Samples required:
    (a) The tetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum for 10 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute the sample as directed in the labeling. Then, using a 
suitable hypodermic needle and syringe, remove all of the withdrawal 
contents if it is represented as a single dose container; or, if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute the sample thus obtained with 
sufficient 0.1N hydrochloric acid to obtain a stock solution of 
convenient concentration containing not less than 150 micrograms of 
tetracycline hydrochloride per milliliter (estimated). Further dilute an 
aliquot of the stock solution with sterile distilled water to the 
reference concentration of 0.24 microgram of tetracycline hydrochloride 
per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use diluting fluid D in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 5.0 milligrams of tetracycline hydrochloride 
per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (8) 4-Epianhydrotetracycline. Proceed as directed in Sec. 436.309 of 
this chapter.
[44 FR 31636, June 1, 1979, as amended at 46 FR 60568, Dec. 11, 1981; 47 
FR 13326, Mar. 30, 1982; 50 FR 19920, May 13, 1985]



Sec. 446.282  Tetracycline phosphate complex for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline phosphate complex for 
injection is a dry mixture of tetracycline phosphate complex, magnesium 
chloride or magnesium ascorbate and one or more suitable buffer 
substances, with or without one

[[Page 813]]

or more suitable preservatives and anesthetic agents, and with or 
without one or more suitable solubilizers and stabilizers. Its potency 
is satisfactory if it contains not less than 90 percent and not more 
than 115 percent of the number of milligrams of tetracycline 
hydrochloride that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its loss on drying is not more than 5 percent. Its pH in 
an aqueous solution containing 10 milligrams per milliliter is not less 
than 2.0 and not more than 3.0. Its 4-epianhydrotetracycline content is 
not more than 3.0 percent. The tetracycline phosphate complex conforms 
to the standards prescribed by Sec. 446.82(a)(1), and it contains no 
depressor substance.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tetracycline phosphate complex used in making the batch for 
potency, moisture, pH, depressor substances, absorptivity, 4-
epianhydrotetracycline content, identity, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, loss on drying, pH, 
and 4-epianhydrotetracycline content.
    (ii) Samples required:
    (a) The tetracycline phosphate complex used in making the batch: 10 
packages, each containing approximately 300 milligrams, and one package 
containing approximately 1 gram.
    (b) The batch: A minimum of 10 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Reconstitute the sample 
as directed in the labeling. Then, using a suitable hypodermic needle 
and syringe, remove all the withdrawable contents if it is represented 
as a single-dose container; or, if the labeling specifies the amount of 
potency in a given volume of the resultant preparation, remove an 
accurately measured representative portion from each container. Dilute 
the sample thus obtained with sufficient 0.1N hydrochloric acid to 
obtain a stock solution of convenient concentration containing not less 
than 150 micrograms of tetracycline hydrochloride per milliliter 
(estimated). Further dilute an aliquot of the stock solution with 
sterile distilled water to the reference concentration of 0.24 microgram 
of tetracycline hydrochloride per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use 50 milligrams in lieu of 300 milligrams of sample.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 5.0 milligrams of tetracycline hydrochloride 
per milliliter.
    (4) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (6) Depressor substances in the tetracycline phosphate complex used 
in making the batch. Proceed as directed in Sec. 436.35 of this chapter. 
Prepare the test solution by dissolving 40 milligrams of sample in 2.0 
milliliters of 0.1N hydrochloric acid and diluting with sterile 
distilled water (diluent 3) to the prescribed concentration.
    (7) 4-Epianhydrotetracycline. Proceed as directed in Sec. 436.309 of 
this chapter.
[43 FR 11168, Mar. 17, 1978, as amended at 46 FR 60568, Dec. 11, 1981; 
50 FR 19920, May 13, 1985]



                   Subpart D--Ophthalmic Dosage Forms



Sec. 446.310  Chlortetracycline hydrochloride ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chlortetracycline hydrochloride 
ophthalmic ointment contains chlortetracycline hydrochloride in a 
suitable and harmless ointment base. Each gram contains 10 milligrams of 
chlortetracycline hydrochloride. Its potency is satisfactory if it 
contains not less than 90 percent and not more than 125 percent of the 
number of milligrams of chlortetracycline hydrochloride that it is 
represented to contain. It is sterile. Its moisture content is not more 
than 0.5 percent. It passes

[[Page 814]]

the test for metal particles. The chlortetracycline hydrochloride used 
conforms to the standards prescribed by Sec. 446.10a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The chlortetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, crystallinity, and identity.
    (b) The batch for potency, sterility, moisture, and metal particles.
    (ii) Samples required:
    (a) The chlortetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the sample into a 
separatory funnel containing approximately 50 milliliters of peroxide-
free ether. Shake the sample and ether until homogeneous. Add 20 to 25 
milliliters of 0.01N hydrochloric acid and shake well. Allow the layers 
to separate. Remove the acid layer and repeat the extraction procedure 
with each of three more 20- to 25-milliliter quantities of 0.01N 
hydrochloric acid. Combine the extractives in a suitable volumetric 
flask and dilute to volume with 0.01N hydrochloric acid. Further dilute 
an aliquot with sterile distilled water to the reference concentration 
of 0.06 microgram of chlortetracycline hydrochloride per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) Metal particles. Proceed as directed in Sec. 436.206 of this 
chapter.
[43 FR 11169, Mar. 17, 1978, as amended at 50 FR 19920, May 13, 1985]



Sec. 446.367  Oxytetracycline hydrochloride ophthalmic dosage forms.



Sec. 446.367c  Oxytetracycline hydrochloride-hydrocortisone acetate ophthalmic suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline hydrochloride-
hydrocortisone acetate ophthalmic suspension is oxytetracycline 
hydrochloride and hydrocortisone acetate in a suitable and harmless oil 
base containing aluminum tristearate. Each milliliter contains 
oxytetracycline hydrochloride equivalent to 5 milligrams of 
oxytetracycline and 15 milligrams of hydrocortisone acetate. Its potency 
is satisfactory if it contains not less than 90 percent and not more 
than 115 percent of the number of milligrams of oxytetracycline that it 
is represented to contain. It is sterile. Its moisture content is not 
more than 1 percent. The oxytetracycline hydrochloride used conforms to 
the standards prescribed by Sec. 446.67a (a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oxytetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, crystallinity, and identity.
    (b) The batch for potency, sterility, and moisture.
    (ii) Samples required:
    (a) The oxytetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of five immediate 
containers.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.

[[Page 815]]

    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place an accurately measured, representative portion of the sample into 
a high-speed glass blender jar containing 1.0 milliliter of polysorbate 
80 and sufficient 0.1N hydrochloric acid to obtain a stock solution of 
convenient concentration containing not less than 150 micrograms of 
oxytetracycline per milliliter (estimated). Blend for 3 to 5 minutes. 
Further dilute an aliquot with sterile distilled water to the reference 
concentration of 0.24 microgram of oxytetracycline per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section, except 
use 0.25 milliliter of the sample in lieu of 1.0 milliliter.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[43 FR 11169, Mar. 17, 1978, as amended at 50 FR 19920, May 13, 1985]



Sec. 446.367e  Oxytetracycline hydrochloride-polymyxin B sulfate ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline hydrochloride-polymyxin B 
sulfate ophthalmic ointment is oxytetracycline hydrochloride and 
polymyxin B sulfate in a suitable and harmless ointment base. Each gram 
contains oxytetracycline hydrochloride equivalent to 5 milligrams of 
oxytetracycline and polymyxin B sulfate equivalent to 10,000 units of 
polymyxin B. Its oxytetracycline content is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of oxytetracycline that it is represented to contain. Its 
polymyxin B content is satisfactory if it is not less than 90 percent 
and not more than 125 percent of the number of units of polymyxin B that 
it is represented to contain. It is sterile. Its moisture content is not 
more than 1 percent. It passes the test for metal particles. The 
oxytetracycline hydrochloride used conforms to the standards prescribed 
by Sec. 446.67a (a)(1). The polymyxin B sulfate used conforms to the 
standards prescribed by Sec. 448.30a(a)(1) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oxytetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, crystallinity, and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
pH, loss on drying, residue on ignition, and identity.
    (c) The batch for oxytetracycline content, polymyxin B content, 
sterility, moisture, and metal particles.
    (ii) Samples required:
    (a) The oxytetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch:
    (1) For all tests except sterility: A minimum of 16 immediate 
containers.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Oxytetracycline 
content. Proceed as directed in Sec. 436.106 of this chapter, preparing 
the sample for assay as follows: Place an accurately weighed 
representative portion of the sample into a separatory funnel containing 
approximately 50 milliliters of peroxide-free ether. Shake the ointment 
and ether until homogeneous. Add 20 to 25 milliliters of 0.1N 
hydrochloric acid and shake well. Allow the layers to separate. Remove 
the acid layer and repeat the extraction procedure with each of three 
more 20- to 25-milliliter quantities of 0.1N hydrochloric acid. Combine 
the acid extractives in a suitable volumetric flask and dilute to volume 
with 0.1N hydrochloric acid to obtain a stock solution of convenient 
concentration containing not less than 150 micrograms of oxytetracycline 
per milliliter (estimated). Further dilute an aliquot of the stock 
solution with sterile distilled water to the reference

[[Page 816]]

concentration of 0.24 microgram of oxytetracycline per milliliter 
(estimated).
    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, preparing the sample for assay as follows: Weigh 
accurately 0.5 to 1 gram of the ointment and place into a 15-milliliter 
centrifuge tube. Add 10 milliliters of peroxide-free ether. Stir until 
contents are homogeneous and centrifuge for 10 minutes at 3,000 
revolutions per minute. Decant the supernatant ether. Repeat washing and 
centrifugation steps once more. Add 10 milliliters of acetone, stir 
until contents are homogeneous, and centrifuge for 10 minutes at 3,000 
revolutions per minute. Decant the supernatant acetone. Repeat acetone 
wash and centrifugation once more. Continue acetone washings until the 
yellow color in the residue disappears. Add 3 to 4 drops of polysorbate 
80 to the residue and mix well. Gently wash the residue into a 100-
milliliter volumetric flask with 10 percent potassium phosphate buffer, 
pH 6.0 (solution 6), and further dilute with solution 6 to the reference 
concentration of 10 units of polymyxin B per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) Metal particles. Proceed as directed in Sec. 436.206 of this 
chapter.
[43 FR 11170, Mar. 17, 1978; 43 FR 34457, Aug. 4, 1978, as amended at 50 
FR 19920, May 13, 1985]



Sec. 446.381  Tetracycline hydrochloride ophthalmic dosage forms.



Sec. 446.381a  Tetracycline hydrochloride ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline hydrochloride ophthalmic 
ointment contains tetracycline hydrochloride in a suitable and harmless 
ointment base. Each gram contains 10 milligrams of tetracycline 
hydrochloride. Its potency is satisfactory if it contains not less than 
90 percent and not more than 125 percent of the number of milligrams of 
tetracycline hydrochloride that it is represented to contain. It is 
sterile. Its moisture content is not more than 0.5 percent. It passes 
the test for metal particles. The tetracycline hydrochloride used 
conforms to the standards prescribed by Sec. 446.81a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, crystallinity, and identity.
    (b) The batch for potency, sterility, moisture, and metal particles.
    (ii) Samples required:
    (a) The tetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the sample into a 
separatory funnel containing approximately 50 milliliters of peroxide-
free ether. Shake the sample and ether until homogeneous. Add 20 to 25 
milliliters of 0.1N hydrochloric acid and shake well. Allow the layers 
to separate. Remove the acid layer and repeat the extraction procedure 
with each of 3 more 20- to 25-milliliter quantities of 0.1N hydrochloric 
acid. Combine the extractives in a suitable volumetric flask and fill to 
volume with 0.1N hydrochloric acid to obtain a stock solution of 
convenient concentration containing not less than 150 micrograms of 
tetracycline hydrochloride per milliliter (estimated). Further dilute an 
aliquot with sterile distilled water to the reference concentration of 
0.24 micrograms of tetracycline hydrochloride per milliliter 
(estimated).

[[Page 817]]

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) Metal particles. Proceed as directed in Sec. 436.206 of this 
chapter.
[43 FR 11170, Mar. 17, 1978, as amended at 50 FR 19920, May 13, 1985]



Sec. 446.381b  Tetracycline hydrochloride ophthalmic suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline hydrochloride ophthalmic 
suspension contains tetracycline hydrochloride in a suitable and 
harmless oily base. Each milliliter contains 10 milligrams of 
tetracycline hydrochloride. Its potency is satisfactory if it contains 
not less than 90 percent and not more than 125 percent of the number of 
milligrams of tetracycline hydrochloride that it is represented to 
contain. It is sterile. Its moisture content is not more than 0.5 
percent. The tetracycline hydrochloride used conforms to the standards 
prescribed by Sec. 446.81a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, crystallinity, and identity.
    (b) The batch for potency, sterility, and moisture.
    (ii) Samples required:
    (a) The tetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of five immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place an accurately measured representative portion of the sample into a 
high-speed glass blender jar with 1 milliliter polysorbate 80 and 
sufficient 0.1N hydrochloric acid to give a stock solution of convenient 
concentration containing not less that 150 micrograms of tetracycline 
hydrochloride per milliliter (estimated). Blend for 3 to 5 minutes. 
Remove an aliquot and further dilute with sterile distilled water to the 
reference concentration of 0.24 microgram of tetracycline hydrochloride 
per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[43 FR 11171, Mar. 17, 1978, as amended at 46 FR 46313, Sept. 18, 1981; 
50 FR 19920, May 13, 1985]



                      Subpart E--Otic Dosage Forms



Sec. 446.467  Oxytetracycline hydrochloride-polymyxin B sulfate otic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline hydrochloride-polymyxin B 
sulfate otic ointment is oxytetracycline hydrochloride and polymyxin B 
sulfate in a suitable and harmless ointment base. Each gram of ointment 
contains oxytetracycline hydrochloride equivalent to 5 milligrams of 
oxytetracycline and polymyxin B sulfate equivalent to 10,000 units of 
polymyxin B. Its oxytetracycline hydrochloride content is satisfactory 
if it contains not less than 90 percent and not more than 120 percent of 
the number of milligrams of oxytetracycline that it is represented to 
contain. Its polymyxin B sulfate content is satisfactory if it contains 
not less than 90 percent and not more than 125 percent of the number of 
units of polymyxin B that it is represented to contain. Its moisture 
content is not more than 1 percent. The oxytetracycline hydrochloride 
used conforms to the standards prescribed by Sec. 446.67(a)(1). The 
polymyxin B sulfate used conforms to the standards prescribed by 
Sec. 448.30(a)(1) of this chapter.

[[Page 818]]

    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oxytetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, identity, and crystallinity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
pH, loss on drying, residue on ignition, and identity.
    (c) The batch for oxytetracycline content, polymyxin B content, and 
moisture.
    (ii) Samples required:
    (a) The oxytetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Oxytetracycline 
content. Proceed as directed in Sec. 436.106 of this chapter, preparing 
the sample for assay as follows: Place an accurately weighed 
representative portion of the sample into a separatory funnel containing 
approximately 50 milliliters of peroxide-free ether. Shake the sample 
and ether until homogeneous. Add 20 to 25 milliliters of 0.1N 
hydrochloric acid and shake well. Allow the layers to separate. Remove 
the acid layer and repeat the extraction procedure with each of three 
more 20- to 25-milliliter quantities of 0.1N hydrochloric acid. Combine 
the acid extractives in a suitable volumetric flask and fill to volume 
with 0.1N hydrochloric acid to obtain a stock solution of convenient 
concentration containing not less than 150 micrograms of oxytetracycline 
per milliliter (estimated). Further dilute an aliquot of the stock 
solution with sterile distilled water to the reference concentration of 
0.24 microgram of oxytetracycline per milliliter (estimated).
    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, preparing the sample for assay as follows: Weigh 
accurately 0.5 to 1.0 gram of the ointment and place into a 15-
milliliter centrifuge tube. Add 10 milliliters of peroxide-free ether. 
Stir until contents are homogeneous and centrifuge for 10 minutes at 
3,000 revolutions per minute. Decant the supernatant ether. Repeat 
washing and centrifugation steps once more. Add 10 milliliters of 
acetone, stir until contents are homogeneous, and centrifuge for 10 
minutes at 3,000 revolutions per minute. Decant the supernatant acetone. 
Repeat acetone wash and centrifugation once more. Continue acetone 
washings until the yellow color in the residue disappears. Add 3 to 4 
drops of polysorbate 80 to the residue and mix well. Gently wash the 
residue into a 100-milliliter volumetric flask with 10 percent potassium 
phosphate buffer, pH 6.0 (solution 6), and further dilute with solution 
6 to the reference concentration of 10 units of polymyxin B per 
milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[43 FR 11171, Mar. 17, 1978, as amended at 50 FR 19920, May 13, 1985]



                  Subpart F--Dermatologic Dosage Forms



Sec. 446.510  Chlortetracycline hydrochloride ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality and purity. Chlortetracycline hydrochloride ointment 
contains chlortetracycline hydrochloride and one or more suitable and 
harmless preservatives in a suitable and harmless ointment base. Each 
gram contains 30 milligrams of chlortetracycline hydrochloride. Its 
potency is satisfactory if it is not less than 90 percent and not more 
than 125 percent of the number of milligrams of chlortetracycline 
hydrochloride that it is represented to contain. Its moisture content is 
not more than 0.5 percent. The chlortetracycline hydrochloride used 
conforms to the standards prescribed by Sec. 446.10(a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5(a)(3) of this chapter, each package shall bear on its label 
or labeling, as hereinafter indicated, the following:

[[Page 819]]

    (i) On the label of the immediate container and on the outside 
wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The chlortetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, crystallinity, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The chlortetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 60 milligrams.
    (b) The batch: A minimum of five immediate containers:
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the sample into a 
separatory funnel containing approximately 50 milliliters of peroxide-
free ether. Shake the sample and ether until homogeneous. Add 20 to 25 
milliliters of 0.01N hydrochloric acid and shake well. Allow the layers 
to separate. Remove the acid layer and repeat the extraction procedure 
with each of three more 20- to 25-milliliter quantities of 0.01N 
hydrochloric acid. Combine the extractives in a suitable volumetric 
flask and dilute to volume with 0.01N hydrochloric acid. Further dilute 
an aliquot with sterile distilled water to the reference concentration 
of 0.06 microgram of chlortetracycline hydrochloride per milliliter 
(estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[43 FR 11172, Mar. 17, 1978, as amended at 50 FR 19920, May 13, 1985]



Sec. 446.542  Meclocycline sulfosalicylate cream.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Meclocycline sulfosalicylate cream 
contains meclocycline sulfosalicylate in a suitable and harmless cream 
base. Each gram contains meclocycline sulfosalicylate equivalent to 10 
milligrams of meclocycline. Its potency is satisfactory if it is not 
less than 90 percent and not more than 125 percent of the number of 
milligrams of meclocycline that it is represented to contain. The 
meclocycline sulfosalicylate used conforms to the standards prescribed 
by Sec. 446.42(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The meclocycline sulfosalicylate used in making the batch for 
potency, moisture, pH, and crystallinity.
    (b) The batch for potency.
    (ii) Samples required:
    (a) The meclocycline sulfosalicylate used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay; potency. Use either of the following 
methods; however, the results obtained from the high-pressure liquid 
chromatography method shall be conclusive.
    (1) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the sample into a 
high-speed glass blender jar containing sufficient 0.01N methanolic 
hydrochloric acid (solution 13) to obtain a stock solution of convenient 
concentration. Blend for 3 to 5 minutes. Further dilute an aliquot of 
the stock solution with distilled water to the reference concentration 
of 0.06 microgram of meclocycline per milliliter (estimated).
    (2) High-pressure liquid chromatography. Proceed as directed in

[[Page 820]]

Sec. 436.329 of this chapter, except, prepare the working standard and 
sample solution and calculate the meclocycline content as follows:
    (i) Preparation of standard solution. Accurately weigh an amount of 
working standard equivalent to approximately 25 milligrams of 
meclocycline into a 50-milliter volumetric flask. Dissolve and dilute to 
volume with methanol and mix. Transfer exactly 2.0 milliliters of this 
solution to a 100-milliliter volumetric flask, dilute to volume with 
mobile phase, and mix.
    (ii) Preparation of sample solution. Accurately weigh approximately 
0.4 to 0.7 gram of sample into a 50-milliliter glass-stoppered 
centrifuge tube. Add 20 milliliters of methanol and 20 milliliters of 
0.025N sulfuric acid. Disperse the sample thoroughly by a combination of 
ultrasonic/vortexing and shaking by hand. Shake for 15 minutes on a 
wrist action shaker. Quantitatively transfer the contents of the 
centrifuge tube into a 50-milliliter volumetric flask. Rinse the 
centrifuge tube with two 5-milliliter portions of methanol and add to 
the flask. Dilute to volume with methanol and mix. Transfer a portion of 
the content of the volumetric flask into an appropriate-sized centrifuge 
tube. Centrifuge for 5 minutes at 2,000 revolutions per minute. Transfer 
5.0 milliliters of this solution into a 50-milliliter volumetric flask 
and dilute to volume with mobile phase and mix. Filter this solution 
through a 0.5 micrometer filter. Inject the filtrate onto the column as 
described in Sec. 436.329(e) of this chapter.
    (iii) Calculations. Calculate the meclocycline content as follows:

    [GRAPHIC] [TIFF OMITTED] TR01JA93.261
    

where:
A=Area or peak height of the sample peak (at a retention time equal to 
          that observed for the standard);
B=Area or peak height of the standard peak.
[46 FR 3837, Jan. 16, 1981; 46 FR 21361, Apr. 10, 1981, as amended at 47 
FR 22515, May 25, 1982; 50 FR 1504, Jan. 11, 1985]



Sec. 446.567  Oxytetracycline hydrochloride dermatologic dosage forms.



Sec. 446.567a  [Reserved]



Sec. 446.567b  Oxytetracycline hydrochloride-polymyxin B sulfate topical ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline hydrochloride-polymyxin B 
sulfate topical ointment is oxytetracycline hydrochloride and polymyxin 
B sulfate in a suitable and harmless ointment base. Each gram contains 
oxytetracycline hydrochloride equivalent to 30 milligrams of 
oxytetracycline and polymyxin B sulfate equivalent to 10,000 units of 
polymyxin B. Its oxytetracycline content is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of oxytetracycline that it is represented to contain. Its 
polymyxin B sulfate content is satisfactory if it is not less than 90 
percent and not more than 125 percent of the number of units of 
polymyxin B that it is represented to contain. Its moisture content is 
not more than 1 percent. The oxytetracycline hydrochloride used conforms 
to the standards prescribed by Sec. 446.67(a)(1). The polymyxin B 
sulfate conforms to the standards prescribed by Sec. 448.30(a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5(a)(3) of this chapter, each package shall bear on its label 
or labeling as hereinafter indicated, the following:
    (i) On the label of the immediate container and on the outside 
wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.

[[Page 821]]

    (ii) On the label of the immediate container or other labeling 
attached to or within the package: Adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The oxytetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, identity, and crystallinity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, residue on ignition, and identity.
    (c) The batch for oxytetracycline content, polymyxin B content, and 
moisture.
    (ii) Samples required:
    (a) The oxytetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Oxytetracycline 
content. Proceed as directed in Sec. 436.106 of this chapter, preparing 
the sample for assay as follows: Place an accurately weighed 
representative portion of the sample into a separatory funnel containing 
approximately 50 milliliters of peroxide-free ether. Shake the ointment 
and ether until homogeneous. Add 20 to 25 milliliters of 0.1N 
hydrochloric acid and shake well. Allow the layers to separate. Remove 
the acid layer and repeat the extraction procedure with each of three 
more 20- to 25-milliliter quantities of 0.1N hydrochloric acid. Combine 
the acid extractives in a suitable volumetric flask and dilute to volume 
with 0.1N hydrochloric acid to obtain a stock solution of convenient 
concentration containing not less than 150 micrograms of oxytetracycline 
per milliliter (estimated). Further dilute an aliquot of the stock 
solution with sterile distilled water to the reference concentration of 
0.24 microgram of oxytetracycline per milliliter (estimated).
    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, preparing the sample for assay as follows: Weigh 
accurately 0.5 to 1 gram of the ointment and place into a 15-milliliter 
centrifuge tube. Add 10 milliliters of peroxide-free ether. Stir until 
contents are homogeneous and centrifuge for 10 minutes at 3,000 
revolutions per minute. Decant the supernatant ether. Repeat washing and 
centrifugation steps once more. Add 10 milliliters of acetone, stir 
until contents are homogeneous, and centrifuge for 10 minutes at 3,000 
revolutions per minute. Decant the supernatant acetone. Repeat acetone 
wash and centrifugation once more. Continue acetone washing until the 
yellow color in the residue disappears. Add 3 to 4 drops of polysorbate 
80 to the residue and mix well. Gently wash the residue into a 100-
milliliter volumetric flask with 10 percent potassium phosphate buffer, 
pH 6.0 (solution 6), and further dilute with solution 6 to the reference 
concentration of 10 units of polymyxin B per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[43 FR 11172, Mar. 17, 1978, as amended at 50 FR 19920, May 13, 1985]



Sec. 446.567c  Oxytetracycline hydrochloride-polymyxin B sulfate topical powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline hydrochloride-polymyxin B 
sulfate topical powder is oxytetracycline hydrochloride and polymyxin B 
sulfate with a suitable filler. Each gram contains 30 milligrams of 
oxytetracycline and 10,000 units of polymyxin B. Its oxytetracycline 
content is satisfactory if it is not less than 90 percent and not more 
than 120 percent of the number of milligrams of oxytetracycline that it 
is represented to contain. Its polymyxin B content is satisfactory if it 
is not less than 90 percent and not more than 120 percent of the number 
of units of polymyxin B that it is represented to contain. The loss on 
drying is not more than 2.0 percent. The oxytetracycline hydrochloride 
used conforms to the

[[Page 822]]

standards prescribed by Sec. 446.67. The polymyxin B sulfate used 
conforms to the standards prescribed by Sec. 448.30(a)(1) of this 
chapter.
    (2) Labeling. Each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On the label of the immediate container and on the outside 
wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (c) An expiration date that is 12 months after the month during 
which the batch was certified, unless the use of a longer expiration 
period has been approved in accordance with the provisions of 
Sec. 432.5(a)(3) of this chapter.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions for lay use of 
the drug.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The oxytetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, identity, and crystallinity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, residue on ignition, and identity.
    (c) The batch for oxytetracycline content, polymyxin content, and 
loss on drying.
    (ii) Samples required:
    (a) The oxytetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Oxytetracycline 
content. Proceed as directed in Sec. 436.106 of this chapter, preparing 
the sample for assay as follows: Place an accurately weighed 
representative portion of the sample into a high-speed glass blender jar 
with sufficient 0.1N hydrochloric acid to obtain a stock solution of 
convenient concentration containing not less than 150 micrograms of 
oxytetracycline hydrochloride per milliliter (estimated). Blend for 3 to 
5 minutes. Remove an aliquot of the stock solution and further dilute 
with sterile distilled water to the reference concentration of 0.24 
microgram of oxytetracycline per milliliter (estimated).
    (ii) Polymyxin content. Proceed as directed in Sec. 436.105 of this 
chapter, preparing the sample for assay as follows: Accurately weigh 1 
gram of the powder and place into a 50-milliliter centrifuge tube. Add 
15 milliliters of acetone and 1 drop of concentrated hydrochloric acid 
and stir well. Add 20 milliliters of acetone and centrifuge for 10 
minutes at 3,000 revolutions per minute. Decant the supernatant liquid 
and repeat the acetone-acid extraction once more. Dissolve and dilute 
the residue with sufficient 10 percent potassium phosphate buffer, pH 
6.0 (solution 6), to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of the stock solution with 
solution 6 to the reference concentration of 10 units of polymyxin B per 
milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[43 FR 11173, Mar. 17, 1978, as amended at 50 FR 19920, May 13, 1985]



Sec. 446.581  Tetracycline hydrochloride dermatologic dosage forms.



Secs. 446.581a--446.581b  [Reserved]



Sec. 446.581c  Tetracycline hydrochloride for topical solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline hydrochloride for topical 
solution is a dry mixture of tetracycline hydrochloride, 4-
epitetracycline hydrochloride, and sodium bisulfite packaged in 
combination with a suitable and harmless aqueous vehicle. When 
reconstituted as directed in the labeling, each milliliter contains 2.2 
miligrams of tetracycline hydrochloride. The tetracycline hydrochloride 
content of the reconstituted solution is satisfactory if it contains not 
less than 90 percent and not more

[[Page 823]]

than 130 percent of the number of milligrams of tetracycline 
hydrochloride per milliliter that it is represented to contain. The 4-
epitetracycline hydrochloride content is satisfactory if it contains not 
less than 135 percent and not more than 165 percent of the amount of 
tetracycline hydrochloride in the reconstituted solution at the time of 
reconstitution. The loss on drying of the dry mixture is not more than 
5.0 percent. When reconstituted as directed in the labeling, its pH is 
not less than 1.9 and not more than 3.5. The tetracycline hydrochloride 
used conforms to the standards prescribed by Sec. 446.81a, except 
sterility, pyrogens, and histamine. The 4-epitetracycline hydrochloride 
used conforms to the following standards: It gives a positive identity 
test for 4-epitetracycline hydrochloride; its 4-epitetracycline content 
is not less than 70 percent; its total anhydrotetracycline and 4-
epianhydrotetracycline content is not more than 2.0 percent; its loss on 
drying is not more than 6.0 percent; its pH in an aqueous solution 
containing 10 milligrams per milliliter is not less than 2.3 and not 
more than 4.0.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, and crystallinity.
    (b) The 4-epitetracycline hydrochloride used in making the batch for 
4-epitetracycline content and identity, total anhydrotetracycline and 4-
epianhydrotetracycline content, loss on drying, and pH.
    (c) The batch for tetracycline hydrochloride content, 4-
epitetracycline hydrochloride content, loss on drying, and pH.
    (ii) Samples required:
    (a) The tetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay of the tetracycline hydrochloride for 
topical solution--(1) Tetracycline hydrochloride content and 4-
epitetracycline hydrochloride content. Proceed as directed in 
Sec. 436.340 of this chapter.
    (2) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter, except use the contents of one immediate container.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution obtained when reconstituted as directed in the labeling.
    (c) Tests and methods of assay of the 4-epitetracycline 
hydrochloride used in making the batch--(1) 4-epitetracycline content 
and identity. Proceed as directed in paragraph (b)(1) of this section, 
except in lieu of Sec. 446.581c(b)(1)(iv) prepare the sample by weighing 
accurately 20 milligrams plus-minus5 milligrams of 4-
epitetracycline hydrochloride bulk powder and transfer to a 25-
milliliter volumetric flask. Dissolve with 1.0 milliliter of methyl 
alcohol and dilute to volume with the buffer solution. Pipet a 2.0-
milliliter aliquot to a 10-milliliter volumetric flask and dilute to 
volume with the buffer solution. Place the column in a suitable support. 
Place a 100-milliliter graduate under the column. Open the column 
stopcock, pipet 2.0 milliliters of solution from the 10-milliliter 
volumetric flask onto the column packing and allow the sample to 
permeate the column packing. Place a solvent reservoir containing 20 
milliliters of benzene on top of the column and begin to collect the 
eluate (at flow rate of approximately 1 milliliter per minute). When the 
benzene level reaches the top of the column packing, replace the empty 
solvent reservoir with a second solvent reservoir containing 60 
milliliters of chloroform and continue elution. When the chloroform 
level reaches the top of the column packing, replace second empty 
solvent reservoir with a third solvent reservoir containing 50-
milliliters of the n-butanol:chloroform mixture and replace the 100-
milliliter graduate with a 10-milliliter graduate. Collect 8.0 
milliliters of eluate. Replace the 10-milliliter graduate with a 50-
milliliter low-actinic volumetric flask and continue collecting the 
eluate containing the 4-

[[Page 824]]

epitetracycline fraction until the column runs dry. Determine the 
absorbance of the 4-epitetracycline eluate as described in paragraph 
(b)(1)(v) of this section.
    Calculate the 4-epitetracycline hydrochloride content of the 4-
epitetracycline hydrochloride bulk powder as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.262

where:
A=Absorbance at 366 nanometers of the low-actinic 50-milliliter 
volumetric flask.
a=Previously established mean absorptivity of the tetracycline 
hydrochloride working standard eluates in liters/gram/centimeter with 
the calculation corrected for potency.
W=Weight of 4-epitetracycline hydrochloride bulk powder in milligrams.


The identity of the 4-epitetracycline hydrochloride is confirmed if the 
absorbance of the sample after column elution is such that the 4-
epitetracycline hydrochloride content is greater than 70 percent by 
weight.
    (2) Total anhydrotetracycline and 4-epianhydrotetracycline content. 
Proceed as directed in Sec. 436.309 of this chapter.
    (3) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 10 milligrams per milliliter.
[42 FR 59066, Nov. 15, 1977; 43 FR 3705, Jan. 27, 1978, as amended at 48 
FR 51291, Nov. 8, 1983; 50 FR 19920, May 13, 1985]



Sec. 446.581d  Tetracycline hydrochloride ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline hydrochloride ointment 
contains tetracycline hydrochloride in a suitable and harmless ointment 
base. Each gram contains 30 milligrams of tetracycline hydrochloride. 
Its potency is satisfactory if it contains not less than 90 percent and 
not more than 125 percent of the number of milligrams of tetracycline 
hydrochloride that it is represented to contain. Its moisture content is 
not more than 1 percent. The tetracycline hydrochloride used conforms to 
the standards prescribed by Sec. 446.81(a)(1), except 4-
epianhydrotetracycline content.
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5(a)(3) of this chapter, each package shall bear on its label 
or labeling as hereinafter indicated, the following:
    (i) On the label of the immediate container and on the outside 
wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (ii) On the label of the immediate container or other labeling 
attached to or inserted within the package: Adequate directions under 
which the layperson can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, crystallinity, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The tetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the sample into a 
separatory funnel containing approximately 50 milliliters of peroxide-
free ether. Shake the sample and ether until homogeneous. Add 20 to 25 
milliliters of 0.1N hydrochloric acid and shake well. Allow the layers 
to separate. Remove the acid layer and repeat the extraction procedure 
with each of three more 20- to 25-milliliter quantities of 0.1N 
hydrochloric acid. Combine the acid extractives in a suitable volumetric 
flask and fill to volume with 0.1N hydrochloric acid to obtain a stock 
solution of convenient concentration containing not less than

[[Page 825]]

150 micrograms of tetracycline hydrochloride per milliliter (estimated). 
Further dilute an aliquot of the stock solution with sterile distilled 
water to the reference concentration of 0.24 microgram of tetracycline 
hydrochloride per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[43 FR 11174, Mar. 17, 1978 as amended at 44 FR 30333, May 25, 1979. 
Redesignated at 45 FR 16472, Mar. 14, 1980, and amended at 50 FR 19920, 
May 13, 1985]



                     Subpart G--Vaginal Dosage Forms



Sec. 446.667  Oxytetracycline hydrochloride-polymyxin B sulfate vaginal tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oxytetracycline hydrochloride-polymyxin B 
sulfate vaginal tablets are tablets composed of oxytetracycline 
hydrochloride and polymyxin B sulfate with one or more suitable 
diluents, binders, lubricants, and preservatives. Each tablet contains 
oxytetracycline hydrochloride equivalent to 100 milligrams of 
oxytetracycline and polymyxin B sulfate equivalent to 100,000 units of 
polymyxin B. Its oxytetracycline content is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of oxytetracycline that it is represented to contain. Its 
polymyxin B content is satisfactory if it is not less than 90 percent 
and not more than 120 percent of the number of units of polymyxin B that 
it is represented to contain. The loss on drying is not more than 3.0 
percent. The oxytetracycline hydrochloride used conforms to the 
standards prescribed by Sec. 446.67(a)(1). The polymyxin B sulfate used 
conforms to the standards prescribed by Sec. 448.30(a)(1) of this 
chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oxytetracycline hydrochloride used in making the batch for 
potency, loss on drying, pH, absorptivity, identity, and crystallinity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, residue on ignition, and identity.
    (c) The batch for oxytetracycline content, polymyxin B content, and 
loss on drying.
    (ii) Samples required:
    (a) The oxytetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (c) The batch: A minimum of 30 tablets.
    (b) Tests and methods of assay--(1) Potency--(i) Oxytetracycline 
content. Proceed as directed in Sec. 436.106 of this chapter, preparing 
the sample for assay as follows: Place a representative number of 
tablets into a high-speed glass blender jar containing sufficient 0.1N 
hydrochloric acid to obtain a stock solution of convenient concentration 
containing not less than 150 micrograms of oxytetracycline per 
milliliter (estimated). Blend for 3 to 5 minutes. Remove an aliquot of 
the stock solution and further dilute with sterile distilled water to 
the reference concentration of 0.24 microgram of oxytetracycline per 
milliliter (estimated).
    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, preparing the sample for assay as follows: Grind a 
representative number of tablets into a fine powder and place this 
powder, accurately weighed, into a filter funnel with a solvent-
resistant membrane filter of 1.0 micrometer porosity. Wash the powder 
with five 20-milliliter portions of acetone or until the yellow color 
has disappeared. Remove the filter and soak in 400 milliliters of 10 
percent potassium phosphate buffer, pH 6.0 (solution 6), and blend. 
Quantitatively transfer to a 500-milliliter volumetric flask and adjust 
to volume with solution 6. Further dilute an aliquot with solution 6 to 
the reference concentration of 10 units of polymyxin B per milliliter 
(estimated).

[[Page 826]]

    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[43 FR 11174, Mar. 17, 1978, as amended at 50 FR 19920, May 13, 1985]



               Subpart H--Rectal Dosage Forms  [Reserved]



                          Subpart I  [Reserved]



            Subpart J--Certain Other Dosage Forms  [Reserved]



PART 448--PEPTIDE ANTIBIOTIC DRUGS--Table of Contents




                          Subpart A--Bulk Drugs

Sec.
448.10  Bacitracin.
448.10a  Sterile bacitracin.
448.13  Bacitracin zinc.
448.13a  Sterile bacitracin zinc.
448.15a  Sterile capreomycin sulfate.
448.20a  Sterile colistimethate sodium.
448.21  Colistin sulfate.
448.23  Cyclosporine.
448.25  Gramicidin.
448.30  Polymyxin B sulfate.
448.30a  Sterile polymyxin B sulfate.
448.75  Tyrothricin.

                      Subpart B--Oral Dosage Forms

448.121  Colistin sulfate for oral suspension.
448.123  Cyclosporine oral dosage forms.
448.123a  Cyclosporine oral solution.
448.123b  Cyclosporine capsules.

                   Subpart C--Injectable Dosage Forms

448.210  Sterile bacitracin.
448.215  Sterile capreomycin sulfate.
448.220  Colistimethate sodium injectable dosage forms.
448.220a  Sterile colistimethate sodium.
448.223  Cyclosporine for infusion.
448.230  Sterile polymyxin B sulfate.

                   Subpart D--Ophthalmic Dosage Forms

448.310  Bacitracin ophthalmic dosage forms.
448.310a  [Reserved]
448.310b  Bacitracin-neomycin sulfate-polymyxin B sulfate ophthalmic 
          ointment.
448.310c  Bacitracin ophthalmic ointment.
448.313  Bacitracin zinc ophthalmic dosage forms.
448.313a  Bacitracin zinc-polymyxin B sulfate ophthalmic ointment.
448.313b  Bacitracin zinc-neomycin sulfate-polymyxin B sulfate 
          ophthalmic ointment; bacitracin zinc-neomycin sulfate-
          polymyxin B sulfate hydrocortisone ophthalmic ointment.
448.321  Colistin sulfate for ophthalmic solution.
448.330  Polymyxin B sulfate-trimethoprim hemisulfate ophthalmic 
          solution.

                      Subpart E--Otic Dosage Forms

448.421  Colistin sulfate-neomycin sulfate-thonzonium bromide-
          hydrocortisone acetate otic suspension.
448.430  Polymyxin B sulfate-hydrocortisone otic solution.

                  Subpart F--Dermatologic Dosage Forms

448.510  Bacitracin dermatologic dosage forms.
448.510a  Bacitracin ointment.
448.510b--448.510c  [Reserved]
448.510d  Bacitracin-neomycin sulfate ointment.
448.510e  Bacitracin-neomycin sulfate-polymyxin B sulfate ointment.
448.510f  Bacitracin-polymyxin B sulfate topical aerosol.
448.513  Bacitracin zinc dermatologic dosage forms.
448.513a  Bacitracin zinc-polymyxin B sulfate ointment.
448.513b  Bacitracin zinc-neomycin sulfate ointment.
448.513c  Bacitracin zinc-neomycin sulfate-polymyxin B sulfate ointment; 
          bacitracin zinc-neomycin sulfate-polymyxin B sulfate 
          hydrocortisone ointment.
448.513d  Bacitracin zinc-polymyxin B sulfate topical powder.
448.513e  Bacitracin zinc-polymyxin B sulfate topical aerosol.
448.513f  Bacitracin zinc ointment.

               Subpart G--Vaginal Dosage Forms  [Reserved]

                        Subparts H-I  [Reserved]

                  Subpart J--Certain Other Dosage Forms

448.910  Bacitracin for prescription compounding.
448.913  Bacitracin zinc for prescription compounding.
448.930  Polymyxin B sulfate in certain other dosage forms.
448.930a  Polymyxin B sulfate for prescription compounding.
448.930b  Sterile polymyxin B sulfate-benzalkonium chloride urethral 
          lubricant.

    Authority:  21 U.S.C. 357.

    Source:  39 FR 19115, May 30, 1974, unless otherwise noted.

[[Page 827]]



                          Subpart A--Bulk Drugs



Sec. 448.10  Bacitracin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin is a white to brown, neutral 
water-soluble polypeptide. It is so purified and dried that:
    (i) Its potency is not less than 40 units of bacitracin per 
milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 5 percent.
    (iv) Its pH in an aqueous solution containing 10,000 units per 
milliliter is not less than 5.5 and not more than 7.5.
    (v) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, and identity.
    (ii) Samples required: 10 packages, each containing approximately 
1.0 gram.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed for 
bacitracin zinc in Sec. 436.105 of this chapter, preparing the sample 
for assay as follows: Dissolve an accurately weighed sample in 
sufficient 1.0 percent potassium phosphate buffer, pH 6.0 (solution 1), 
to obtain a stock solution of convenient concentration. Remove an 
aliquot of the stock solution, add sufficient hydrochloric acid so that 
the amount of acid in the final solution will be the same as in the 
reference concentration of the working standard and further dilute with 
solution 1 to the reference concentration of 1.0 unit of bacitracin per 
milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10,000 units per milliliter.
    (5) Identity. Proceed as directed in Sec. 436.319 of this chapter.
[42 FR 27229, May 27, 1977, as amended at 50 FR 19920, May 13, 1985]



Sec. 448.10a  Sterile bacitracin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin is a white to brown, neutral 
water-soluble polypeptide. It is so purified and dried that:
    (i) Its potency is not less than 50 units per milligram. If it is 
packaged for dispensing, its content is satisfactory if it is not less 
than 90 percent and not more than 115 percent of the number of units of 
bacitracin that it is represented to contain.
    (ii) It is sterile.
    (iii) [Reserved]
    (iv) It is nonpyrogenic.
    (v) Its loss on drying is not more than 5 percent.
    (vi) Its pH in an aqueous solution containing 10,000 units per 
milliliter is not less than 5.5 and not more than 7.5.
    (vii) Its residue on ignition is not more than 3.0 percent.
    (viii) It passes the identity test.
    (ix) Its heavy metals content is not more than 30 parts per million.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, pH, residue on ignition, identity, and heavy 
metals.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 1 gram.
    (2) For sterility testing: 1 package, containing approximately 12 
grams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests ands methods of assay--(1) Potency. Proceed as directed 
for bacitracin zinc in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Dissolve an accurately weighed

[[Page 828]]

sample in sufficient 1.0 percent potassium phosphate buffer, pH 6.0 
(solution 1), to obtain a stock solution of convenient concentration; 
also, if it is packaged for dispensing, reconstitute as directed in the 
labeling. Then using a suitable hypodermic needle and syringe, remove 
all of the withdrawable contents if it is represented as a single dose 
container, or if the labeling specifies the amount of potency in a given 
volume of the resultant preparation, remove an accurately measured 
representative portion from each container. Dilute with solution 1 to 
obtain a stock solution of convenient concentration. Remove an aliquot 
of the stock solution, add sufficient hydrochloric acid so that the 
amount of acid in the final solution will be the same as in the 
reference concentration of the working standard and further dilute with 
solution 1 to the reference concentration of 1.0 unit of bacitracin per 
milliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 300 units of bacitracin per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10,000 units per milliliter.
    (7) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (8) Identity. Proceed as directed in Sec. 436.319 of this chapter.
    (9) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
[42 FR 27229, May 27, 1977, as amended at 50 FR 19920, May 13, 1985]



Sec. 448.13  Bacitracin zinc.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin zinc is the zinc salt of a 
kind of bacitracin or a mixture of two or more such salts. It is so 
purified and dried that:
    (i) Its potency is not less than 40 units per milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 5 percent.
    (iv) Its pH is not less than 6.0 and not more than 7.5.
    (v) Its zinc content is not more than 10 percent by weight on an 
anhydrous basis.
    (vi) It passes the identity test.
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, each package shall bear on the outside wrapper or 
container and the immediate container the statement ``For use only in 
the manufacture of nonparenteral drugs''.
    (3) Requests for certification; samples. In addition to complying 
with the requriements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, zinc content,and identity.
    (ii) Samples required: 10 packages, each containing approximately 
1.0 gram.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample (usually 25 to 35 milligrams) in 
sufficient 0.01N hydrochloric acid to give a bacitracin concentration of 
100 units per milliliter (estimated). Further dilute an aliquot with 
solution 1 to the reference concentration of 1.0 unit of bacitracin per 
milliliter (estimated).

    Note: The final sample solution must contain the same amount of 
hydrochloric acid as the reference concentration of the working 
standard.

    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
saturated solution (approximately 100 milligrams of the sample per 
milliliter).
    (5) Zinc content. Proceed as directed in Sec. 436.312 of this 
chapter.
    (6) Identity. Proceed as directed in Sec. 436.319 of this chapter.
[42 FR 27229, May 27, 1977, as amended at 50 FR 19920, May 13, 1985]



Sec. 448.13a  Sterile bacitracin zinc.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality,

[[Page 829]]

and purity. Sterile bacitracin zinc is the zinc salt of a kind of 
bacitracin or a mixture of two or more such salts. It is so purified and 
dried that:
    (i) It contains not less than 40 units of bacitracin per milligram.
    (ii) It is sterile.
    (iii) [Reserved]
    (iv) Its loss on drying is not more than 5.0 percent.
    (v) Its pH is not less than 6.0 and not more than 7.5.
    (vi) Its zinc content is not more than 10 percent by weight on a 
moisture-free basis.
    (vii) It passes the identity test.
    (2) Labeling. In addition to the labeling requirements of Sec. 432.5 
of this chapter, each package shall bear on the outside wrapper or 
container and the immediate container the statement ``For use in the 
manufacture of topical drugs only''.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
loss on drying, pH, zinc content, and identity.
    (ii) Samples required:
    (a) For all tests except sterility: Six packages, each containing 
approximately 1.0 gram.
    (b) For sterility testing: 20 packages, each containing 
approximately 500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed for 
bacitracin in Sec. 436.105 of this chapter, except add to each standard 
response line concentration sufficient 0.01N hydrochloric acid to yield 
the same ratio of 0.01N hydrochloric acid to 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1) as present in the sample solution 
diluted to the reference concentration. Prepare the sample for assay as 
follows: Dissolve an accurately weighed sample (usually 25 to 35 
milligrams) in sufficient 0.01N hydrochloric acid to give a bacitracin 
concentration of 100 units per milliliter (estimated). Further dilute 
with solution 1 to the reference concentration of 1.0 unit of bacitracin 
per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use diluting fluid F in lieu of diluting fluid A.
    (3) [Reserved]
    (4) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
saturated solution (approximately 100 milligrams of the sample per 
milliliter).
    (6) Zinc content. Proceed as directed in Sec. 436.312 of this 
chapter.
    (7) Identity. Proceed as directed in Sec. 436.319 of this chapter.
[39 FR 19115, May 30, 1974, as amended at 40 FR 15088, Apr. 4, 1975; 40 
FR 19194, May 2, 1975; 42 FR 27230, May 27, 1977; 50 FR 19920, May 13, 
1985]



Sec. 448.15a  Sterile capreomycin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile capreomycin sulfate is the 
amorphous sulfate salt of capreomycin. It is a white or essentially 
white powder. Capreomycin has been separated chromatographically into 
components designated capreomycins Ia, Ib, IIa, and IIb. Each component 
has been partially characterized according to its type and amino acid 
content. Capreomycin Ia contains serine and no alanine. Capreomycin Ib 
contains alanine and no serine. Capreomycin I is a mixture of 
capreomycins Ia and Ib. It is so purified and dried that:
    (i) Its potency is not less than 700 micrograms and not more than 
1,050 micrograms of capreomycin per milligram on an ``as is'' basis. If 
it is packaged for dispensing, its potency is satisfactory if it is not 
less than 90 percent and not more than 115 percent of the number of 
milligrams of capreomycin that it is represented to contain.
    (ii) It is sterile.
    (iii) [Reserved]
    (iv) It is nonpyrogenic.
    (v) It contains no depressor substance.
    (vi) Its loss on drying is not more than 10 percent.
    (vii) Its pH in an aqueous solution containing 30 milligrams per 
milliliter (or if packaged for dispensing, after reconstitution as 
directed in the labeling) is not less than 4.5 and not more than 7.5.

[[Page 830]]

    (viii) Its capreomycin I content is not less than 90 percent of the 
total capreomycins.
    (ix) Its residue on ignition is not more than 3 percent.
    (x) Its heavy metals content is not more than 30 parts per million.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, depressor substances, loss on drying, pH, capreomycin I 
content, residue on ignition, and heavy metals.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 500 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient sterile distilled 
water to give a stock solution of convenient concentration; also, if it 
is packaged for dispensing, reconstitute as directed in the labeling. 
Then using a suitable hypodermic needle and syringe, remove all of the 
withdrawable contents if it is represented as a single dose container; 
or if the labeling specifies the amount of potency in a given volume of 
the resultant preparation, remove an accurately measured representative 
portion from each container. Dilute with sterile distilled water to give 
a stock solution of convenient concentration. Further dilute the stock 
solution with sterile distilled water to the reference concentration of 
100 micrograms of capreomycin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) [Reserved]
    (4) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 10 milligrams per milliliter.
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) Loss on drying. Proceed as directed in Sec. 436.200(e) of this 
chapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 30 milligrams per milliliter; however, if 
it is packaged for dispensing, use the solution obtained after 
reconstituting the drug as directed in the labeling.
    (8) Capreomycin I content--(i) Equipment--(a) Sheet 
(chromatographic). Whatman No. 1 filter paper for chromatography 20  x  
50 centimeters.
    (b) Chamber (chromatographic). Square glass chromatography jar, 30 
x  30  x  60 centimeters, designed for descending chromatography. The 
bottom of the tank is filled with 1.5 inches of a mixture of 70 percent 
n- propyl alcohol and 30 percent distilled water (v/v) and allowed to 
equilibrate for 2 days. The mobility of the capreomycin factors, 
capreomycin I and capreomycin II, depends to a large extent upon the 
amount of water vapor present in the chromatographic chamber. The 
mobility can be restricted by using more n- propyl alcohol and less 
water in the equilibrating solvent, or it can be increased by raising 
the water content. The Rf of value (the distance traveled by 
a particular antibiotic factor divided by the distance traveled by the 
solvent front) should be approximately 0.50 for capreomycin I and 
approximately 0.60 for capreomycin II.
    (ii) Preparation of solutions--(a) 0.1N citrate buffer, pH 6.2. 
Dissolve 21.0 grams of citric acid monohydrate in 1 liter of distilled 
water. Adjust the pH to 6.2 with 50 percent aqueous sodium hydroxide.
    (b) Developing solvent. Mix n- propyl alcohol, distilled water, 
triethylamine, and glacial acetic acid in volumetric proportions of 
75:33:8:8, respectively.

[[Page 831]]

    (iii) Preparation of the capreomycin sample solution. Dissolve 
approximately 200 milligrams of the sample, accurately weighed, with 
distilled water in a 10-milliliter volumetric flask. Dilute to volume 
with distilled water. This sample should be refrigerated when not in 
use.
    (iv) Preparation of the chromatogram. Use separate sheets for each 
capreomycin sample solution and for blanks without sample application. 
Evenly apply a 100-microliter aliquot of the capreomycin sample solution 
to the origin line of a sheet. A U-shaped glass rod is placed under the 
chromatogram during spotting. Dry the streak thoroughly with warm air. 
Place the sample sheets and a blank sheet in the chamber and develop 
them in a descending manner for 16 hours. Remove the sheets from the 
chamber and air dry for about 1 hour.
    (v) Processing the chromatogram. Examine each sheet under short-
wave-length (254 nanometers) ultraviolet light and locate the main 
streak (Rf approximately 0.5) and the preceding streak 
(capreomycin II, Rf approximately 0.6). Outline the main zone 
lightly with a pencil. Outline an area on the blank sheet approximately 
equal in size and in the same location as those outlined on the sample 
sheets. Cut the marked areas from the sheets and then cut them into 
approximately 1.5-centimeter squares. For each sheet, place the squares 
into a glass-stoppered 50-milliliter Erlenmeyer flask.
    (vi) Elution. To each flask, add 10 milliliters of 0.1N citrate 
buffer, pH 6.2, and agitate on a reciprocating shaker for 1 hour. Filter 
each of the shaken solutions through Whatman No. 1 filter paper into 
separate 10-milliliter glass-stoppered Erlenmeyer flasks. Transfer 3 
milliliters of each filtrate into separate 50-milliliter volumetric 
flasks and dilute to volume with distilled water.
    (vii) Capreomycin sample solution for direct measurement of 
absorbance. Pipette 1.0 milliliter of the sample solution prepared as 
described in paragraph (b)(8)(iii) of this section into a 100-milliliter 
glass-stoppered volumetric flask. Dilute to volume with 0.1N citrate 
buffer, pH 6.2. Transfer 3.0 milliliters of this solution into a 50-
milliliter volumetric flask and dilute to volume with distilled water.
    (viii) Absorbance measurement. Using a suitable spectrophotometer, 
1.0-centimeter quartz cells, and distilled water as the reference 
solvent, determine the absorbance of each eluate and of each sample 
solution at the absorption maximum at about 268 nanometers.
    (ix) Calculation of percent capreomycin I in samples. Calculate as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.263

where:
AI=Absorbance of the eluate from the main zone of the sample 
sheet;
AB=Absorbance of the eluate from the area of the blank sheet 
corresponding to the area of the capreomycin I of the sample sheet;
As=Absorbance of the capreomycin sample solution described in 
paragraph (b)(8)(vii) of this section.


If the assay of capreomycin I from the chromatogram is less than 90 
percent of total capreomycins, repeat the procedure described in 
paragraph (b)(8) (iv), (v), (vi), (vii), and (viii) of this section two 
more times and at the same time determine the recovery of total 
capreomycins from the unchromatographed sheet as described in paragraph 
(b)(8)(x) of this section. The average of three valid assays should then 
be reported.
    (x) Recovery of total capreomycins from the unchromatographed 
sheet--(a) Procedure. Evenly apply a 100-microliter aliquot of the 
capreomycin sample solution (prepared as described in paragraph 
(b)(8)(iii) of this section) to the origin line of a sheet. Dry the 
streak thoroughly with warm air. The paper is not chromatographed before 
elution. Cut the area containing the streak from the sheet and then cut 
into approximately 1.5-centimeter squares. Place the squares into a 
glass-stoppered 50-milliliter Erlenmeyer flask and proceed as directed 
in paragraph (b)(8) (vi), (vii), and (viii) of this section. Likewise, 
cut an equal-sized area from an untreated part of the sheet and cut it 
into approximately 1.5-centimeter squares. Place the squares

[[Page 832]]

in a glass-stoppered 50-milliliter Erlenmeyer flask and also proceed as 
directed in paragraph (b)(8) (vi), (vii), and (viii) of this section.
    (b) Calculation. Calculate the recovery of total capreomycins as 
follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.066

where:
At=Absorbance of the eluate from the unchromatographed sheet;
Ab=Absorbance of the eluate from the unchromatographed blank sheet;
As=Absorbance of the capreomycin sample solution described in paragraph 
(b)(8)(vii) of this section.


To be a valid assay, the recovery of total capreomycins from the 
unchromatographed sheet must be 100plus-minus2 percent.
    (9) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter, except ignite at 700 deg. C.
    (10) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
[39 FR 19115, May 30, 1974, as amended at 46 FR 60568, Dec. 11, 1981; 50 
FR 19920, May 13, 1985]



Sec. 448.20a  Sterile colistimethate sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Colistimethate sodium is the sodium salt 
of a kind of colistin methane sulfonate or a mixture of two or more such 
salts. It is a white to slightly yellow, odorless, fine powder which is 
freely soluble in water. It is so purified and dried that:
    (i) Its potency is not less than 390 micrograms of colistin base 
equivalent per milligram. If it is packaged for dispensing, its potency 
is satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of colistin base equivalent that it 
is represented to contain.
    (ii) It is sterile.
    (iii) [Reserved]
    (iv) It is nonpyrogenic.
    (v) Its loss on drying is not more than 7.0 percent.
    (vi) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 6.5 and not more than 8.5.
    (vii) It gives a positive identity test for colistimethate sodium.
    (viii) It passes the test for free colistin.
    (ix) Its heavy metals content is not more than 30 parts per million.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, pH, identity, free colistin, and heavy metals.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 10 containers, each containing 
approximately 500 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 12 vials or if each 
vial contains less than 150 milligrams of colistimethate, a minimum of 
60 vials.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
If the batch is packaged for repacking or for use in manufacturing 
another drug, dissolve an accurately weighed sample in 2 milliliters of 
sterile distilled water and further dilute with sufficient 10-percent 
potassium phosphate buffer, pH 6.0 (solution 6), to give a stock 
solution of convenient concentration. If it is packaged for dispensing, 
reconstitute as directed in the labeling. Then, using a suitable 
hypodermic needle and syringe, remove all of the withdrawable contents 
if the container is represented as a single dose container; or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Further dilute the

[[Page 833]]

stock solution with solution 6 to the reference concentration of 1.0 
microgram of colistin base equivalent per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 10 milligrams of colistin base equivalent 
per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
1-percent aqueous solution prepared in the following manner: Weigh 
accurately 0.5 gram of sample and transfer to a 125-milliliter 
Erlenmeyer flask. Add 50 milliliters of freshly boiled distilled water, 
stopper, and shake until the sample is in solution.
    (7) Identity--(i) Infrared. Proceed as directed in Sec. 436.211 of 
this chapter, using a 1-percent potassium bromide disc prepared as 
described in paragraph (b)(1) of that section.
    (ii) Iodine reduction. Dissolve 40 milligrams of sample in 1.0 
milliliter of 1.0N hydrochloric acid and add 0.5 milliliter of 0.02N 
iodine. The color is rapidly discharged.
    (8) Free colistin. Dissolve 80 milligrams of sample in 3.0 
milliliters of distilled water and add 0.05 milliliter of 10 percent w/v 
solution of silico-tungstic acid. It passes the test for free colistin 
if no immediate precipitate is produced.
    (9) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
[39 FR 19115, May 30, 1974, as amended at 44 FR 10381, Feb. 20, 1979; 44 
FR 22059, Apr. 13, 1979; 50 FR 19920, May 13, 1985]



Sec. 448.21  Colistin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Colistin sulfate is the white to slightly 
yellow, odorless sulfate salt of a kind of colistin or a mixture of two 
or more such salts. It is so purified and dried that:
    (i) Its potency is not less than 500 micrograms of colistin per 
milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 7.0 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 4.0 and not more than 7.0.
    (v) It gives a positive identity test for colistin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b).
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, and identity.
    (ii) Samples required on the batch: 10 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in 2 milliliters of sterile 
distilled water and further dilute with sufficient 10 percent potassium 
phosphate buffer, pH 6.0 (solution 6), to give a stock solution of 
convenient concentration. Further dilute the stock solution with 
solution 6 to the reference concentration of 1.0 microgram of colistin 
per milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Identity. To about 20 milligrams of sample, add 2.0 milliliters 
of pH 7.0 buffer (prepared by adding 29.63 milliliters of 1 N sodium 
hydroxide to 50 milliliters of 1 M potassium dihydrogen phosphate, 
adjusting to pH 7.0 if necessary, and diluting to 100 milliliters with 
distilled water) and 0.2 milliliter of a 0.5 percent aqueous 
triketohydrindene hydrate solution, and bring to boil. A purple color is 
produced.
[39 FR 19115, May 30, 1974, as amended at 50 FR 19920, May 13, 1985]

[[Page 834]]



Sec. 448.23  Cyclosporine.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cyclosporine is a cyclic polypeptide 
consisting of 11 amino acids. It is a white or essentially white finely 
crystalline powder. It is so purified and dried that:
    (i) Its cyclosporine content is not less than 975 micrograms per 
milligram and not more than 1,020 micrograms per milligram on the 
anhydrous basis.
    (ii) Its loss on drying is not more than 3.0 percent.
    (iii) Its heavy metals content is not more than 20 parts per 
million.
    (iv) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for cyclosporine 
content, loss on drying, heavy metals, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Cyclosporine content. Proceed as 
directed in Sec. 436.346 of this chapter, except prepare the working 
standard and sample solutions and calculate the cyclosporine content as 
described in paragraphs (b)(1) (i) and (ii) of this section. A typically 
suitable column for cyclosporine is a 250-millimeter column having an 
inside diameter of 4 millimeters packed with octyl silane chemically 
bonded to totally porous microsilica particles, 5 to 7 microns in 
diameter.
    (i) Preparation of working standard and sample solutions.

    Note: Dissolve working standards and samples immediately before 
analysis.

    (a) Preparation of working standard solution. Dissolve an accurately 
weighed portion of the working standard in ethanol by shaking for at 
least 15 minutes. If necessary, ultrasonicate until the solution becomes 
completely clear. Dilute with ethanol to obtain a solution containing 
1,000 micrograms of cyclosporine activity per milliliter.
    (b) Preparation of sample solutions. Prepare all sample solutions as 
directed for preparation of working standard solutions, except dilute 
with ethanol to obtain a solution containing 1,000 micrograms of 
cyclosporine per milliliter (estimated).
    (ii) Calculations. Calculate the micrograms of cyclosporine per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.264

where:

Au=Area of the cyclosporine peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cyclosporine peak in the chromatogram of the 
          cyclosporine working standard;
Ps=Cyclosporine activity in the cyclosporine working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of cyclosporine per milliliter of sample 
          solution; and
m=Percent loss on drying of the sample.

    (2) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (3) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
    (4) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraphs (b)(1) of this section compares 
qualitatively to that of the cyclosporine working standard.
[49 FR 22632, May 31, 1984, as amended at 55 FR 11584, Mar. 29, 1990]



Sec. 448.25  Gramicidin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Gramicidin is the white, or nearly white, 
odorless, crystalline compound of a kind of gramicidin or a mixture of 
two or more such compounds. It is so purified and dried that:
    (i) It has a potency of not less than 900 micrograms of gramicidin 
per milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 3 percent.
    (iv) Its residue on ignition is not more than 1.0 percent.

[[Page 835]]

    (v) Its melting point is not below 229 deg. C after drying in vacuum 
at 60 deg. C for 3 hours.
    (vi) When calculated on the anhydrous basis, the difference between 
the absorptivity value at the maximum occurring at 282 nanometers and 
the absorptivity value at the minimum occurring at 247 nanometers is 
100plus-minus4 percent of the difference obtained with the 
gramicidin working standard.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, residue on ignition, melting point, identity, and crystallinity.
    (ii) Samples required of the batch: Ten packages, each containing 
approximately 500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient alcohol U.S.P. XX to 
obtain a stock solution of convenient concentration. Further dilute the 
stock solution volumetrically with alcohol U.S.P. XX to the reference 
concentration of 0.04 microgram of gramicidin per milliliter 
(estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (5) Melting point. Proceed as directed in Sec. 436.209 of this 
chapter.
    (6) Identity. Accurately weigh about 20 milligrams of the sample and 
dilute in ethyl alcohol to give a concentration of 0.05 milligram 
(estimated) of gramicidin per milliliter. Prepare a solution of the 
gramicidin working standard to contain 0.05 milligram per milliliter in 
ethyl alcohol. Using a suitable recording spectrophotometer with 1-
centimeter cells, record the ultraviolet absorbance spectrum of each 
solution from 220 nanometers to 320 nanometers. The ultraviolet 
absorbance spectrum of the sample solution should compare qualitatively 
to that of the working standard solution. Determine the absorptivities 
of each at the maximum occurring at 282 nanometers and at the minimum 
occurring at 247 nanometers (the exact position of the maximum and 
minimum of the gramicidin working standard should be determined for the 
particular instrument used).
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19115, May 30, 1974, as amended at 41 FR 24883, June 21, 1976; 47 
FR 23710, June 1, 1982; 50 FR 19920, May 13, 1985]



Sec. 448.30  Polymyxin B sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Polymyxin B sulfate is the sulfate salt 
of a kind of polymyxin or a mixture of two or more such salts. It is a 
white to buff-colored powder. It is so purified and dried that:
    (i) Its potency is not less than 6,000 units of polymyxin B per 
milligram on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 7.0 percent.
    (iv) Its pH is an aqueous solution containing 5 milligrams per 
milliliter is not less than 5.0 and not more than 7.5.
    (v) It gives positive color identity tests for polymyxin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, and identity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Add 2.0 milliliters of sterile distilled water to each 5 milligrams of 
an accurately weighed portion of the sample. Dilute with sufficient 10 
percent potassium phosphate buffer, pH 6.0 (solution 6), to give a stock 
solution containing 10,000 units of polymyxin B

[[Page 836]]

per milliliter (estimated). Further dilute an aliquot of the stock 
solution with solution 6 to the reference concentration of 10 units of 
polymyxin B per milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 5 milligrams per milliliter.
    (5) Identity. (i) To a solution of 2 milligrams of the sample in 5.0 
milliliters of water, add 0.5 milliliter of triketohydrindene solution 
(1:1,000) and 2 drops of pyridine. Boil for 1 minute and cool. A blue 
color is a positive test.
    (ii) To a solution of 2 milligrams of the sample in 5 milliliters of 
water, add 5 milliliters of sodium hydroxide solution (1:10) and mix 
well. Add, dropwise, 5 drops of a cupric sulfate solution (1:100), 
mixing after the addition of each drop. A reddish-violet color is a 
positive test.
[40 FR 22253, May 22, 1975, as amended at 50 FR 19920, May 13, 1985]



Sec. 448.30a  Sterile polymyxin B sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Polymyxin B sulfate is the sulfate salt 
of a kind of polymyxin or a mixture of two or more such salts. It is a 
white to buff-colored powder. It is so purified and dried that:
    (i) Its potency is not less than 6,000 units of polymyxin B per 
milligram, on an anhydrous basis. If it is packaged for dispensing, its 
content is satisfactory if it is not less than 90 percent and not more 
than 120 percent of the number of units of polymyxin B that it is 
represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its loss on drying is not more than 7.0 percent.
    (vi) Its pH in an aqueous solution containing 5 milligrams per 
milliliter is not less than 5.0 and not more than 7.5.
    (vii) Its residue on ignition is not more than 5 percent.
    (viii) If it is intended for systemic medication, its heavy metals 
content is not more than 100 parts per million.
    (ix) It gives positive color identity tests for polymyxin.
    (2) Labeling. In addition to the requirements of Sec. 432.5 of this 
chapter, if the drug is packaged for dispensing its labeling shall bear 
the statement, ``Caution: This drug should be given intramuscularly and/
or intrathecally only to hospitalized patients so as to provide constant 
supervision by a physician''.
    (3) Request for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, pH, residue on ignition, heavy metals, and 
identity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use as an 
ingredient in the manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If the drug is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers plus one additional package containing 1 gram of the batch.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in 2 milliliters of sterile 
distilled water for each 5 milligrams of weighed sample. Further dilute 
an aliquot of this solution with sufficient 10 percent potassium 
phosphate buffer, pH 6.0 (solution 6), to give a stock solution of 
convenient concentration; also, if it is packaged for dispensing, 
reconstitute as directed in the labeling. Then using a suitable 
hypodermic needle and syringe, remove all of the withdrawable contents 
if it is represented as a single dose container; or if the labeling 
specifies the amount of potency in a given volume of the resultant 
preparation, remove an accurately measured representative portion

[[Page 837]]

from each container. Dilute with sufficient solution 6 to give a stock 
solution of convenient concentration. Further dilute an aliquot of the 
stock solution with solution 6 to the reference concentration of 10 
units of polymyxin B per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 20,000 units of polymyxin B per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 5 milligrams per milliliter.
    (7) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (8) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
    (9) Identity. (i) To a solution of 2 milligrams of polymyxin B 
sulfate in 5 milliliters of water, add 0.5 milliliter of 
triketohydrindene solution (1:1,000) and 2 drops of pyridine, boil for 1 
minute, and cool; a blue color develops; and
    (ii) To a solution of 2 milligrams of polymyxin B sulfate in 5 
milliliters of water, add 5 milliliters of sodium hydroxide solution 
(1:10), mix well, and add, dropwise, 5 drops of cupric sulfate solution 
(1:100), mixing after the addition of each drop; a reddish-violet color 
is produced.
[39 FR 19115, May 30, 1974, as amended at 46 FR 16683, Mar. 13, 1981; 46 
FR 22359, Apr. 17, 1981; 50 FR 19920, May 13, 1985]



Sec. 448.75  Tyrothricin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tyrothricin is a white to brownish-white 
compound of a kind of tyrothricin or a mixture of two or more such 
compounds. It consists principally of gramicidin and tyrocidine. It is 
so purified and dried that:
    (i) Its potency is not less than 900 micrograms and not more than 
1,400 micrograms of tyrothricin per milligram.
    (ii) Its loss on drying is not more than 5 percent.
    (iii) It gives a positive identity test for tyrothricin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, and identity.
    (ii) Samples required: five packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 95 percent ethyl 
alcohol, U.S.P. XVIII or equivalent, to give a stock solution of 
convenient concentration. Further dilute the stock solution with 95 
percent ethyl alcohol, U.S.P. XVIII or equivalent, to the reference 
concentration of 0.20 microgram of tyrothricin per milliliter 
(estimated). Average the absorbance values for the tyrothricin sample 
and read the gramicidin concentration from the gramicidin standard 
response line. Multiply by 5 to obtain the number of micrograms of 
tyrothricin in the sample.
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Identity. To 5 milliliters of p- dimethylaminobenzaldehyde 
(T.S.) add about 5 milligrams of tyrothricin. Shake well for 2 minutes; 
then add 2 drops of 0.1M sodium nitrite and 5 milliliters of water. A 
blue color is produced.



                      Subpart B--Oral Dosage Forms



Sec. 448.121  Colistin sulfate for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Colistin sulfate for oral suspension is a 
dry mixture of colistin sulfate, with or without one or more suitable 
and harmless buffer substances, suspending and dispersing agents, 
diluents, colorings, and flavorings. The colistin sulfate content is 5.0 
milligrams of colistin per milliliter of the reconstituted suspension. 
Its potency

[[Page 838]]

is satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of colistin that it is represented 
to contain. The loss on drying is not more than 3.0 percent. The pH of 
the reconstituted suspension is not less than 5.0 and not more than 6.0. 
The colistin sulfate used conforms to the standards prescribed by 
Sec. 448.21(a)(1).
    (2) Labeling. It shall be labeled as prescribed in Sec. 432.5 of 
this chapter,
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The colistin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (b) The batch for potency, loss on drying, and pH.
    (ii) Samples required:
    (a) The colistin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute the suspension as directed in the label. Remove an 
accurately measured representative portion of the reconstituted 
suspension with a hypodermic needle and syringe and dilute with 10 
percent potassium phosphate buffer, pH 6.0 (solution 6), to the 
reference concentration of 1.0 microgram of colistin per milliliter 
(estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the suspension when reconstituted as directed in the labeling.
[39 FR 19115, May 30, 1974, as amended at 50 FR 19920, May 13, 1985]



Sec. 448.123  Cyclosporine oral dosage forms.



Sec. 448.123a  Cyclosporine oral solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cyclosporine oral solution contains, in 
each milliliter, 100 milligrams of cyclosporine in a suitable and 
harmless alcohol-vegetable oil solution. Its cyclosporine content is 
satisfactory if it is not less than 90 percent and not more than 110 
percent of the number of milligrams of cyclosporine that it is 
represented to contain. The cyclosporine used conforms to the standards 
prescribed by Sec. 448.23, except heavy metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cyclosporine used in making the batch for cyclosporine 
content, loss on drying, and identity.
    (b) The batch for cyclosporine content.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The cyclosporine used in making the batch: Six packages, each 
containing approximately 500 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay; cyclosporine content. Proceed as 
directed in Sec. 436.346 of this chapter, except prepare the working 
standard and sample solutions and calculate the cyclosporine content as 
described in paragraphs (b) (1) and (2) of this section. A typically 
suitable column for cyclosporine dosage forms is 250 millimeters long 
having an inside diameter of 4 millimeters packed with dimethyl silane 
chemically bonded to porous silica particles 10 microns in diameter [RP-
2 (E.M. Science, S. Plainfield, NJ)].
    (1) Preparation of working standard and sample solutions.

    Note: Prepare working standard and sample solutions immediately 
before analysis.

    (i) Preparation of working standard solution. Dissolve an accurately 
weighed portion of the working standard in ethanol by shaking for at 
least 15 minutes. If necessary, ultrasonicate until the solution becomes 
completely clear. Dilute with ethanol to obtain a solution containing 1 
milligram of cyclosporine activity per milliliter.

[[Page 839]]

    (ii) Preparation of sample solution. Place an accurately measured 
representative volume of the cyclosporine oral solution into a 
volumetric flask. Add sufficient ethanol to obtain a concentration of 1 
milligram of cyclosporine activity per milliliter (estimated).
    (2) Calculations. Calculate the cyclosporine content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.265
    
where:

Au=Area of the cyclosporine peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cyclosporine peak in the chromatogram of the 
          cyclosporine working standard;
Ps=Cyclosporine activity of the cyclosporine working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.
[49 FR 22633, May 31, 1984, as amended at 55 FR 11584, Mar. 29, 1990. 
Redesignated at 55 FR 19873, May 14, 1990]



Sec. 448.123b  Cyclosporine capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cyclosporine capsules are composed of 
cyclosporine in a suitable and harmless alcohol-vegetable oil solution 
enclosed by a soft gelatin capsule. Its cyclosporine content is 
satisfactory if it is not less than 90 percent and not more than 110 
percent of the number of milligrams of cyclosporine that it is 
represented to contain. The capsules shall disintegrate within 30 
minutes. The cyclosporine used conforms to the standards prescribed by 
Sec. 448.23, except heavy metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The cyclosporine used in making the batch for cyclosporine 
content, loss on drying, and identity.
    (B) The batch for cyclosporine content and disintegration time.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research:
    (A) The cyclosporine used in making the batch: Six packages, each 
containing approximately 500 milligrams.
    (B) The batch: A minimum of 36 capsules.
    (b) Test and methods of assay--(1) Cyclosporine content. Proceed as 
directed in Sec. 436.346 of this chapter, except prepare the working 
standard and sample solutions and calculate the cyclosporine content as 
described in paragraphs (b)(1)(i) (A) and (B) and (b)(1)(ii) of this 
section. A typically suitable column for cyclosporine dosage forms is 
250 millimeters long having an inside diameter of 4 millimeters packed 
with dimethyl silane chemically bonded to porous silica particles 10 
microns in diameter (RP-2 (E.M. Science, S. Plainfield, NJ)).
    (i) Preparation of working standard and sample solutions.

    Note: Prepare working standard and sample solutions immediately 
before analysis.

    (A) Working standard solution. Dissolve an accurately weighed 
portion of the working standard in ethanol by shaking for at least 15 
minutes. If necessary, ultrasonicate until the solution becomes 
completely clear. Dilute with ethanol to obtain a solution containing 1 
milligram of cyclosporine activity per milliliter.
    (B) Sample solution. Cut open a representative number of capsules 
with a sharp blade and quantitatively transfer the capsule contents to a 
volumetric flask. Add sufficient ethanol to obtain a concentration of 1 
milligram of cyclosporine activity per milliliter (estimated).
    (ii) Calculations. Calculate the cyclosporine content in milligrams 
per capsule as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.067

where:

    Au=Area of the cyclosporine peak in the chromatogram of 
the sample (at a retention time equal to that observed for the 
standard);
    As=Area of the cyclosporine peak in the chromatogram of 
the cyclosporine working standard;

[[Page 840]]

    Ps=Cyclosporine content in the cyclosporine working 
standard solution in micrograms per milliliter;
    d=Dilution factor of the sample; and
    n=Number of capsules in the sample assayed.

    (2) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure described in Sec. 436.212(e)(5).
[55 FR 19873, May 14, 1990; 55 FR 22014, May 30, 1990]



                   Subpart C--Injectable Dosage Forms



Sec. 448.210  Sterile bacitracin.

    The requirements for certification and the tests and methods of 
assay for sterile bacitracin packaged for dispensing are described in 
Sec. 448.10a.



Sec. 448.215  Sterile capreomycin sulfate.

    The requirements for certification and the tests and methods of 
assay for sterile capreomycin sulfate packaged for dispensing are 
described in Sec. 448.15a.



Sec. 448.220  Colistimethate sodium injectable dosage forms.



Sec. 448.220a  Sterile colistimethate sodium.

    The requirements for certification and the tests and methods of 
assay for sterile colistimethate sodium packaged for dispensing are 
described in Sec. 448.20a.



Sec. 448.223  Cyclosporine for infusion.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cyclosporine for infusion is a solution 
of cyclosporine in a suitable and harmless alcohol derivatized vegetable 
oil vehicle. Its cyclosporine content is satisfactory if it is not less 
than 90 percent and not more than 110 percent of the number of 
milligrams of cyclosporine that it is represented to contain. It is 
sterile. It contains not more than 42 endotoxin units per milliliter 
(United States Pharmacopeia endotoxin units). The cyclosporine used 
conforms to the standards prescribed by Sec. 448.23.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The cyclosporine used in making the batch for cyclosporine 
content, loss on drying, heavy metals, and identity.
    (b) The batch for cyclosporine content, sterility, and bacterial 
endotoxins.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The cyclosporine used in making the batch: Six packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Cyclosporine content. Proceed as 
directed in Sec. 436.346 of this chapter, except prepare the working 
standard and sample solutions and calculate the cyclosporine content as 
described in paragraphs (b)(1) (i) and (ii) of this section. A typically 
suitable column for cyclosporine dosage forms is 250 millimeters long 
having an inside diameter of 4 millimeters packed with dimethyl silane 
chemically bonded to porous silica particles 10 microns in diameter [RP-
2 (E.M. Science, S. Plainfield, NJ)].
    (i) Preparation of working standard and sample solutions.

    Note: Prepare working standard and sample solutions immediately 
before analysis.

    (a) Preparation of working standard solution. Dissolve an accurately 
weighed portion of the working standard in ethanol by shaking for at 
least 15 minutes. If necessary, ultrasonicate until the solution becomes 
completely clear. Dilute with ethanol to obtain a solution containing 1 
milligram of cyclosporine activity per milliliter.
    (b) Preparation of sample solution. Using a suitable hypodermic 
needle and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the concentration of cyclosporine in a given volume of the resultant 
preparation, remove an

[[Page 841]]

accurately measured portion from each container. Dilute with ethanol to 
obtain a stock solution of 1 milligram of cyclosporine activity per 
milliliter (estimated).
    (ii) Calculations. Calculate the cyclosporine content of the vial as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.266

where:
Au=Area of the cyclosporine peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the cyclosporine peak in the chromatogram of the 
          cyclosporine working standard;
Ps=Cyclosporine activity in the cyclosporine working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section.
    (3) Bacterial endotoxins. Proceed as directed in the United States 
Pharmacopeia XX bacterial endotoxins test.
[49 FR 22633, May 31, 1984, as amended at 55 FR 11584, Mar. 29, 1990]



Sec. 448.230  Sterile polymyxin B sulfate.

    The requirements for certification and the tests and methods of 
assay for sterile polymyxin B sulfate packaged for dispensing are 
described in Sec. 448.30a.
[44 FR 10379, Feb. 20, 1979]



                   Subpart D--Ophthalmic Dosage Forms



Sec. 448.310  Bacitracin ophthalmic dosage forms.



Sec. 448.310a  [Reserved]



Sec. 448.310b  Bacitracin-neomycin sulfate-polymyxin B sulfate ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin-neomycin sulfate-polymyxin B 
sulfate ophthalmic ointment contains bacitracin, neomycin sulfate, and 
polymyxin B sulfate in a suitable and harmless ointment base. Each gram 
contains 500 units of bacitracin, 3.5 milligrams of neomycin, and 10,000 
units of polymyxin B. Its bacitracin content is satisfactory if it is 
not less than 90 percent and not more than 140 percent of the number of 
units of bacitracin that it is represented to contain. Its neomycin 
content is satisfactory if it is not less than 90 percent and not more 
than 140 percent of the number of milligrams of neomycin that it is 
represented to contain. Its polymyxin B content is satisfactory if it is 
not less than 90 percent and not more than 140 percent of the number of 
units of polymyxin B that it is represented to contain. It is sterile. 
Its moisture content is not more than 0.5 percent. It passes the test 
for metal particles. The bacitracin used conforms to the standards 
prescribed by Sec. 448.10a(a)(1), except pyrogens, residue on ignition, 
and heavy metals. The neomycin sulfate used conforms to the standards 
prescribed by Sec. 444.42a(a)(1) of this chapter, except pyrogens. The 
polymixin B sulfate used conforms to the standards prescribed by 
Sec. 448.30a(a)(1), except pyrogens, residue on ignition, and heavy 
metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacitracin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (c) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (d) The batch for bacitracin content, neomycin content, polymyxin B 
content, sterility, moisture, and metal particles.
    (ii) Samples required:
    (a) The bacitracin used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (b) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 1.0 gram.

[[Page 842]]

    (c) The polymixin B sulfate used in making the batch: 10 packages, 
each containing approximately 1.0 gram.
    (d) The batch:
    (1) For all tests except sterility: A minimum of 17 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assays--(1) Potency--(i) Bacitracin 
content. Proceed as directed for bacitracin zinc in Sec. 436.105 of this 
chapter, preparing the sample for assay as follows: Place an accurately 
weighed representative portion of the sample into a separatory funnel 
containing approximately 50 milliliters of peroxide-free ether. Shake 
the sample and ether until homogeneous. Add 20 to 25 milliliters of 1 
percent potassium phosphate buffer, pH 6.0 (solution 1), and shake well. 
Allow the layers to separate. Remove the buffer layer and repeat the 
extraction procedure with each of three more 20- to 25-milliliter 
quantities of solution 1. Combine the buffer extractives in a suitable 
volumetric flask and dilute to volume with solution 1. Remove an 
aliquot, add sufficient hydrochloric acid so that the amount of acid in 
the final solution will be the same as in the reference concentration of 
the working standard and further dilute with solution 1 to the reference 
concentration of 1.0 unit of bacitracin per milliliter (estimated).
    (ii) Neomycin content. Proceed as directed in Sec. 436.105 of this 
chapter, preparing the sample for assay as follows: Place an accurately 
weighed representative portion of the sample into a separatory funnel 
containing approximately 50 milliliters of peroxide-free ether. Shake 
the sample and ether until homogeneous. Add 20 to 25 milliliters of 0.1M 
potassium phosphate buffer, pH 8.0 (solution 3), and shake well. Allow 
the layers to separate. Remove the buffer layer and repeat the 
extraction procedure with each of three more 20- to 25-milliliter 
quantities of solution 3. Combine the buffer extractives in a suitable 
volumetric flask and dilute to volume with solution 3. Remove an aliquot 
and further dilute with solution 3 to the reference concentration of 1.0 
microgram of neomycin per milliliter (estimated).
    (iii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, except add to each concentration of the polymyxin standard 
response line a quantity of neomycin to yield the same concentration of 
neomycin as that present when the sample is diluted to contain 10 units 
of polymyxin B per milliliter. Prepare the sample for assay as follows: 
Place an accurately weighed representative portion of the sample into a 
separatory funnel containing approximately 50 milliliters of peroxide-
free ether. Shake the sample and ether until homogeneous. Add 20 to 25 
milliliters of 10 percent potassium phosphate buffer, pH 6.0 (solution 
6), and shake well. Allow the layers to separate. Remove the buffer 
layer and repeat the extraction procedure with each of three more 20- to 
25-milliliter quantities of solution 6. Combine the buffer extractives 
in a suitable volumetric flask and dilute to volume with solution 6. 
Remove an aliquot and futher dilute with solution 6 to the reference 
concentration of 10 units of Polymyxin B per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) Metal particles. Proceed as directed in Sec. 436.206 of this 
chapter.
[42 FR 27230, May 27, 1977, as amended at 47 FR 23442, May 28, 1982; 50 
FR 19920, May 13, 1985]



Sec. 448.310c  Bacitracin ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strenght, quality, and purity. Bacitracin ophthalmic ointment contains 
bacitracin in a suitable and harmless ointment base. Each gram contains 
500 units of bacitracin. Its potency is satisfactory if it is not less 
than 90 percent and not more than 140 percent of the number of units of 
bacitracin that it is represented to contain. It is sterile. Its 
moisture content is not more than 0.5 percent. It passes the test for 
metal particles. The bacitracin used conforms to the standards 
prescribed by Sec. 448.10a(a)(1), except pyrogens, residue on ignition, 
and heavy metals.

[[Page 843]]

    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacitracin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency, sterility, moisture, and metal particles.
    (ii) Samples required:
    (a) The bacitracin used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 15 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed for 
bacitracin zinc in Sec. 436.105 of this chapter, preparing the sample 
for assay as follows: Place an accurately weighed representative portion 
of the sample into a separatory funnel containing approximately 50 
milliliters of peroxide-free ether. Shake the sample and ether until 
homogeneous. Add 20 to 25 milliliters of 1 percent potassium phosphate 
buffer, pH 6.0 (solution 1), and shake well. Allow the layers to 
separate. Remove the buffer layer and repeat the extraction procedure 
with each of three more 20- to 25-milliliter quantities of solution 1. 
Combine the buffer extractives in a suitable volumetric flask and dilute 
to volume with solution 1. Remove an aliquot, add sufficient 
hydrochloric acid so that the amount of acid in the final solution will 
be the same as in the reference concentration of the working standard 
and further dilute with solution 1 to the reference concentration of 1.0 
unit of bacitracin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) Metal particles. Proceed as directed in Sec. 436.206 of this 
chapter.
[42 FR 27230, May 27, 1977, as amended at 50 FR 19920, May 13, 1985]



Sec. 448.313  Bacitracin zinc ophthalmic dosage forms.



Sec. 448.313a  Bacitracin zinc-polymyxin B sulfate ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin zinc-polymyxin B sulfate 
ophthalmic ointment contains in each gram 500 units of a bacitracin and 
10,000 units of polymyxin B in a suitable and harmless ointment base. 
Its bacitracin content is satisfactory if it is not less than 90 percent 
and not more than 130 percent of the number of units of bacitracin that 
it is represented to contain. Its polymyxin B content is satisfactory if 
it is not less than 90 percent and not more than 130 percent of the 
number of units of polymyxin B that it is represented to contain. It is 
sterile. Its moisture content is not more than 0.5 percent. It passes 
the test for metal particles. The bacitracin zinc used conforms to the 
standards prescribed by Sec. 448.13a(a)(1). The polymyxin B sulfate used 
conforms to the standards prescribed by Sec. 448.30a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacitracin zinc used in making the batch for potency, loss 
on drying, pH, zinc content, and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (c) The batch for bacitracin content, polymyxin B content, 
sterility, moisture, and metal particles.
    (ii) Samples required:
    (a) The bacitracin zinc used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 1.0 gram.
    (c) The batch:

[[Page 844]]

    (1) For all tests except sterility: A minimum of 17 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Bacitracin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Place an accurately weighed representative 
portion of the sample into a separatory funnel containing approximately 
50 milliliters of peroxide-free ether. Shake the sample and ether until 
homogeneous. Add 20 to 25 milliliters of 0.01N hydrochoric acid and 
shake well. Allow the layers to separate. Remove the acid layer and 
repeat the extraction procedure with each of three more 20- to 25-
milliliter quantities of 0.01N hydrochloric acid. Combine the acid 
extractives in a suitable volumetric flask and dilute to volume with 
0.01N hydrochloric acid. (If the bacitracin content is less than 100 
units per milliliter in 0.01N hydrochloric acid, add sufficient 
additional hydrochloric acid to each concentration of the standard 
response line so that each standard solution contains the same amount of 
acid as the final sample solution.) Remove an aliquot and further dilute 
with solution 1 to the reference concentration of 1.0 unit of bacitracin 
per milliliter (estimated).
    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, preparing the sample for assay as follows: Place an 
accurately weighed representative portion of the sample into a 
separatory funnel containing approximately 50 milliliters of peroxide-
free ether. Shake the sample and ether until homogeneous. Add 20 to 25 
milliliters of 10 percent potassium phosphate buffer, pH 6.0 (solution 
6) and shake well. Allow the layers to separate. Remove the buffer layer 
and repeat the extraction procedure with each of three more 20- to 25-
milliliter quantities of solution 6. Combine the buffer extractives in a 
suitable volumetric flask and dilute to volume with solution 6. Remove 
an aliquot and further dilute with solution 6 to the reference 
concentration of 10 units of polymyxin B per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) Metal particles. Proceed as directed in Sec. 436.206 of this 
chapter.
[42 FR 27231, May 27, 1977, as amended at 49 FR 34351, Aug. 30, 1984; 50 
FR 19920, May 13, 1985]



Sec. 448.313b  Bacitracin zinc-neomycin sulfate-polymyxin B sulfate ophthalmic ointment; bacitracin zinc-neomycin sulfate-polymyxin B sulfate-hydrocortisone 
          ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin zinc-neomycin sulfate-
polymyxin B sulfate ophthalmic ointment is bacitracin zinc, neomycin 
sulfate, and polymyxin B sulfate in a suitable and harmless ointment 
base. Each gram contains:
    (i) 400 units of bacitracin, 3.5 milligrams of neomycin, 10,000 
units of polymyxin B with or without 10 milligrams of hydrocortisone 
acetate;
    (ii) 500 units of bacitracin, 3.5 milligrams of neomycin, 10,000 
units of polymyxin B; or
    (iii) 400 units of bacitracin, 3.5 milligrams of neomycin, 10,000 
units of polymyxin B, and 10 milligrams of hydrocortisone.

Its bacitracin content is satisfactory if it is not less than 90 percent 
and not more than 140 percent of the number of units of bacitracin that 
it is represented to contain. Its neomycin content is satisfactory if it 
is not less than 90 percent and not more than 140 percent of the number 
of milligrams of neomycin that it is represented to contain. Its 
polymyxin B content is satisfactory if it is not less than 90 percent 
and not more than 140 percent of the number of units of polymyxin B that 
it is represented to contain. It is sterile. Its moisture content is not 
more than 0.5 percent. It passes the test for metal particles. The 
bacitracin zinc used conforms to the standards prescribed by 
Sec. 448.13a(a)(1). The neomycin sulfate used conforms to the standards 
prescribed by Sec. 444.42a(a)(1) of this chapter, except pyrogens. The 
polymyxin B sulfate used conforms to the standards prescribed by 
Sec. 448.30a(a)(1), except

[[Page 845]]

pyrogens, residue on ignition, and heavy metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacitracin zinc used in making the batch for potency, loss 
on drying, pH, zinc content, and identity.
    (b) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (c) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (d) the batch for bacitracin content, neomycin content, polymyxin B 
content, sterility, moisture, and metal particles.
    (ii) Samples required:
    (a) The bacitracin zinc used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (b) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (c) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 1.0 gram.
    (d) The batch:
    (1) For all tests except sterility: A minimum of 17 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Bacitracin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Place an accurately weighed representative 
portion of the sample into a separatory funnel containing approximately 
50 milliliters of peroxide-free ether. Shake the sample and ether until 
homogeneous. Add 20 to 25 milliliters of 0.01N hydrochloric acid and 
shake well. Allow the layers to separate. Remove the acid layer and 
repeat the extration procedure with each of three more 20- to 25-
milliliter quantities of 0.01N hydrochloric acid. Combine the acid 
extractives in a suitable volumetric flask and dilute to volume with 
0.01N hydrochloric acid. (If the bacitracin content is less than 100 
units per milliliter in 0.01N hydrochloric acid, add sufficient 
additional hydrochloric acid to each concentration of the standard 
response line so that each standard solution contains the same amount of 
acid as the 1.0 unit per milliliter sample solution.) Remove an aliquot 
and further dilute with 1 percent potassium phosphate buffer, pH 6.0 
(solution 1) to the reference concentration of 1.0 unit of bacitracin 
per milliliter (estimated).
    (ii) Neomycin content. Proceed as directed in Sec. 436.105 of this 
chapter, preparing the sample for assay as follows: Place an accurately 
weighed representative portion of the sample into a separatory funnel 
containing approximately 50 milliliters of peroxide-free ether. Shake 
the sample and ether until homogeneous. Add 20 to 25 milliliters of 
0.01M potassium phosphate buffer, pH 8.0 (solution 3), and shake well. 
Allow the layers to separate. Remove the buffer layer and repeat the 
extraction procedure with each of three more 20- to 25-milliliter 
quantities of solution 3. Combine the buffer extractives in a suitable 
volumetric flask and dilute to volume with solution 3. Remove an aliquot 
and further dilute with solution 3 to the reference concentration of 1.0 
microgram of neomycin per milliliter (estimated).
    (iii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, except add to each concentration of the polymyxin B 
standard response line a quantity of neomycin to yield the same 
concentration of neomycin as that present when the sample is diluted to 
contain 10 units of polymyxin B per milliliter. Prepare the sample for 
assay as follows: Place an accurately weighed representative portion of 
the sample into a separatory funnel containing approximately 50 
milliliters of peroxide-free ether. Shake the sample and ether until 
homogeneous. Add 20 to 25 milliliters of 10 percent potassium phosphate 
buffer, pH 6.0 (solution 6), and shake well. Allow the layers to 
separate. Remove the buffer layer and repeat the extraction procedure 
with each of 3 more 20- to 25-milliliter quantities of solution 6. 
Combine the buffer extractives in a suitable volumetric flask and dilute 
to volume with solution 6. Remove an aliquot and further

[[Page 846]]

dilute with solution 6 to the reference concentration of 10 units of 
polymyxin B per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) Metal particles. Proceed as directed in Sec. 436.206 of this 
chapter.
[42 FR 27231, May 27, 1977, as amended at 47 FR 23443, May 28, 1982; 48 
FR 21564, May 13, 1983; 50 FR 19920, May 13, 1985]



Sec. 448.321  Colistin sulfate for ophthalmic solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Colistin sulfate for ophthalmic solution 
is a dry mixture of colistin sulfate and mannitol packaged in 
combination with a suitable and harmless diluting solution which 
contains buffers and a preservative. When reconstituted as directed in 
the labeling, each milliliter contains 1.2 milligrams of colistin. Its 
potency is satisfactory if it contains not less than 90 percent and not 
more than 120 percent of the number of milligrams of colistin that it is 
represented to contain. It is sterile. Its loss on drying is not more 
than 5 percent. When reconstituted as directed in the labeling, its pH 
is not less than 5.5 and not more than 6.3. The colistin sulfate used 
conforms to the standards prescribed by Sec. 448.21(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The colistin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (b) The batch for potency, sterility, loss on drying, and pH.
    (ii) Samples required:
    (a) The colistin sulfate used in making the batch: 10 containers, 
each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 6 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Remove an accurately measured 
representative portion of the reconstituted solution and dilute with 10 
percent potassium phosphate buffer, pH 6.0 (solution 6), to the 
reference concentration of 1.0 microgram of colistin per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution reconstituted as directed in the labeling.
[39 FR 19115, May 30, 1974, as amended at 50 FR 19920, May 13, 1985]



Sec. 448.330  Polymyxin B sulfate-trimethoprim hemisulfate ophthalmic solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Polymyxin B sulfate-trimethoprim 
hemisulfate ophthalmic solution contains, in each milliliter, 10,000 
units of polymyxin B and 1.0 milligram of trimethoprim in a suitable and 
harmless isotonic aqueous vehicle. It contains suitable and harmless 
buffers and preservatives. Its polymyxin B content is satisfactory if it 
is not less than 90 percent and not more than 130 percent of the number 
of units of polymyxin B that it is represented to contain. Its 
trimethoprim content is satisfactory if it is not less than 90 percent 
and not more than 110 percent of the number of milligrams of 
trimethoprim that it is represented to contain. It is sterile. Its pH is 
not less than 3.0 and not more than 5.5. The polymyxin B sulfate used 
conforms to the standards prescribed by Sec. 448.30(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:

[[Page 847]]

    (i) Results of tests and assays on:
    (A) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (B) The trimethoprim used in making the batch for all U.S.P. 
specifications.
    (C) The batch for polymyxin B content, trimethoprim content, 
sterility, and pH.
    (ii) Samples if required by the Director, Center for Drug Evaluation 
and Research:
    (A) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (B) The trimethoprim hemisulfate used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (C) The batch:
    (1) For all tests except sterility: A minimum of 7 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Polymyxin content. Proceed as 
directed in Sec. 436.105 of this chapter, preparing the sample for assay 
as follows: Dilute an accurately measured representative portion of the 
sample with 10 percent potassium phosphate buffer, pH 6.0 (solution 10), 
to obtain a stock solution of convenient concentration. Further dilute 
an aliquot of the stock solution with solution 6 to the reference 
concentration of 10 units of polymyxin B per milliliter (estimated).
    (2) Trimethoprim content. Proceed as directed in Sec. 436.216 of 
this chapter, using ambient temperature, an ultraviolet detection system 
operating at a wavelength of 254 nanometers, and a column packed with 
octyl, octadecyl, or phenyl groups chemically bonded to porous silica 
ranging from 3 to 10 micrometers in particle size. Reagents, working 
standard solution, sample solution, resolution test solution, system 
suitability requirements and calculations are as follows:
    (i) Reagents--(A) Diluting fluid. 13 percent acetonitrile in 0.01M 
hydrochloric acid.
    (B) Mobile phase. Mix 0.015M ethanesulfonic acid in acetonitrile: 
water (130:870) and adjust to pH 3.5 with 50 percent w/w sodium 
hydroxide and hydrochloric acid solution. Filter the mobile phase 
through a suitable filter capable of removing particulate matter to 0.5 
micron in diameter. Degas the mobile phase just prior to its 
introduction into the chromatograph.
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(A) Working standard solution. Place approximately 40 
milligrams of trimethoprim working standard, accurately weighed, into a 
50-milliliter volumetric flask. Dissolve and dilute to volume with 13 
percent acetonitrile in 0.01M hydrochloric acid, and mix. Transfer 5 
milliliters of this solution to a 50-milliliter volumetric flask, and 
dilute to volume with 13 percent acetonitrile in 0.01M hydrochloric 
acid, and mix.
    (B) Sample solution. Transfer 4.0 milliliters of the sample into a 
50-milliliter volumetric flask and dilute to volume with 13 percent 
acetonitrile in 0.01M hydrochloric acid.
    (C) Resolution test solution. Place approximately 40 milligrams of 
trimethoprim working standard and 15 milligrams of 2-amino-4-hydroxy-5-
(3',4'5')-trimethoxybenzyl pyrimidine, accurately weighed, into a 50-
milliliter volumetric flask. Dissolve and dilute to volume with 13 
percent acetonitrile in 0.01M hydrochloric acid, and mix. Transfer 5 
milliliters of this solution to a 50-milliliter volumetric flask, and 
dilute to volume with 13 percent acetonitrile in 0.01M hydrochloric 
acid, and mix. Prepare the resolution test solution just prior to its 
introduction into the chromatograph pumping system.
    (iii) System suitability requirements--(A) Asymmetry factor. The 
asymmetry factor (As is satisfactory if it is not more than 
1.4 at 10 percent of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,500 theoretical plates.
    (C) Resolution. The resolution (R) between 2-amino-4-hydroxy-5-
(3',4',5')-trimethoxybenzyl pyrimidine (AHTP) and trimethoprim is 
satisfactory if it is not more than 1.5.
    (D) Coefficient of variation. The coefficient of variation (SR 
in percent) of five replicate injections is satisfactory if it

[[Page 848]]

is not more than 2.0 percent. If the system suitability parameters have 
been met, then proceed as described in Sec. 436.216(b) of this chapter.
    (iv) Calculations. Calculate the milligrams of trimethoprim per 
milliliter of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.267

where:

Au=Area of the trimethoprim peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the trimethoprim peak in the chromatogram of the 
          trimethroprim working standard;
Ws=Weight of the trimethoprim working standard in milligrams; 
          and
0.855=Gravimetric conversion factor trimethoprim hemisulfate to 
          trimethoprim.

    (3) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[54 FR 38376, Sept. 18, 1989, as amended at 59 FR 8399, Feb. 22, 1994]



                      Subpart E--Otic Dosage Forms



Sec. 448.421  Colistin sulfate-neomycin sulfate-thonzonium bromide-hydrocortisone acetate otic suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Colistin sulfate-neomycin sulfate-
thonzonium bromide-hydrocortisone acetate otic suspension is a 
suspension containing colistin sulfate, neomycin sulfate, thonzonium 
bromide, and hydrocortisone acetate, and one or more preservatives, 
dispersing agents, and buffer substances. Each milliliter contains 3.0 
milligrams of colistin, 3.3 milligrams of neomycin, 0.5 milligram of 
thonzonium bromide, and 10 milligrams of hydrocortisone acetate. Its 
content of colistin is satisfactory if it is not less than 90 percent 
and not more than 135 percent of the number of milligrams of colistin 
per milliliter that it is represented to contain. Its content of 
neomycin is satisfactory if it is not less than 90 percent and not more 
than 125 percent of the number of milligrams of neomycin per milliliter 
that it is represented to contain. It is sterile. Its pH is not less 
than 4.8 and not more than 5.2. The colistin sulfate used conforms to 
the standards prescribed therefor by Sec. 448.21(a)(1). The neomycin 
sulfate used conforms to the standards prescribed in Sec. 444.42a(a)(1) 
(i), (v), (vi), and (vii) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The colistin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (b) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (c) The batch for colistin content, neomycin content, sterility, and 
pH.
    (ii) Samples required:
    (a) The colistin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (c) The batch:
    (1) For all tests except sterility: A minimum of six immediate 
containers.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Colistin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Thoroughly mix the sample and transfer an 
accurately measured representative portion of the sample into a 100-
milliliter volumetric flask. Fill the flask to mark with 10 percent 
potassium phosphate buffer, pH 6.0 (solution 6). Further dilute with 
solution 6 to the reference concentration of 1.0 microgram of colistin 
per milliliter (estimated).
    (ii) Neomycin content. Proceed as directed in Sec. 436.105 of this 
chapter, preparing the sample for assay as follows:

[[Page 849]]

Thoroughly mix the sample and transfer an accurately measured 
representative portion into a 100-milliliter volumetric flask. Fill the 
flask to mark with 0.1M potassium phosphate buffer, pH 8.0 (solution 3). 
Further dilute with solution 3 to the reference concentration of 1.0 
microgram of neomycin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section, except 
transfer 0.25 milliliter of sample in lieu of 1 milliliter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted suspension.
[39 FR 19115, May 30, 1974, as amended at 50 FR 19920, May 13, 1985]



Sec. 448.430  Polymyxin B sulfate-hydrocortisone otic solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Polymyxin B sulfate-hydrocortisone otic 
solution contains in each milliliter 10,000 units of polymyxin B and 5 
milligrams of hydrocortisone in a suitable and harmless vehicle. Its 
polymyxin B sulfate content is satisfactory if it contains not less than 
90 percent and not more than 130 percent of the number of units of 
polymyxin B that it is represented to contain. It is sterile. Its pH is 
not less than 3.0 and not more than 5.0. The polymyxin B sulfate used 
conforms to the standards prescribed by Sec. 448.30(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain the following:
    (i) Results of tests and assays on--
    (a) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity; and
    (b) The batch for potency, sterility, and pH.
    (ii) Samples required:
    (a) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 5 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dilute an accurately measured representative portion of the sample 
(usually 1.0 milliliter) with 10 percent potassium phosphate buffer, pH 
6.0 (solution 6), to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of the stock solution with 
solution 6 to the reference concentration of 10 units of polymyxin B per 
milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
if the steroid prevents solubilization, use 0.25 milliliter of the 
sample in lieu of 1 milliliter and proceed as directed in paragraph 
(e)(2) of that section.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter using 
the undiluted solution.
[44 FR 5880, Jan. 30, 1979, as amended at 50 FR 1504, Jan. 11, 1985; 50 
FR 19920, May 13, 1985]



                  Subpart F--Dermatologic Dosage Forms



Sec. 448.510  Bacitracin dermatologic dosage forms.



Sec. 448.510a  Bacitracin ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin ointment is composed of 500 
units of bacitracin per gram in a suitable ointment base. It may contain 
a suitable local anesthetic. Its potency is satisfactory if it is not 
less than 90 percent and not more than 140 percent of the number of 
units of bacitracin that it is represented to contain. Its moisture 
content is not more than 0.5 percent. The bacitracin used conforms to 
the standards described by Sec. 448.10(a)(1).
    (2) Labeling. (i) On the label of the immediate container and on the 
outside wrapper or container, if any:
    (a) The batch mark.

[[Page 850]]

    (b) The name and quantity of each active ingredient contained in the 
drug.
    (c) An expiration date that conforms to the requirements prescribed 
by Sec. 432.5(a)(3) of this chapter.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacitracin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The bacitracin used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed for 
bacitracin zinc in Sec. 436.105 of this chapter, preparing the sample 
for assay as follows: Place an accurately weighed representative portion 
of the sample into a separatory funnel containing approximately 50 
milliliters of peroxide-free ether. Shake the sample and ether until 
homogeneous. Add 20 to 25 milliliters of 1 percent potassium phosphate 
buffer, pH 6.0 (solution 1) and shake well. Allow the layers to 
separate. Remove the buffer layer and repeat the extraction procedure 
with each of three more 20- to 25-milliliter quantities of solution 1. 
Combine the buffer extractives in a suitable volumetric flask and dilute 
to volume with solution 1. remove an aliquot, add sufficient 
hydrochloric acid so that the amount of acid in the final solution will 
be the same as in the reference concentration of the working standard 
and further dilute with solution 1 to the reference concentration of 1.0 
unit of bacitracin per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[42 FR 27232, Nov. 27, 1977, as amended at 50 FR 19920, May 13, 1985]



Secs. 448.510b--448.510c  [Reserved]



Sec. 448.510d  Bacitracin-neomycin sulfate ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin-neomycin sulfate ointment 
contains bacitracin and neomycin sulfate in a suitable ointment base. 
Each gram contains 500 units of bacitracin and 3.5 milligrams of 
neomycin. Its bacitracin content is satisfactory if it is not less than 
90 percent and not more than 130 percent of the number of units of 
bacitracin that it is represented to contain. Its neomycin content is 
satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of milligrams of neomycin that it is represented 
to contain. The moisture content is not more than 0.5 percent. The 
bacitracin used conforms to the standards prescribed by 
Sec. 448.10(a)(1). The neomycin sulfate used conforms to the standards 
prescribed by Sec. 444.42(a)(1) of this chapter.
    (2) Labeling. (i) On the label of the immediate container and on the 
outside wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (c) An expiration date that conforms to the requirements prescribed 
by Sec. 432.5(a)(3) of this chapter.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safety and efficaciously.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacitracin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (c) The batch for bacitracin content, neomycin content, and 
moisture.
    (ii) Samples required:
    (a) The bacitracin used in making the batch: 10 packages, each 
containing approximately 1.0 gram.

[[Page 851]]

    (b) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (c) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Bacitracin content. 
Proceed as directed for bacitracin zinc in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Place an accurately weighed 
representative portion of the sample into a separatory funnel containing 
approximately 50 milliliters of peroxide-free ether. Shake the sample 
and ether until homogeneous. Add 20 to 25 milliliters of 1 percent 
potassium phosphate buffer, pH 6.0 (solution 1), and shake well. Allow 
the layers to separate. Remove the buffer layer and repeat the 
extraction procedure with each of three more 20- to 25 milliliter 
quantities of solution 1. Combine the buffer extractives in a suitable 
volumetric flask and dilute to volume with solution. 1. Remove an 
aliquot, add sufficient hydrochloric acid so that the amount of acid in 
the final solution will be the same as in the reference concentration of 
the working standard and further dilute with solution 1 to the reference 
concentration of 1.0 unit of bacitracin per milliliter (estimated).
    (ii) Neomycin content. Proceed as directed in Sec. 436.105 of this 
chapter, preparing the sample for assay as follows: Place an accurately 
weighed representative portion of the sample into a separatory funnel 
containing approximately 50 milliliters of peroxide-free ether. Shake 
the sample and ether until homogeneous. Add 20 to 25 milliliters of 0.1M 
potassium phosphate buffer, pH 8.0 (solution 3), and shake well. Allow 
the layers to separate. Remove the buffer layer and repeat the 
extraction procedure with each of three more 20- to 25-milliliter 
quantities of solution 3. Combine the buffer extractives in a suitable 
volumetric flask and dilute to volume with solution 3. Remove an aliquot 
and further dilute with solution 3 to the reference concentration of 1.0 
microgram of neomycin per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[42 FR 27233, May 27, 1977, as amended at 50 FR 19920, May 13, 1985]



Sec. 448.510e  Bacitracin-neomycin sulfate-polymyxin B sulfate ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin-neomycin sulfate-polymyxin B 
sulfate ointment, in a suitable and harmless ointment base, contains in 
each gram the following:
    (i) 500 units of bacitracin, 3.5 milligrams of neomycin, and 5,000 
units of polymyxin B; or
    (ii) 400 units of bacitracin, 3.5 milligrams of neomycin, and 5,000 
units of polymyxin B.

It may contain a suitable local anesthetic. Its bacitracin content is 
satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of units of bacitracin that it is represented to 
contain. Its neomycin content is satisfactory if it is not less than 90 
percent and not more than 130 percent of the number of milligrams of 
neomycin that it is represented to contain. Its polymyxin B content is 
satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of units of polymyxin B that is represented to 
contain. Its moisture content is not more than 0.5 percent. The 
bacitracin used conforms to the standards prescribed by 
Sec. 448.10(a)(1). The neomycin sulfate used conforms to the standards 
prescribed by Sec. 444.42(a)(1) of this chapter. The polymyxin B sulfate 
used conforms to the standards prescribed by Sec. 448.30(a)(1).
    (2) Labeling. (i) On the label of the immediate container and on the 
outside wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each ingredient contained in the drug.
    (c) An expiration date that conforms to the requirements prescribed 
by Sec. 432.5(a)(3) of this chapter.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each request shall 
contain:
    (i) Results of tests and assays on:
    (a) The bacitracin used in making the batch for potency, loss on 
drying, pH, and identity.

[[Page 852]]

    (b) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (c) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (d) The batch for bacitracin content, neomycin content, polymyxin B 
content, and moisture.
    (ii) Samples required:
    (a) The bacitracin used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (b) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (c) The polymxin B sulfate used in making the batch: 10 packages, 
each containing approximately 1.0 gram.
    (d) The batch: A minimum of 7 immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Bacitracin content. 
Proceed as directed for bacitracin zinc in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Place an accurately weighed 
representative portion of the sample into a separatory funnel containing 
approximately 50 milliliters of peroxide-free ether. Shake the sample 
and ether until homogeneous. Add 20 to 25 milliliters of 1.0 percent 
potassium phosphate buffer, pH 6.0 (solution 1), and shake well. Allow 
the layers to separate. Remove the buffer layer and repeat the 
extraction procedure with each of three more 20- to 25-milliliter 
quantities of solution 1. Combine the buffer extractives in a suitable 
volumetric flask and dilute to volume with solution 1. Remove an 
aliquot, add sufficient hydrochloric acid so that the amount of acid in 
the final solution will be the same as in the reference concentration of 
the working standard and further dilute with solution 1 to the reference 
concentration of 1.0 unit of bacitracin per milliliter (estimated).
    (ii) Neomycin content. Proceed as directed in Sec. 436.105 of this 
chapter, preparing the sample for assay as follows: Place an accurately 
weighed representative portion of the sample into a separatory funnel 
containing approximately 50 milliliters of peroxide-free ether. Shake 
the sample and ether until homogeneous. Add 20 to 25 milliliters of 0.1M 
potassium phosphate buffer, pH 8.0 (solution 3), and shake well. Allow 
the layers to separate. Remove the buffer layer and repeat the 
extraction procedure with each of three more 20- to 25-milliliter 
quantities of solution 3. Combine the buffer extractives in a suitable 
volumetric flask and dilute to volume with solution 3. Remove an aliquot 
and further dilute with solution 3 to the reference concentration of 1.0 
microgram of neomycin per milliliter (estimated).
    (iii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, except add to each concentration of the polymyxin B 
standard response line a quantity of neomycin to yield the same 
concentration of neomycin as that present when the sample is diluted to 
contain 10 units of polymyxin B per milliliter. Prepare the sample for 
assay as follows: Place an accurately weighed representative portion of 
the sample into a separatory funnel containing approximately 50 
milliliters of peroxide-free ether. Shake the sample and ether until 
homogeneous. Add 20 to 25 milliliters of 10 percent potassium phosphate 
buffer, pH 6.0 (solution 6), and shake well. Allow the layers to 
separate. Remove the buffer layer and repeat the extraction procedure 
with each of three more 20- to 25-milliliter quantities of solution 6. 
Combine the buffer extractives in a suitable volumetric flask and dilute 
to volume with solution 6. Remove an aliquot and further dilute with 
solution 6 to the reference concentration of 10 units of polymyxin B per 
milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[42 FR 27233, May 27, 1977, as amended at 50 FR 19920, May 13, 1985]



Sec. 448.510f  Bacitracin-polymyxin B sulfate topical aerosol.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin-polymyxin B sulfate topical 
aerosol is bacitracin and polymyxin B sulfate in a suitable and harmless 
vehicle, packaged in a pressurized container with a suitable and 
harmless inert gas. Each gram contains 500 units of bacitracin and 5,000 
units of polymyxin B. It may contain a suitable local anesthetic. Its 
bacitracin content is satisfactory if it is not less than 90 percent and 
not more than 130 percent

[[Page 853]]

of the number of units of bacitracin that it is represented to contain. 
Its polymyxin B content is satisfactory if it is not less than 90 
percent and not more than 130 percent of the number of units of 
polymyxin B that it is represented to contain. Its moisture content is 
not more than 0.5 percent. The bacitracin used conforms to the standards 
prescribed by Sec. 448.10(a)(1). The polymyxin B sulfate used conforms 
to the standards prescribed by Sec. 448.30(a)(1).
    (2) Labeling. (i) On the label of the immediate container and on the 
outside wrapper or container, if any:
    (a) The batch mark;
    (b) The name and quantity of each active ingredient contained in the 
drug; and
    (c) An expiration date that conforms to the requirements prescribed 
by Sec. 432.5(a)(3) of this chapter.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacitracin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The polymyxin B sulfate used in making the batch for potentcy, 
loss on drying, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The bacitracin used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing 1.0 gram.
    (c) The batch: A minimum of 12 immediate containers.
    (b) Tests and methods of assay. The container must remain inverted 
throughout the sampling procedure. Freeze the container overnight at -70 
 deg.C. Remove from the freezer and puncture the container to allow the 
propellant to dissipate. Open the container, mix well, and proceed as 
described in paragraphs (b) (1) and (2) of this section.
    (1) Potency--(i) Bacitracin content. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the sample from 
the container into a separatory funnel containing approximately 50 
milliliters of peroxide-free ether. Shake the sample and ether until 
homogeneous. Add 20 to 25 milliliters of 1.0 percent potassium phosphate 
buffer, pH 6.0 (solution 1), and shake well. Allow the layers to 
separate. Remove the buffer layer and repeat the extraction procedure 
with each of three more 20- to 25-milliliter quantities of solution 1. 
Combine the buffer extractives in a suitable volumetric flask and dilute 
to volume with solution 1. Remove an aliquot, add sufficient 
hydrochloric acid so that the amount of acid in the final solution will 
be the same as in the reference concentration of the working standard, 
and further dilute with solution 1 to the reference concentration of 1.0 
unit of bacitracin per milliliter (estimated).
    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, preparing the sample for assay as follows: Place an 
accurately weighed portion of the sample from the container into a 
separatory funnel containing approximately 50 milliliters of peroxide-
free ether. Shake the sample and ether until homogeneous. Add 20 to 25 
milliliters of 10 percent potassium phosphate buffer, pH 6.0 (solution 
6), and shake well. Allow the layers to separate. Remove the buffer 
layer and repeat the extraction procedure with each of three more 20- to 
25-milliliter quantities of solution 6. Combine the buffer extractives 
in a suitable volumetric flask and dilute to volume with solution 6. 
Remove an aliquot and further dilute with solution 6 to the reference 
concentration of 10 units of polymyxin B per milliliter (estimated).
    (2) Moisture. Proceed as directed Sec. 436.201 of this chapter, 
using the titration procedure and calculation in paragraph (e)(3) of 
that section and 1- to 2-milliliter portions of the sample from the 
container.
[51 FR 35212, Oct. 2, 1986, as amended at 55 FR 9722, Mar. 15, 1990; 55 
FR 11584, Mar. 29, 1990]

[[Page 854]]



Sec. 448.513  Bacitracin zinc dermatologic dossage forms.



Sec. 448.513a  Bacitracin zinc-polymyxin B sulfate ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin zinc-polymyxin B sulfate 
ointment contains bacitracin zinc and polymyxin B sulfate in a suitable 
and harmless ointment base. It may contain a suitable local anesthetic. 
Each gram contains 500 units of bacitracin and 10,000 units of polymyxin 
B. Its bacitracin content is satisfactory if it is not less than 90 
percent and not more than 130 percent of the number of units of 
bacitracin that it is represented to contain. Its polymyxin B content is 
satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of units of polymyxin B that it is represented to 
contain. Its moisture content is not more than 0.5 percent. The 
bacitracin zinc used conforms to the standards prescribed by 
Sec. 448.13(a)(1). The polymyxin B sulfate used conforms to the 
standards prescribed by Sec. 448.30(a)(1).
    (2) Labeling. (i) On the label of the immediate container and on the 
outside wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (c) An expiration date that conforms to the requirements prescribed 
by Sec. 432.5(a)(3) of this chapter.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacitracin zinc used in making the batch for potency, loss 
on drying, pH, zinc content, and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (c) The batch for bacitracin content, polymyxin B content, and 
moisture.
    (ii) Samples required:
    (a) The bacitracin zinc used in making the batch: 10 packages, each 
containing equal portions of approximately 1.0 gram.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 1.0 gram.
    (c) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Bacitracin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows:
    (a) If the ointment is not water miscible. Place an accurately 
weighed representative portion of the sample into a separatory funnel 
containing approximately 50 milliliters of peroxide-free ether. Shake 
the sample and ether until homogeneous. Add 20 to 25 milliliters of 
0.01N hydrochloric acid and shake well. Allow the layers to separate. 
Remove the acid layer and repeat the extraction procedure with each of 
three more 20- to 25-milliliter quantities of 0.01N hydrochloric acid. 
Combine the acid extractives in a suitable volumetric flask and dilute 
to volume with 0.01N hydrochloric acid. (If the bacitracin content is 
less than 100 units per milliliter in 0.01N hydrochloric acid, add 
sufficient addition hydrochloric acid to each concentration of the 
standard response line so that each standard solution contains the same 
amount of acid as the 1.0 unit per milliliter sample solution.) Remove 
an aliquot and further dilute with 1.0 percent potassium phosphate 
buffer, pH 6.0 (solution 1), to the reference concentration of 1.0 unit 
of bacitracin per milliliter (estimated).
    (b) If the ointment is water miscible. Place an accurately weighed 
representative portion of the sample into a high-speed glass blender jar 
containing 1.0 milliliter polysorbate 80 and sufficient solution 1 to 
give a stock solution of convenient concentration. Blend for 3 to 5 
minutes. Remove an aliquot, add sufficient hydrochloric acid so that the 
amount of acid in the final solution will be the same as in the 
reference concentration of the working standard and further dilute with 
solution 1 to the reference concentration of 1.0 unit of bacitracin per 
milliliter (estimated).

[[Page 855]]

    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, preparing the sample for assay as follows:
    (a) If the ointment is not water miscible. Place an accurately 
weighed representative portion of the sample into a separatory funnel 
containing approximately 50 milliliters of peroxide-free ether. Shake 
the sample and ether until homogeneous. Add 20 to 25 milliliters of 10 
percent potassium phosphate buffer, pH 6.0 (solution 6), and shake well. 
Allow the layers to separate. Remove the buffer layer and repeat the 
extraction procedure with each of three more 20- to 25-milliliter 
quantities of solution 6. Combine the buffer extractives in a suitable 
volumetric flask and dilute to volume with solution 6. Remove an aliquot 
and further dilute with solution 6 to the reference concentration of 10 
units of polymyxin B per milliliter (estimated).
    (b) If the ointment is water miscible. Place an accurately weighed 
representative portion of the sample into a high speed glass blender jar 
containing 1.0 milliliter polysorbate 80 and sufficient 10 percent 
potassium phosphate buffer, pH 6.0 (solution 6), to give a stock 
solution of convenient concentration. Blend for 3 to 5 minutes. Remove 
an aliquot and further dilute with solution 6 to the reference 
concentration of 10 units of polymyxin B per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[42 FR 27234, May 27, 1977, as amended at 55 FR 50173, Dec. 5, 1990]



Sec. 448.513b  Bacitracin zinc-neomycin sulfate ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin zinc-neomycin sulfate ointment 
contains bacitracin zinc and neomycin sulfate in a suitable and harmless 
ointment base. Each gram contains 500 units of bacitracin and 3.5 
miligrams of neomycin. Its bacitracin content is satisfactory if it is 
not less than 90 percent and not more than 130 percent of the number of 
units of bacitracin that it is represented to contain. Its neomycin 
content is satisfactory if it is not less than 90 percent and not more 
than 130 percent of the number of milligrams of neomycin that it is 
represented to contain. Its moisture content is not more than 0.5 
percent. The bacitracin zinc used conforms to the standards prescribed 
by Sec. 448.13(a)(1). The neomycin sulfate used conforms to the 
standards prescribed by Sec. 444.42(a)(1) of this chapter.
    (2) Labeling--(i) On the label of the immediate container and on the 
outside wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (c) An expiration date that conforms to the requirements prescribed 
by Sec. 432.5(a)(3) of this chapter.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacitracin zinc used in making the batch for potency, loss 
on drying, pH, zinc content, and identity.
    (b) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (c) The batch for bacitracin content, neomycin content, and 
moisture.
    (ii) Samples required:
    (a) The bacitracin zinc used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (b) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (c) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Bacitracin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows:
    (a) If the ointment is not water miscible. Place an accurately 
weighed representative portion of the sample into a separatory funnel 
containing approximately 50 milliliters of peroxide-free ether. Shake 
the sample and ether until homogeneous. Add 20 to 25 milliliters of 
0.01N hydrochloric acid and shake well. Allow the layers to separate. 
Remove the acid layer and repeat the extraction procedure with each of

[[Page 856]]

three more 20- to 25-milliliter quantities of 0.01N hydrochloric acid. 
Combine the acid extractives in a suitable volumetric flask and dilute 
to volume with 0.01N hydrochloric acid. (If the bacitracin content is 
less than 100 units per milliliter in 0.01N hydrochloric acid, add 
sufficient additional hydrochloric acid to each concentration of the 
standard response line so that each standard solution will have the same 
amount of acid as the final sample solution.) Remove an aliquot and 
further dilute with 1 percent potassium phosphate buffer, pH 6.0 
(solution 1), to the reference concentration of 1.0 unit of bacitracin 
per milliliter (estimated).
    (b) If the ointment is water miscible. Place an accurately weighed 
representative portion of the sample into a high-speed glass blender jar 
containing 1.0 milliliter polysorbate 80 and sufficient 1 percent 
potassium phosphate buffer, pH 6.0 (solution 1), to give a stock 
solution of convenient concentration. Blend for 3 to 5 minutes. Remove 
an aliquot, add sufficient hydrochloric acid so that the amount of acid 
in the final solution will be the same as in the reference concentration 
of the working standard and further dilute with solution 1 to the 
reference concentration of 1.0 unit of bacitracin per milliliter 
(estimated).
    (ii) Neomycin content. Proceed as directed in Sec. 436.105 of this 
chapter, preparing the sample for assay as follows:
    (a) If the ointment is not water miscible. Place an accurately 
weighed representative portion of the sample into a separatory funnel 
containing approximately 50 milliliters of peroxide-free ether. Shake 
the sample and ether until homogeneous. Add 20 to 25 milliliters of 0.1M 
potassium phosphate buffer, pH 8.0 (solution 3), and shake well. Allow 
the layers to separate. Remove the buffer layer and repeat the 
extraction procedure with each of three more 20- to 25-milliliter 
quantities of solution 3. Combine the buffer extractives in a suitable 
volumetric flask and dilute to volume with solution 3. Remove an aliquot 
and further dilute with solution 3 to the reference concentration of 1.0 
microgram of neomycin per milliliter (estimated).
    (b) If the ointment is water miscible. Place an accurately weighed 
representative portion of the sample into a high-speed glass blender jar 
containing 1.0 milliliter polysorbate 80 and sufficient 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), to give a stock solution of 
convenient concentration. Blend for 3 to 5 minutes. Remove an aliquot 
and further dilute with solution 3 to the reference concentration of 1.0 
microgram of neomycin per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[42 FR 27234, May 27, 1977, as amended at 50 FR 19920, May 13, 1985]



Sec. 448.513c  Bacitracin zinc-neomycin sulfate-polymyxin B sulfate ointment; bacitracin zinc-neomycin sulfate-polymyxin B sulfate hydrocortisone ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. This drug, in a suitable and harmless 
ointment base, contains in each gram the following:
    (i) 400 units of bacitracin, 3 milligrams of neomycin, and 8,000 
units of polymyxin B; or
    (ii) 400 units of bacitracin, 3.5 milligrams of neomycin, and 5,000 
units of polymyxin B, with or without 10 milligrams of hydrocortisone; 
or
    (iii) 500 units of bacitracin, 3.5 milligrams of neomycin, and 5,000 
units of polymyxin B; or
    (iv) 500 units of bacitracin, 3.5 milligrams of neomycin, and 10,000 
units of polymyxin B.

It may contain a suitable local anesthetic except for combinations in 
paragraph (a)(1)(ii) of this section that contain hydrocortisone. Its 
bacitracin content is satisfactory if it is not less than 90 percent and 
not more than 130 percent of the number of units of bacitracin that it 
is represented to contain. Its neomycin content is satisfactory if it is 
not less than 90 percent and not more than 130 percent of the number of 
milligrams of neomycin that it is represented to contain. Its polymyxin 
B content is satisfactory if it is not less than 90 percent and not more 
than 130 percent of the number of units of polymyxin B that it is 
represented to contain. Its moisture content is not more

[[Page 857]]

than 0.5 percent. The bacitracin zinc used conforms to the standards 
prescribed by Sec. 448.13(a)(1). The neomycin sulfate used conforms to 
the standards prescribed by Sec. 444.42(a)(1) of this chapter. The 
polymyxin B sulfate used conforms to the standards prescribed by 
Sec. 448.30(a)(1).
    (2) Labeling. If it contains a steroid, it shall be labeled in 
accordance with the requirements of Sec. 432.5 of this chapter. If it 
does not contain a steroid, each package shall bear on its label or 
labeling, as hereinafter indicated, the following:
    (i) The batch mark.
    (ii) The name and quantity of each active ingredient contained in 
the drug.
    (iii) An expiration date that conforms to the requirements 
prescribed by Sec. 432.5(a)(3) of this chapter.
    (iv) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacitracin zinc used in making the batch for potency, loss 
on drying, pH, zinc content, and identity.
    (b) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (c) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (d) The batch for bacitracin content, neomycin content, polymyxin B 
content, and moisture.
    (ii) Samples required:
    (a) The bacitracin zinc used in making the batch: 10 packages, each 
containing 1.0 gram.
    (b) The neomycin sulfate used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (c) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 1.0 gram.
    (d) The batch: A minimum of 7 immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Bacitracin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows:
    (a) If the ointment is not water miscible. Place an accurately 
weighed representative portion of the sample into a separatory funnel 
containing approximately 50 milliliters of peroxide-free ether. Shake 
the sample and ether until homogeneous. Add 20 to 25 milliliters of 
0.01N hydrochloric acid and shake well. Allow the layers to separate. 
Remove the acid layer and repeat the extraction procedure with each of 
three more 20- to 25-milliliter quantities of 0.01N hydrochloric acid. 
Combine the acid extractives in a suitable volumetric flask and dilute 
to volume with 0.01N hydrochloric acid. (If the bacitracin content is 
less than 100 units per milliliter in 0.01N hydrochloric acid, add 
sufficient additional hydrochloric acid to each concentration of the 
standard response line so that each standard solution contains the same 
amount of acid as the 1.0 unit per milliliter sample solution.) Remove 
an aliquot and further dilute with 1.0 percent potassium phosphate 
buffer, pH 6.0 (solution 1), to the reference concentration of 1.0 unit 
of bacitracin per milliliter (estimated).
    (b) If the ointment is water miscible. Place an accurately weighed 
representative portion of the sample into a high-speed glass blender jar 
containing 1.0 milliliter polysorbate 80 and sufficient 1 percent 
potassium phosphate buffer, pH 6.0 (solution 1) to give a stock solution 
of convenient concentration. Blend for 3 to 5 minutes. Remove an 
aliquot, add sufficient hydrochloric acid so that the amount of acid in 
the final solution will be the same as in the reference concentration of 
the working standard and further dilute with solution 1 to the reference 
concentration of 1.0 unit of bacitracin per milliliter (estimated).
    (ii) Neomycin content. Proceed as directed in Sec. 436.105 of this 
chapter, preparing the sample for assay as follows:
    (a) If the ointment is not water miscible. Place an accurately 
weighed representative portion of the sample into a separatory funnel 
containing approximately 50 milliliters of peroxide-free ether. Shake 
the sample and ether until homogeneous. Add 20 to 25 milliliters of 0.1M 
potassium phosphate

[[Page 858]]

buffer, pH 8.0 (solution 3), and shake well. Allow the layers to 
separate. Remove the buffer layer and repeat the extraction procedure 
with each of three more 20- to 25- milliliter quantities of solution 3. 
Combine the buffer extractives in a suitable volumetric flask and dilute 
to volume with solution 3. Remove an aliquot and further dilute with 
solution 3 to the reference concentration of 1.0 microgram of neomycin 
per milliliter (estimated).
    (b) If the ointment is water miscible. Place an accurately weighed 
representative portion of the sample into a high-speed glass blender jar 
containing 1.0 milliliter of polysorbate 80 and sufficient 0.1M 
potassium phosphate buffer, pH 8.0 (solution 3), to give a stock 
solution of convenient concentration. Blend for 3 to 5 minutes. Remove 
an aliquot and further dilute with solution 3 to the reference 
concentration of 1.0 microgram of neomycin per milliliter (estimated).
    (iii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, except add to each concentration of the polymyxin B 
standard response line a quantity of neomycin to yield the same 
concentration of neomycin as that present when the sample is diluted to 
contain 10 units of polymyxin B per milliliter. Prepare the sample for 
assay as follows:
    (a) If the ointment is not water miscible. Place an accurately 
weighed representative portion of the sample into a separatory funnel 
containing approximately 50 milliliters of peroxide-free ether. Shake 
the sample and ether until homogeneous. Add 20 to 25 milliliters of 10 
percent potassium phosphate buffer, pH 6.0 (solution 6), and shake well. 
Allow the layers to separate. Remove the buffer layer and repeat the 
extraction procedure with each of three more 20- to 25-milliliter 
quantities of solution 6. Combine the buffer extractives in a suitable 
volumetric flask and dilute to volume with solution 6. Remove an aliquot 
and further dilute with solution 6 to the reference concentration of 10 
units of polymyxin B per milliliter (estimated).
    (b) If the ointment is water miscible. Place an accurately weighed 
representative portion of the sample into a high-speed glass blender jar 
containing 1.0 milliliter polysorbate 80 and sufficient 10 percent 
potassium phosphate buffer, pH 6.0 (solution 6), to give a stock 
solution of convenient concentration. Blend for 3 to 5 minutes. Remove 
an aliquot and further dilute with solution 6 to the reference 
concentration of 10 units of polymyxin B per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[42 FR 27235, May 27, 1977, as amended at 55 FR 50173, Dec. 5, 1990]



Sec. 448.513d  Bacitracin zinc-polymyxin B sulfate topical powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin zinc-polymyxin B sulfate 
topical powder contains bacitracin zinc and polymyxin B sulfate in a 
suitable and harmless base. Each gram contains 500 units of bacitracin 
and 10,000 units of polymyxin B. Its bacitracin content is satisfactory 
if it is not less than 90 percent and not more than 120 percent of the 
number of units of bacitracin that it is represented to contain. Its 
polymyxin B content is satisfactory if it is not less than 90 percent 
and not more than 120 percent of the number of units of polymyxin B that 
it is represented to contain. Its moisture content is not more than 7.0 
percent. It contains not more than an average of 10 microoganisms per 
gram. The bacitracin zinc used conforms to the standards prescribed by 
Sec. 448.13(a)(1). The polymyxin B sulfate used conforms to the 
standards prescribed by Sec. 448.30(a)(1).
    (2) Labeling--(i) On the label of the immediate container and on the 
outside wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (c) An expiration date that conforms to the requirements prescribed 
by Sec. 432.5(a)(3) of this chapter.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.

[[Page 859]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of test and assays on:
    (a) The bacitracin zinc used in making the batch for potency, loss 
on drying, pH, zinc content, and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (c) The batch for bacitracin content, polymyxin B content, moisture, 
and a microorganism count.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The bacitracin zinc used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 1.0 gram.
    (c) The batch: A minimum of 12 immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Bacitracin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Wash an accurately weighed sample (usually 
2 grams) into a 100-milliliter volumetric flask with 0.01N hydrochloric 
acid. Dilute to volume with 0.01N hydrochloric acid. Further dilute an 
aliquot with solution 1 to the reference concentration of 1.0 unit of 
bacitracin per milliliter (estimated).

    Note: The final sample solution must contain the same amount of 
hydrochloric acid as the reference concentration of the working 
standard.

    (ii) Polymyxin B content. Proceed as directed in Sec. 436.105 of 
this chapter, preparing the sample for assay as follows: Dissolve an 
accurately weighed representative portion of the sample (usually 1 gram) 
in 20 milliliters of sterile distilled water. Wash into an appropriate-
sized volumetric flask with 10 percent potassium phosphate buffer, pH 
6.0 (solution 6). Further dilute with solution 6 to the reference 
concentration of 10 units of polymyxin B per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Microorganism count--(i) Conduct of test for bacteria. Using 
approximately 200 milligrams of powder from each of five separate 
immediate containers, proceed as directed in Sec. 436.20(e)(1) of this 
chapter, except after the three washings transfer the entire filter 
membrane to the surface of medium N as described in Sec. 436.20(c)(14) 
of this chapter. Incubate the plate for 7 days at 30 deg. C. to 32 deg. 
C. Count the number of colonies appearing on the filter pad and 
calculate therefrom the number of viable microorganisms per gram of 
powder.
    (ii) Conduct of test for molds and yeasts. Proceed as directed in 
Sec. 436.20(e)(1) of this chapter, using approximately 200 milligrams 
from each of the five containers tested, except transfer the entire 
filter membrane to the surface of medium N as described in 
Sec. 436.20(c)(14) of this chapter, and incubate at 22 deg. C. to 
25 deg. C. for 7 days. Count the number of colonies appearing on the 
filter pad and calculate therefrom the number of viable microorganisms 
per gram of powder.
[42 FR 27236, May 27, 1977, as amended at 50 FR 15110, Apr. 17, 1985; 55 
FR 11584, Mar. 29, 1990]



Sec. 448.513e  Bacitracin zinc-polymyxin B sulfate topical aerosol.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin zinc-polymyxin B sulfate 
topical aerosol is bacitarcin zinc, polymyxin B sulfate in a suitable 
and harmless vehicle, packaged in a pressurized container with suitable 
and harmless inert gases. Each gram contains 120 units of bacitracin and 
2,350 units of polymyxin B. Its bacitracin content is satisfactory if it 
is not less than 90 percent and not more than 130 percent of the number 
of units of bacitracin that it is represented to contain. Its polymyxin 
B content is satisfactory if it is not less than 90 percent and not more 
than 130 percent of the number of units of polymyxin B that it is 
represented to contain. Its moisture content is not more than 0.5 
percent. It contains not more than an average of 10 microorganisms per 
container. The bacitracin zinc used conforms to the standards prescribed 
by Sec. 448.13(a)(1). The polymyxin B sulfate used conforms to the 
standards prescribed by Sec. 448.30(a)(1).

[[Page 860]]

    (2) Labeling. (i) On the label of the immediate container and on the 
outside wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (c) An expiration date that conforms to the requirements prescribed 
by Sec. 432.5(a)(3) of this chapter.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacitracin zinc used in making the batch for potency, loss 
on drying, pH, zinc content, and identity.
    (b) The polymyxin B sulfate used in making the batch for potency, 
loss on drying, pH, and identity.
    (c) The batch for bacitracin content, polymyxin B content, moisture, 
and a microorganism count.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The bacitracin zinc used in making the batch: 10 packages, each 
containing approximately 1.0 gram.
    (b) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 1.0 gram.
    (c) The batch: A minimum of 12 immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Sample preparation. 
Spray, as directed in the labeling, the entire contents of each 
container to be tested into a separate 2-liter Erlenmeyer flask, held in 
a horizontal position. Add 500 milliliters of 0.01N hydrochloric acid 
and shake to dissolve the contents. Immediately remove aliquots of this 
sample solution and proceed as directed paragraph (b)(1)(i)(a) and (b) 
of this section for each antibiotic to be tested.
    (a) Bacitracin content. Proceed as directed in Sec. 436.105 of this 
chapter, diluting an aliquot of the sample solution with 1 percent 
potassium phosphate buffer, pH 6.0 (solution 1), to the reference 
concentration of 1.0 unit of bacitracin per milliliter (estimated).

    Note: The final sample solution must contain the same amount of 
hydrochloric acid as the reference concentration of the working 
standard.

    (b) Polymyxin B content. Proceed as directed in Sec. 436.105 of this 
chapter, diluting an aliquot of the sample solution with 10 percent 
potassium phosphate buffer, pH 6.0 (solution 6), to the reference 
concentration of 10.0 units of polymyxin B per milliliter (estimated).
    (ii) [Reserved]
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Microorganism count--(i) Conduct of test for bacteria. 
Thoroughly cleanse the valve of each container to be tested with a 
suitable disinfectant. Into an empty, sterile Erlenmeyer flask, 
stoppered with a cotton plug, spray about one-half of the contents of 
each of five separate immediate containers by removing the cotton plug 
temporarily and using aseptic technique. Allow the propellant to 
evaporate. To the dry residue, which should not exceed 1 gram, add 500 
milliliters of diluting fluid C as described in Sec. 436.20(d)(3) of 
this chapter. Stopper the flask and swirl to dissolve the drug. As soon 
as the sample has completely dissolved, proceed as directed in 
Sec. 436.20(e)(1)(ii) of this chapter, except after the three washings 
transfer the entire filter membrane to the surface of medium N as 
described in Sec. 436.20(c)(14) of this chapter. Incubate the plate for 
7 days at 30 deg. C. to 32 deg. C. Count the number of colonies 
appearing on the filter pad and calculate therefrom the number of viable 
microorganisms per gram of powder.
    (ii) Conduct of test for molds and yeasts. Proceed as directed in 
paragraph (b)(3)(i) of this section, except transfer the entire filter 
membrane to the surface of medium N as described in Sec. 436.20(c)(14) 
of this chapter, and incubate at 25 deg. C. for 7 days.
[42 FR 27237, May 27, 1977, as amended at 50 FR 15110, Apr. 17, 1985; 55 
FR 11584, Mar. 29, 1990; 55 FR 40381, Oct. 3, 1990]



Sec. 448.513f  Bacitracin zinc ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality,

[[Page 861]]

and purity. Bacitracin zinc ointment is composed of 500 units of 
bacitracin zinc per gram in a suitable ointment base. Its potency is 
satisfactory if it is not less than 90 percent and not more than 140 
percent of the number of units of bacitracin that it is represented to 
contain. Its moisture content is not more than 0.5 percent. The 
bacitracin zinc used conforms to the standards prescribed by 
Sec. 448.13(a)(1).
    (2) Labeling--(i) On the label of the immediate container and on the 
outside wrapper or container, if any:
    (a) The batch mark.
    (b) The name and quantity of each active ingredient contained in the 
drug.
    (c) An expiration date that conforms to the requirements prescribed 
by Sec. 432.5(a)(3) of this chapter.
    (ii) On the label of the immediate container or other labeling 
attached to or within the package, adequate directions under which the 
layman can use the drug safely and efficaciously.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacitracin zinc used in making the batch for potency, loss 
on drying, pH, zinc content, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The bacitracin zinc used in making the batch: 10 packages, each 
containing 1.0 gram.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the sample into a 
separtory funnel containing approximately 50 milliliters of peroxide-
free ether. Shake the sample and ether until homogeneous. Add 20 to 25 
milliliters of 0.01N hydrochloric acid and shake well. Allow the layers 
to separate. Remove the aid layer and repeat the extraction procedure 
with each of three more 20- to 25-milliliter quantities of 0.01N 
hydrochloric acid. Combine the acid extractives in a suitable volumetric 
flask and dilute to volume with 0.01N hydrochloric acid. (If the 
bacitracin content is less than 100 units per milliliter in 0.01N 
hydrochloric acid, add sufficient additional hydrochloic acid to each 
concentration of the standard response line so that each standard 
solution contains the same amout of acid as the 1.0 unit per milliliter 
sample solution.) Remove an aliquot and further dilute with 1.0 percent 
potassium phosphate buffer, pH 6.0 (solution 1), to the reference 
concentration of 1.0 unit of bacitracin per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[42 FR 27237, May 27, 1977, as amended at 50 FR 19920, May 13, 1985]



               Subpart G--Vaginal Dosage Forms  [Reserved]



                        Subparts H-I  [Reserved]



                  Subpart J--Certain Other Dosage Forms



Sec. 448.910  Bacitracin for prescription compounding.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin for prescription compounding 
is a white to brown, neutral, water-soluble polypeptide intended for use 
in the extemporaneous compounding of prescriptions by practicing 
pharmacists. It is so purified and dried that:
    (i) Its potency is not less than 40 units of bacitracin per 
milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 5.0 percent.
    (iv) Its pH in an aqueous solution containing 10,000 units per 
millilter is not less than 5.5 and not more than 7.5.
    (v) It passes the identity test.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass, and it shall be a tight container as defined by the 
United States Pharmacopeia (U.S.P.). It shall be so sealed that the 
contents cannot be used without destroying such seal. Each such 
container shall contain 500,000 or 5 million units of bacitracin.
    (3) Labeling. Each package shall bear on its outside wrapper or 
container and

[[Page 862]]

on the immediate container the following:
    (i) The statement ``Caution: Federal law prohibits dispensing 
without prescription''.
    (ii) The statement ``Not sterile''.
    (iii) The batch mark.
    (iv) The number of units of bacitracin activity in each milligram 
and the number of grams of bacitracin in the immediate container.
    (v) The statement ``Expiration date ------------'', the blank being 
filled in with the date that is 12 months after the month during which 
the batch was certified, unless the use of a longer dating period has 
been approved in accordance with Sec. 432.5(a)(3) of this chapter.
    (vi) The statement ``The potency of this drug cannot be assured for 
longer than 60 days after the container is first opened for compounding 
a prescription''.
    (vii) The statements, ``For use only in extemporaneous prescription 
compounding. Not for manufacturing use''.
    (4) Requests for certification; samples. In addition to the 
requirements of Sec. 432.1 of this chapter, each request shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, and identity.
    (ii) Samples required: A 0.5-gram portion for each 5,000 packages in 
the batch, but in no case less than 10 such portions. Each such portion 
shall be collected at such intervals throughout the entire time of 
packaging the batch that the quantities packaged during the intervals 
are approximately equal.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed for 
bacitracin zinc in Sec. 436.105 of this chapter, preparing the sample 
for assay as follows: Dissolve an accurately weighed sample in 
sufficient 1.0 percent potassium phosphate buffer, pH 6.0 (solution 1), 
to obtain a stock solution of convenient concentration. Remove an 
aliquot of the stock solution, add sufficient hydrochloric acid so that 
the amount of acid in the final solution will be the same as in the 
reference concentration of the working standard and further dilute with 
solution 1 to the reference concentration of 1.0 unit of bacitracin per 
milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10,000 units per milliliter.
    (5) Identity. Proceed as directed in Sec. 436.319 of this chapter.
[42 FR 27238, May 27, 1977, as amended at 50 FR 19920, May 13, 1985]



Sec. 448.913  Bacitracin zinc for prescription compounding.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin zinc for prescription 
compounding is the zinc salt of a kind of bacitracin or a mixture of two 
or more such salts intended for use in the extemporaneous compounding of 
prescriptions by practicing pharmacists. It is so purified and dried 
that:
    (i) Its potency is not less than 40 units of bacitracin per 
milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 5.0 percent.
    (iv) Its pH in a saturated aqueous solution is not less than 6.0 and 
not more than 7.5.
    (v) Its zinc content is not more than 10 percent by weight on a 
moisture-free basis.
    (vi) It passes the identity test.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
United States Pharmacopeia (U.S.P.). It shall be so sealed that the 
contents cannot be used without destroying such seal. Each such 
container shall contain 500,000 or 5 million units of bacitracin.
    (3) Labeling. Each package shall bear on its outside wrapper or 
container and on the immediate container the following:
    (i) The statement ``Caution: Federal law prohibits dispensing 
without prescription''.
    (ii) The statement ``Not sterile''.
    (iii) The batch mark.
    (iv) The number of units of bacitracin activity in each milligram of 
the bacitracin zinc, and the number of grams of bacitracin zinc in the 
immediate container.

[[Page 863]]

    (v) The statement ``Expiration date ----------'', the blank being 
filled in with the date that is 12 months after the month during which 
the batch was certified, unless use of a longer dating period has been 
approved in accordance with the provisions of Sec. 432.5(a)(3) of this 
chapter.
    (vi) The statement, ``The potency of this drug cannot be assured for 
longer than 60 days after the container is first opened for compounding 
a prescription''.
    (vii) The statements, ``For use only in extemporaneous prescription 
compounding. Not for manufacturing use''.
    (4) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, zinc content, and identity.
    (ii) Samples required: A 0.5-gram portion for each 5,000 packages in 
the batch, but in no case less than 10 such portions. Each such portion 
shall be collected at such intervals throughout the entire time of 
packaging the batch that the quantities packaged during the intervals 
are approximately equal.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample (usually 25 to 35 milligrams) in 
sufficient 0.01N hydrochloric acid to give a bacitracin concentration of 
100 units per milliliter (estimated). Further dilute an aliquot with 
solution 1 to the reference concentration of 1.0 unit of bacitracin per 
milliliter (estimated).

    Note: The final sample solution must contain the same amount of 
hydrochloric acid as the reference concentration of the working 
standard.

    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
saturated solution (approximately 100 milligrams of the sample per 
milliliter).
    (5) Zinc content. Proceed as directed in Sec. 436.312 of this 
chapter.
    (6) Identity. Proceed as directed in Sec. 436.319 of this chapter.
[42 FR 27238, May 27, 1977, as amended at 50 FR 19920, May 13, 1985]



Sec. 448.930  Polymyxin B sulfate in certain other dosage forms.



Sec. 448.930a  Polymyxin B sulfate for prescription compounding.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Polymyxin B sulfate for prescription 
compounding is the sulfate salt of a kind of polymyxin or a mixture of 
two or more such salts intended for use in the extemporaneous 
compounding of prescriptions by practicing pharmacists. It is a white to 
buff-colored powder. It is so purified and dried that:
    (i) Its potency is not less than 6,000 units of polymyxin B per 
milligram, on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 7.0 percent.
    (iv) Its pH in an aqueous solution containing 5 milligrams per 
milliliter is not less than 5.0 and not more than 7.5.
    (v) Its residue on ignition is not more than 5 percent.
    (vi) It gives positive color identity tests for polymyxin.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. Each such container shall contain 100 million 
units of polymyxin B.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its outside wrapper or 
container and on the immediate container the following:
    (i) The statement ``Caution: Federal law prohibits dispensing 
without prescription''.
    (ii) The statement ``Not sterile''.
    (iii) The batch mark.
    (iv) The number of units of polymyxin B activity in each milligram 
of the polymyxin B sulfate and the number of grams of polymyxin B 
sulfate in the immediate container.

[[Page 864]]

    (v) The statement, ``The potency of this drug cannot be assured for 
longer than 60 days after the container is first opened for compounding 
a prescription''.
    (vi) The statements, ``For use only in extemporaneous prescription 
compounding. Not for manufacturing use''.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, residue on ignition, and identity.
    (ii) Samples required: A 0.5-gram portion for each 5,000 packages in 
the batch, but in no case less than 10 such portions. Each such portion 
shall be collected at such intervals throughout the entire time of 
packaging the batch that the quantities packaged during the intervals 
are approximately equal.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in 2 milliliters of sterile 
distilled water for each 5 milligrams of weighed sample. Further dilute 
an aliquot with sufficient 10 percent potassium phosphate buffer, pH 6.0 
(solution 6), to give a stock solution of convenient concentration. 
Further dilute an aliquot of the stock solution with solution 6 to the 
reference concentration of 10 units of polymyxin B per milliliter 
(estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 5 milligrams per milliliter.
    (5) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (6) Identity. (i) To a solution of 2 milligrams of polymyxin B 
sulfate in 5 milliliters of water, add 0.5 milliliter of 
triketohydrindene solution (1:1,000) and 2 drops of pyridine, boil for 1 
minute, and cool; a blue color develops; and
    (ii) To a solution of 2 milligrams of polymyxin B sulfate in 5 
milliliters of water, add 5 milliliters of sodium hydroxide solution 
(1:10), mix well, and add, dropwise, 5 drops of a cupric sulfate 
solution (1:100) mixing after the addition of each drop; a reddish-
violet color is produced.
[39 FR 19115, May 30, 1974, as amended at 46 FR 16684, Mar. 13, 1981; 50 
FR 19920, May 13, 1985]



Sec. 448.930b  Sterile polymyxin B sulfate-benzalkonium chloride urethral lubricant.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile polymyxin B sulfate-benzalkonium 
chloride urethral lubricant is polymyxin B sulfate and benzalkonium 
chloride, with one or more suitable and harmless suspending agents, in a 
suitable and harmless base. It contains, in each gram, 5,000 units of 
polymyxin B and 330 micrograms of benzalkonium chloride. Its content of 
polymyxin B is satisfactory if it contains not less than 90 percent and 
not more than 130 percent of the number of units of polymyxin B that it 
is represented to contain. It is sterile. Its pH is not less than 4.0 
and not more than 5.5. The polymyxin B sulfate used conforms to the 
standards prescribed by Sec. 448.30a(a)(1), except sterility, pyrogens, 
and heavy metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The polymyxin B sulfate used in making the batch for potency, 
pH, loss on drying, residue on ignition, and identity.
    (b) The batch for potency, sterility, and pH.
    (ii) Samples required:
    (a) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch.
    (1) For all tests except sterility: A minimum of five immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of

[[Page 865]]

this chapter, preparing the sample for assay as follows: Place an 
accurately weighed representative portion of the sample into a high-
speed glass blender jar containing 1.0 milliliter polysorbate 80 and 
sufficient 10 percent potassium phosphate buffer, pH 6.0 (solution 6), 
to obtain a stock solution of convenient concentration. Blend for 3 to 5 
minutes. Further dilute an aliquot of the stock solution with solution 6 
to the reference concentration of 10 units of polymyxin B per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20(e)(1) of this 
chapter, except dissolve the ointment as follows: Aseptically transfer a 
portion of 0.25 gram from each of 10 immediate containers of the drug to 
400 milliliters of diluting fluid D in an Erlenmeyer flask. Repeat the 
procedure on another 10 immediate containers. Swirl the flasks to 
dissolve the ointment.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[39 FR 19115, May 30, 1974, as amended at 46 FR 16684, Mar. 13, 1981; 50 
FR 19920, May 13, 1985]



PART 449--ANTIFUNGAL ANTIBIOTIC DRUGS--Table of Contents




                          Subpart A--Bulk Drugs

Sec.
449.1--449.3  [Reserved]
449.4  Amphotericin B.
449.4a  Amphotericin B for use in parenteral products.
449.10  Candicidin.
449.20  Griseofulvin.
449.40  Natamycin.
449.50  Nystatin.

                      Subpart B--Oral Dosage Forms

449.104  Amphotericin B oral suspension.
449.120  Griseofulvin oral dosage forms.
449.120a  Griseofulvin tablets.
449.120b  Griseofulvin capsules.
449.120c  Griseofulvin oral suspension.
449.120d  Griseofulvin (ultramicrosize) tablets.
449.150  Nystatin oral dosage forms.
449.150a  Nystatin tablets.
449.150b  Nystatin oral suspension.
449.150c  Nystatin for oral suspension.
449.150d  Nystatin pastilles.

                   Subpart C--Injectable Dosage Forms

449.204  Amphotericin B for injection.

                   Subpart D--Ophthalmic Dosage Forms

449.340  Natamycin ophthalmic suspension.

                          Subpart E  [Reserved]

                  Subpart F--Dermatologic Dosage Forms

449.504  Amphotericin B dermatologic dosage forms.
449.504a  Amphotericin B ointment.
449.504b  Amphotericin B cream.
449.504c  Amphotericin B lotion.
449.550  Nystatin dermatologic dosage forms.
449.550a  Nystatin ointment.
449.550b  Nystatin-iodochlorhydroxyquin ointment.
449.550c  Nystatin-neomycin sulfate-gramicidin-triamcinolone acetonide 
          ointment; nystatin-neomycin sulfate-gramicidin-fludrocortisone 
          acetate ointment.
449.550d  Nystatin cream.
449.550e  Nystatin-neomycin sulfate-gramicidin-triamcinolone acetonide 
          cream.
449.550f  Nystatin topical powder.
449.550g  Nystatin-neomycin sulfate-gramicidin topical powder.
449.550h  Nystatin lotion.

                     Subpart G--Vaginal Dosage Forms

449.610  Candicidin vaginal dosage forms.
449.610a  Candicidin vaginal ointment.
449.610b  Candicidin vaginal tablets.
449.610c  Candicidin vaginal capsules.
449.650  Nystatin vaginal dosage forms.
449.650a  Nystatin vaginal tablets.
449.650b  Nystatin vaginal suppositories.

    Authority:  21 U.S.C. 357.

    Source:  39 FR 19134, May 30, 1974, unless otherwise noted.



                          Subpart A--Bulk Drugs



Secs. 449.1--449.3  [Reserved]



Sec. 449.4  Amphotericin B.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amphotericin B is a yellow to golden-
orange powder. It is insoluble in water at pH. 6.0 to 7.0, anhydrous 
alcohols, esters, ethers, benzene, and toluene. It is soluble in 
dimethylformamide and dimethylsulfoxide. It is so purified and dried 
that:
    (i) Its potency is not less than 750 micrograms of amphotericin B 
per milligram on an anhydrous basis.

[[Page 866]]

    (ii) It contains not more than 15 percent of amphotericin A.
    (iii) [Reserved]
    (iv) Its loss on drying is not more than 5.0 percent.
    (v) It contains not more than 3.0 percent residue on ignition.
    (vi) It passes the identity test.
    (2) Labeling. In addition to the labeling prescribed by 
Sec. 432.5(b) of this chapter, each package shall bear on its label the 
statements ``Store below 10 deg. C.'' and ``Protect from light and 
moisture''.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of test and assays on the batch for potency, 
amphotericin A content, loss on drying, residue on iginition, and 
identity.
    (ii) Samples required on the batch: 10 packages, each containing not 
less than 500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient dimethylsulfoxide to 
give a stock solution of convenient concentration. Further dilute an 
aliquot with dimethylsulfoxide to a concentration of 20 micrograms of 
amphotericin B per milliliter (estimated). Remove an aliquot; dilute 
with 0.2M potassium phosphate buffer, pH 10.5 (solution 10), to the 
reference concentration of 1.0 microgram of amphotericin B per 
milliliter (estimated).
    (2) Amphotericin A content--(i) Amphotericin A. Dry approximately 20 
milligrams of the nystatin working standard as described in 
Sec. 436.200(a) of this chapter. Accurately weigh the dried working 
standard and quantitatively transfer into a 200-milliliter volumetric 
flask. Add exactly 40.0 milliliters of dimethylsulfoxide and dissolve. 
Make to mark with methyl alcohol and mix thoroughly. Pipette 4.0 
milliliters of this solution into a 50-milliliter volumetric flask. Add 
methyl alcohol to mark and mix thoroughly.
    (ii) Amphotericin B. Dry approximately 50 milligrams of the 
amphotericin B working standard as described in Sec. 436.200(a) of this 
chapter. Accurately weigh the dried working standard and quantitatively 
transfer into a 50-milliliter volumetric flask. Add 10 milliliters of 
dimethylsulfoxide and dissolve. Make to mark with methyl alcohol and mix 
thoroughly. Pipette 4.0 milliliters of this solution into a 50-
milliliter volumetric flask. Add methyl alcohol to mark and mix 
thoroughly.

The standard solution should be used for 1 day only.
    (iii) Sample. Accurately weigh about 50 milligrams of the sample to 
be tested and quantitatively transfer into a 50-milliliter volumetric 
flask. Add 10 milliliters of dimethylsulfoxide and dissolve. Make to 
mark with methyl alcohol and mix thoroughly. Pipette 4.0 milliliters of 
this solution into a 50-milliliter volumetric flask. Add methyl alcohol 
to mark and mix thoroughly.
    (iv) Blank. Pipette 10 milliliters of dimethylsulfoxide into a 50-
milliliter volumetric flask. Make to mark with methyl alcohol and mix. 
Pipette 4.0 milliliters of this solution into a 50-milliliter volumetric 
flask. Make to mark with methyl alcohol and mix thoroughly.
    (v) Procedure. Use a suitable ultraviolet spectrophotometer and 1-
centimeter silica cells. Adjust the instrument to zero with the blank 
solution. Measure the absorbances of the solutions of nystatin standard, 
amphotericin B standard, and the sample at 304 nanometers and at 282 
nanometers. Calculate the absorptivity of each standard at both 
wavelengths:
[GRAPHIC] [TIFF OMITTED] TR01JA93.268

where:

A=Absorptivity of nystatin standard at 282 nanometers;

[[Page 867]]

    B=Absorptivity of amphotericin B standard at 282 nanometers;
    a=Absorptivity of nystatin standard at 304 nanometers;
    b=Absorptivity of amphotericin B standard at 304 nanometers;
    S1=Absorbance of sample at 282 nanometers;
    S2=Absorbance of sample at 304 nanometers;
    Ws=Weight of sample in grams (on an anhydrous basis).

    (3) [Reserved]
    (4) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (5) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (6) Identity. Using the solutions prepared as described in 
paragraphs (b)(2) (ii), (iii), and (iv) of this section, record the 
absorption spectrum from 320 to 240 nanometers. Then dilute these 
solutions (1+9) with methyl alcohol and record the absorption spectrum 
from 400 to 320 nanometers. The sample exhibits absorption peaks at 
identical wavelengths with that of the amphotericin B standard. 
Depending on the amphotericin A content of the sample, a peak may occur 
at 304 nanometers.
[39 FR 19115, May 30, 1974, as amended at 46 FR 16684, Mar. 13, 1981; 49 
FR 2242, Jan. 19, 1984; 50 FR 19920, May 13, 1985]



Sec. 449.4a  Amphotericin B for use in parenteral products.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amphotericin B is a yellow to golden-
orange powder. It is insoluble in water at pH 6.0 to 7.0, anhydrous 
alcohols, esters, ethers, benzene, and toluene. It is soluble in 
dimethylformamide and dimethylsulfoxide. It is so purified and dried 
that:
    (i) Its potency is not less than 750 micrograms of amphotericin B 
per milligram on an anhydrous basis.
    (ii) It contains not more than 5 percent of amphotericin A.
    (iii) [Reserved]
    (iv) Its loss on drying is not more than 5.0 percent.
    (v) It contains not more than 0.5 percent residue on ignition.
    (vi) It passes the identity test.
    (2) Labeling. In addition to the labeling prescribed by 
Sec. 432.5(b) of this chapter, each package shall bear on its label the 
statements ``Store below 10 deg. C.'' and ``Protect from light and 
moisture''.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, 
amphotericin A content, loss on drying, residue on ignition, and 
identity.
    (ii) Samples required on the batch: 10 packages, each containing not 
less than 500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient dimethylsulfoxide to 
give a stock solution of convenient concentration. Further dilute with 
dimethylsulfoxide to give a concentration of 20 micrograms of 
amphotericin B per milliliter (estimated). Dilute an aliquot with 0.2M 
potassium phosphate buffer, pH 10.5 (solution 10), to the reference 
concentration of 1.0 microgram of amphotericin B per milliliter 
(estimated).
    (2) Amphotericin A content. Proceed as directed in Sec. 449.4(b)(2).
    (3) [Reserved]
    (4) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (5) Residue of ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (6) Identity. Proceed as directed in Sec. 449.4(b)(7).
[39 FR 19134, May 30, 1974, as amended at 49 FR 2243, Jan. 19, 1984; 50 
FR 19920, May 13, 1985]



Sec. 449.10  Candicidin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Candicidin is a brown to yellow powder. 
It is sparingly soluble in water; very slightly soluble in ethyl 
alcohol, butyl alcohol, and acetone. It is so purified and dried that:
    (i) Its potency is not less than 1,000 micrograms of candicidin per 
milligram on an anhydrous basis.
    (ii) Its loss on drying is not more than 4 percent.
    (iii) Its pH is not less than 8.0 nor more than 10.0 in a 1 percent 
aqueous suspension.

[[Page 868]]

    (iv) Its ultraviolet absorption spectrum is characteristic of a 
conjugated heptaene and is qualitatively the same as that of the 
candicidin working standard.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, and identity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve a portion of the sample in sufficient dimethylsulfoxide to 
yield an estimated concentration of 1,000 micrograms of candicidin 
activity per milliliter. Further dilute an aliquot with sterile 
distilled water to the reference concentration of 0.06 microgram of 
candicidin activity per milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
1 percent aqueous suspension.
    (4) Identity--(i) Preparation of aqueous alcohol solution. Prepare 
an aqueous alcohol solution by mixing 53 volumes of ethyl alcohol and 47 
volumes of water.
    (ii) Preparation of standard solution. Grind a small portion of the 
candicidin working standard to a fine powder with a mortar and pestle. 
Accurately weigh an amount equivalent to 20,000 micrograms of candicidin 
activity and transfer it to a 100-milliliter volumetric flask. Add about 
50 milliliters of the aqueous alcohol solution and shake to effect 
complete dissolution. Bring to volume with the aqueous alcohol solution 
and mix well. Transfer a 25-milliliter aliquot to a 100-milliliter 
volumetric flask and bring to volume with the aqueous alcohol solution. 
This solution contains 50 micrograms of candicidin activity per 
milliliter.
    (iii) Preparation of sample solution. Proceed as directed in 
paragraph (b)(4)(ii) of this section.
    (iv) Procedure. Using a suitable recording spectrophotometer, record 
the absorption spectra of the standard solution and the sample solution 
between the wavelengths of 330 and 410 nanometers with the aqueous 
alcohol solution as the reference solution. Compare the absorption 
spectra of the standard solution and the sample solution. They should 
exhibit absorption maxima and minima at the same wavelengths, which are 
approximately 342, 359, 378, and 397 nanometers for the maxima and 348, 
366, and 390 nanometers for the minima.
[39 FR 19134, May 30, 1974, as amended at 44 FR 30333, May 25, 1979; 49 
FR 2243, Jan. 19, 1984]



Sec. 449.20  Griseofulvin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Griseofulvin is a microsize, white to 
pale-cream compound with the following chemical name: 7-chloro-2',4,6-
trimethoxy-6'-methylspiro[benzofuran-2(3H),1'-[2]cyclohexene]-
3,4'-dione. It is so purified and dried that:
    (i) Its griseofulvin content is not less than 900 micrograms and not 
more than 1,050 micrograms of griseofulvin per milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 1.0 percent.
    (iv) Its melting point, after drying, is not less than 217 deg. C. 
and not more than 224 deg. C.
    (v) Its specific rotation in dimethylformamide at 25 deg. C. is not 
less than +348 deg. and not more than +364 deg..
    (vi) Its ultraviolet absorption spectrum in methyl alcohol compares 
qualitatively with that of the griseofulvin reference standard.
    (vii) Its residue on ignition is not more than 0.2 percent.
    (viii) Its heavy metals content is not more than 25 parts per 
million.
    (ix) Its specific surface area is not less than 1.3 and not more 
than 1.7 square meters per gram.
    (x) It is crystalline.

[[Page 869]]

    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for griseofulvin 
content, loss on drying, melting point, specific rotation, identity, 
residue on ignition, heavy metals, specific surface area, and 
crystallinity.
    (ii) Samples required: 10 packages, each containing not less than 1 
gram.
    (b) Tests and methods of assay--(1) Griseofulvin content (gas liquid 
chromatography). Proceed as directed in Sec. 436.321 of this chapter.
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) Melting point. Proceed as directed in Sec. 436.209 of this 
chapter.
    (5) Specific rotation. Accurately weigh approximately 250 milligrams 
of the sample in a 25-milliliter glass-stoppered volumetric flask and 
dissolve in about 15 milliliters of dimethylformamide. Bring to volume 
with dimethylformamide, stopper, and mix well. Proceed as directed in 
Sec. 436.210 of this chapter, using a 2.0-decimeter polarimeter tube.
    (6) Identity. Dissolve an accurately weighed portion of the sample 
and of the griseofulvin working standard and dissolve each in sufficient 
methyl alcohol to obtain a concentration of 10 micrograms of 
griseofulvin per milliliter and mix well. (The standard solution can be 
kept under refrigeration and used for up to 1 month.) Record the 
ultraviolet absorption spectrum of solutions of the sample and standard 
from 240 to 320 nanometers. The spectral curves shall be similar, and 
each shall have a maximum at 292plus-minus 2 nanometers and a 
minimum at 269plus-minus 2 nanometers.
    (7) Residue on ignition. Proceed as directed in Sec. 436.207 of this 
chapter.
    (8) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
    (9) Specific surface area--(i) Procedure. Determine the apparent 
particle size in microns by the air-permeation method, using a suitable 
subsieve sizer. Weigh 1.819 grams plus-minus0.001 gram of the 
sample and transfer to the compression tube of the apparatus. Compact 
the sample with moderate pressure so that it has a uniform porosity. 
Pass compressed dry air through the sample and measure the air pressure 
with a water manometer. Observe the porosity and calculate the apparent 
particle size from the instrument equation or read it from a chart that 
has been calculated in accordance with the equation. Repeat the readings 
at successively higher degrees of compaction until the apparent particle 
size reaches a minimum. Calculate the observed specific surface area 
(SSA) in square meters per gram of sample, as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.269

where F is a factor used to correct the apparent particle size to the 
true particle size:

                                                                        
------------------------------------------------------------------------
                        Porosity reading                            F   
------------------------------------------------------------------------
0.80...........................................................   1.3771
0.76...........................................................   1.4142
0.72...........................................................   1.4573
0.68...........................................................   1.5082
0.64...........................................................   1.5690
0.60...........................................................   1.6432
0.56...........................................................   1.7353
0.52...........................................................   1.8528
0.48...........................................................   2.0076
0.44...........................................................   2.2203
0.40...........................................................   2.5298
------------------------------------------------------------------------


    (ii) Standard. Determine the observed specific surface area of the 
griseofulvin specific surface area standard by the method prescribed in 
paragraph (b)(9)(i) of this section, using the same instrument and the 
same air pressure setting.

[[Page 870]]

    (iii) Calculations. Calculate the corrected specific surface area of 
the sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.270

    (10) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19134, May 30, 1974, as amended at 44 FR 20660, Apr. 6, 1979; 50 
FR 19920, May 13, 1985]



Sec. 449.40  Natamycin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Natamycin is 22-[(3 - amino - 3,6 - 
dideoxy --D-mannopyranosyl) - oxy] - 1,3,26-trihydroxy - 12-
methyl-10-oxo-6,11,28-trioxatricyclo [22.3.1.05,7] octacosa-
8,14,16,18,20 - pentaene-25-carboxylic acid. It is an off-white to cream 
colored powder which may contain up to 3 moles of water. It is 
practically insoluble in water, slightly soluble in methanol, and 
soluble in glacial acetic acid and dimethylformamide. It is so purified 
and dried that:
    (i) Its potency is not less than 900 micrograms of natamycin per 
milligram on an anhydrous basis.
    (ii) Its moisture content is not less than 6.0 percent and not more 
than 9.0 percent.
    (iii) Its pH in a 1 percent aqueous suspension is not less than 5.0 
and not more than 7.5.
    (iv) It passes the identity test.
    (v) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay. Dilute solutions of natamycin are 
very sensitive to light and should be kept in the dark as much as 
possible or substantial decomposition will take place.
    (1) Potency. Proceed as directed in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Dissolve an accurately 
weighed sample in dimethylsulfoxide and further dilute with sufficient 
dimethylsulfoxide to give a concentration of 100 micrograms of natamycin 
per milliliter (estimated). Further dilute with 0.2M potassium phosphate 
buffer, pH 10.5 (solution 10), to the reference concentration of 5.0 
micrograms of natamycin per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
1.0 percent aqueous suspension.
    (4) Identity. Accurately weigh approximately 50 milligrams of the 
sample into a 200-milliliter volumetric flask. Add approximately 5.0 
milliliters of distilled water, and completely moisten the sample. Then 
add approximately 100 milliliters of an acid-alcohol solvent (0.1 
percent glacial acetic acid in methyl alcohol) and stir or shake 
mechanically in the dark until solution is complete. Dilute to volume 
with the acid-alcohol solvent. Transfer 2.0 milliliters of this solution 
to a 100-milliliter volumetric flask and dilute to volume with the acid-
alcohol solvent. Using a suitable spectrophotometer with 1-centimeter 
cells and the acid-alcohol solvent as a blank, record the ultraviolet 
absorption spectrum from 215 to 330 nanometers. The spectrum compares 
qualitatively to that of the natamycin working standard similarly 
treated.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[43 FR 55384, Nov. 28, 1978, as amended at 46 FR 16684, Mar. 13, 1981]

[[Page 871]]



Sec. 449.50  Nystatin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nystatin is the yellow to light-tan 
compound of a kind of nystatin or a mixture of two or more such 
compounds. It is very slightly soluble in water, moderately soluble in 
methyl alcohol, butyl alcohol, or propyl alcohol. It is so purified and 
dried that:
    (i) Its potency is not less than 4,400 units of nystatin per 
milligram; except, if it is packaged for extemporaneous preparation of 
oral suspensions, its potency is not less than 5,000 units of nystatin 
per milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 5.0 percent.
    (iv) Its pH in a 3 percent aqueous suspension is not less than 6.5 
and not more than 8.0.
    (v) It passes the identity test.
    (vi) If it is packaged for extemporaneous preparation of oral 
suspensions, it passes the suspendibility test.
    (vii) If it is packaged for extemporaneous preparation of oral 
suspensions, it is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, and identity. In addition, if it is packaged for 
extemporaneous preparation of oral suspensions, results of tests and 
assays on the batch for suspendibility and crystallinity.
    (ii) Samples required on the batch: 10 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient dimethylformamide to 
give a nystatin concentration of 400 units per milliliter (estimated). 
Further dilute with 10 percent potassium phosphate buffer, pH 6.0 
(solution 6), to the reference concentration of 20 units of nystatin per 
milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
3 percent aqueous suspension of the drug.
    (5) Identity. Weigh approximately 100 milligrams of the sample into 
a 200-milliliter, glass-stoppered, volumetric flask. Add 50 milliliters 
of absolute methyl alcohol and 10 milliliters of glacial acetic acid. 
When the sample has dissolved, dilute to volume with methyl alcohol. 
Transfer 2 milliliters of this solution to a 100-milliliter volumetric 
flask and dilute to volume with methyl alcohol. Use the same dilution of 
acetic acid in methyl alcohol as the blank. Immediately determine the 
absorption peaks at 230, 291, 305, and 319 nanometers, and the shoulders 
at 279plus-minus2 nanometers, using a suitable ultraviolet 
spectrophotometer and quartz cells. Set the instrument to 100 percent 
transmission with the blank. If a recording spectrophotometer is used, 
record the ultraviolet absorption spectrum from 220 nanometers to 350 
nanometers. If a nonrecording spectrophotometer is used, the exact 
positions of the peaks and shoulder should be determined for the 
particular instrument used. The ratio of the two absorbances

                    (A230/A279)

should be not less than 0.90 and not more than 1.25.
    (6) Suspendibility test. Transfer 200 milligrams of the sample into 
a 250-milliliter beaker containing 200 milliliters of water. Swirl the 
suspension gently with a stirring rod. Allow the beaker to remain still 
for 2 minutes and observe the bottom. It passes the test if the powder 
remains in suspension. If a significant amount of sediment is observed, 
withdraw an accurately measured aliquot of the undisturbed suspension 
and assay as directed in Sec. 449.150c(b)(1) of this chapter. It passes 
the test if the suspension contains not less than 90 percent of the 
number of units of nystatin that it is represented to contain.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19134, May 30, 1974, as amended at 39 FR 43833, Dec. 19, 1974; 49 
FR 5098, Feb. 10, 1984; 50 FR 19920, May 13, 1985]

[[Page 872]]



                      Subpart B--Oral Dosage Forms



Sec. 449.104  Amphotericin B oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amphotericin B oral suspension is a 
mixture of amphotericin B with one or more suitable and harmless 
preservatives, colorings, sweetening ingredients, flavorings, buffer 
substances, lubricants, suspending agents, and sequestrants in an 
aqueous vehicle. Each milliliter contains 100 milligrams of amphotericin 
B. Its potency is satisfactory if it is not less than 90 percent and not 
more than 125 percent of the number of milligrams of amphotericin B that 
it is represented to contain. Its pH is not less than 4.5 and not more 
than 6.0. The amphotericin B conforms to the standards prescribed by 
Sec. 449.4(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The amphotericin B used in making the batch for potency, 
amphotericin A content, loss on drying, pH, residue on ignition, and 
identity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The amphotericin B used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place an accurately measured representative portion into a high-speed 
glass blender with sufficient dimethylsulfoxide to give a stock solution 
of convenient concentration. Blend for 3 to 5 minutes. Dilute an aliquot 
of the stock solution with dimethylsulfoxide to give a concentration of 
20 micrograms of amphotericin B per milliliter (estimated). Further 
dilute an aliquot with 0.2M potassium phosphate buffer, pH 10.5 
(solution 10), to the reference concentration of 1.0 microgram of 
amphotericin B per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted suspension.
[39 FR 19134, May 30, 1974, as amended at 50 FR 19920, May 13, 1985]



Sec. 449.120  Griseofulvin oral dosage forms.



Sec. 449.120a  Griseofulvin tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Griseofulvin tablets are tablets composed 
of griseofulvin, with or without one or more suitable fillers, 
colorings, lubricants, and binders. Each tablet contains 125, 250, or 
500 milligrams of griseofulvin. The griseofulvin content is satisfactory 
if it is not less than 90 percent and not more than 115 percent of the 
number of milligrams of griseofulvin that it is represented to contain. 
The loss on drying is not more than 5.0 percent. The tablets shall 
disintegrate within 1 hour. The griseofulvin used conforms to the 
standards prescribed by Sec. 449.20(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The griseofulvin used in making the batch for griseofulvin 
content, loss on drying, melting point, specific rotation, identity, 
residue on ignition, heavy metals, specific surface area, and 
crystallinity.
    (b) The batch for griseofulvin content, loss on drying, and 
disintegration time.
    (ii) Samples required:
    (a) The griseofulvin used in making the batch: 10 packages, each 
containing not less than 1 gram.
    (b) The batch for griseofulvin content, loss on drying, and 
disintegration time.
    (b) Tests and methods of assay--(1) Griseofulvin content (gas liquid 
chromatography). Proceed as directed in Sec. 436.321 of this chapter, 
except:
    (i) Prepare the sample solution as follows: Accurately weigh 20 
tablets

[[Page 873]]

and determine the average tablet weight. Grind the tablets to a fine 
powder in a mortar and transfer an accurately weighed sample to a 
volumetric flask of such size that for each 50 milliliters of volume 
there are 40 milligrams of griseofulvin (estimated). Add chloroform to 
about one-fourth volume of the flask. Swirl the flask and apply gentle 
heat to aid in dissolution of the griseofulvin. Allow the mixture to 
cool and then dilute to volume with chloroform and mix. Allow to settle 
and transfer 2.0 milliliters of the supernate to a conical centrifuge 
tube and evaporate to dryness under a current of dry air. Add 1.0 
milliliter of the internal standard solution to the centrifuge tube and 
mix vigorously to obtain a uniform solution; and,
    (ii) Calculate the milligrams of griseofulvin per tablet as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.271
    
where:
Ru=Area of the griseofulvin sample peak (at a retention time 
equal to that observed for the griseofulvin standard)/Area of the 
internal standard peak;
Rs=Area of the griseofulvin working standard peak/Area of the 
internal standard peak;
f=Potency of the griseofulvin working standard in micrograms per 
milligram;
Wa=Average tablet weight in milligrams;
Ws=Weight of the griseofulvin working standard in milligrams;
Wu=Weight of the ground tablet powder sample in milligrams;
Vu=Volume of the dissolved ground tablet powder sample in 
milliliters.

    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure described in paragraph (e)(1) of that 
section.
[39 FR 19134, May 30, 1974, as amended at 44 FR 20660, Apr. 6, 1979; 50 
FR 19920, May 13, 1985]



Sec. 449.120b  Griseofulvin capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Griseofulvin capsules are gelatin 
capsules containing griseofulvin with a suitable filler and binder, with 
or without a suitable lubricant. Each capsule contains 125 or 250 
milligrams of griseofulvin. The griseofulvin content is satisfactory if 
it is not less than 90 percent and not more than 115 percent of the 
number of milligrams of griseofulvin that it is represented to contain. 
The loss on drying is not more than 1.0 percent. The griseofulvin used 
conforms to the standards prescribed by Sec. 449.20(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The griseofulvin used in making the batch for griseofulvin 
content, loss on drying, melting point, specific rotation, identity, 
residue on ignition, heavy metals, specific surface area, and 
crystallinity.
    (b) The batch for griseofulvin content and loss on drying.
    (ii) Samples required:
    (a) The griseofulvin used in making the batch: 10 packages, each 
containing not less than 1 gram.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Griseofulvin content (gas liquid 
chromatography). Proceed as directed in Sec. 436.321 of this chapter, 
except:
    (i) Prepare the sample solution as follows: Empty the contents of 20 
capsules into a tared weighing bottle. Weigh the powder and calculate 
the average capsule weight. Mix the powder and transfer an accurately 
weighed sample to a volumetric flask of such size that for each 50 
milliliters of volume there are 40 milligrams of griseofulvin 
(estimated). Add chloroform to about one-fourth volume of the flask. 
Swirl the flask and apply gentle heat to aid in dissolution of the 
griseofulvin. Allow the mixture to cool and then dilute to volume with 
chloroform and mix. Allow to settle and transfer 2.0 milliliters of the 
supernate to a conical centrifuge tube and evaporate to dryness under a 
current of dry air. Add 1.0 milliliter of the internal standard solution 
to the centrifuge tube and mix vigorously to obtain a uniform solution; 
and,

[[Page 874]]

    (ii) Calculate the milligrams of griseofulvin per capsule as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.272

where:
Ru=Area of the griseofulvin sample peak (at a retention time 
equal to that observed for the griseofulvin standard)/Area of the 
internal standard peak;
Rs=Area of the griseofulvin working standard peak/Area of the 
internal standard peak;
Ws=Weight of the griseofulvin working standard in milligrams;
f=Potency of the griseofulvin working standard in micrograms per 
milligram;
Wa=Average capsule fill weight in milligrams;
Wu=Weight of the ground tablet powder sample in milligrams;
Vu=Volume of the dissolved capsule powder sample in 
milliliters.

    (2) Loss of drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[39 FR 19134, May 30, 1974, as amended at 43 FR 9800, Mar. 10, 1978; 44 
FR 20661, Apr. 6, 1979; 50 FR 19920, May 13, 1985]



Sec. 449.120c  Griseofulvin oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Griseofulvin oral suspension is 
griseofulvin oral suspension with one or more suitable flavorings, 
colorings, wetting agents, preservatives, and diluents in an aqueous 
vehicle. Each milliliter contains 25 milligrams of griseofulvin. Its 
griseofulvin content is satisfactory if it is not less than 90 percent 
and not more than 115 percent of the number of milligrams of 
griseofulvin that it is represented to contain. Its pH is not less than 
6.5 and not more than 7.5. The griseofulvin used conforms to the 
standards prescribed by Sec. 449.20(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The griseofulvin used in making the batch for griseofulvin 
content, loss on drying, melting point, specific rotation, identity, 
residue on ignition, heavy metals, specific surface area, and 
crystallinity.
    (b) The batch for griseofulvin content and pH.
    (ii) Samples required:
    (a) The griseofulvin used in making the batch: 10 packages, each 
containing not less than 1 gram.
    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Griseofulvin content (gas liquid 
chromatography). Proceed as directed in Sec. 436.321 of this chapter, 
except:
    (i) Prepare the sample solution as follows: Transfer an accurately 
measured portion of the oral suspension equivalent to 100 milligrams of 
griseofulvin into a 50-milliliter round-bottomed glass-stoppered 
centrifuge tube. Add 5 milliliters of water and 20 milliliters of a 
solvent mixture of ethyl acetate and chloroform (85:15). Shake the tube 
for 1 minute and centrifuge it briefly to separate the layers. Transfer 
most of the upper layer to a 100-milliliter volumetric flask being 
careful not to remove any of the lower aqueous layer. Repeat the 
extraction step with two additional 20-milliliter portions of the 
solvent mixture combining the extracts in the volumetric flask with the 
first 20-milliliter extract. Dilute to volume with the solvent mixture 
and mix. Place 2.0 milliliters of this solution in a conical centrifuge 
tube and evaporate the contents to dryness on a steam bath under a 
current of dry air. Add 1.0 milliliter of the internal standard solution 
to the centrifuge tube and mix vigorously to obtain a uniform solution; 
and,
    (ii) Calculate the milligrams of griseofulvin per milliliter as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.273

where:
Ru=Area of the griseofulvin sample peak (at a retention time 
equal to that observed for the griseofulvin standard)/Area of the 
internal standard peak;
Rs=Area of the griseofulvin working standard peak/Area of the 
internal standard peak;
Ws=Weight of the griseofulvin working standard in milligrams;

[[Page 875]]

f=Potency of the griseofulvin working standard in micrograms per 
milligram;
Vo=Volume of oral suspension taken in milliliters.

    (2) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using the undiluted suspension.
[39 FR 19134, May 30, 1974, as amended at 44 FR 20662, Apr. 6, 1979; 50 
FR 19920, May 13, 1985]



Sec. 449.120d  Griseofulvin (ultramicrosize) tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Griseofulvin (ultramicrosize) tablets are 
composed of ultramicrosize crystals of griseofulvin which may or may not 
be dispersed in polyethylene glycol 6,000. Each tablet contains 125, 
165, 250, or 330 milligrams of griseofulvin. The griseofulvin content is 
satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of milligrams of griseofulvin that it is 
represented to contain. The loss on drying is not more than 5.0 percent. 
It passes the solubility characteristic test. If it is dispersed in 
polyethylene glycol 6,000, the griseofulvin used conforms to the 
standards prescribed by Sec. 449.20(a)(1). If it is not dispersed in 
polyethylene glycol 6,000, the griseofulvin used conforms to the 
standards prescribed by Sec. 449.20(a)(1), except specific surface area.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The griseofulvin used in making the batch for potency, loss on 
drying, melting point, specific rotation, identity, residue on ignition, 
heavy metals, specific surface area (if it is dispersed in polyethylene 
glycol 6,000), and crystallinity.
    (b) The batch for griseofulvin content, loss on drying, and 
solubility characteristic.
    (ii) Samples required:
    (a) The griseofulvin used in making the batch: 10 packages, each 
containing not less than 1 gram.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Griseofulvin content (gas liquid 
chromatography). Proceed as directed in Sec. 436.321 of this chapter, 
except:
    (i) Prepare the sample solution as follows: Accurately weigh 20 
tablets and determine the average tablet weight. Grind the tablets to a 
fine powder in a mortar and transfer an accurately weighed sample to a 
volumetric flask of such size that for each 50 milliliters of volume 
there are 40 milligrams of griseofulvin (estimated). Add chloroform to 
about one-fourth volume of the flask. Swirl the flask and apply gentle 
heat to aid in dissolution of the griseofulvin. Allow the mixture to 
cool and then dilute to volume with chloroform. Mix and allow to settle. 
Using gentle vacuum, remove and discard the waxy substance that forms on 
the top of the chloroform. Transfer 2.0 milliliters of the chloroform 
solution to a conical centrifuge tube and evaporate to dryness under a 
current of dry air. Add 1.0 milliliter of the internal standard solution 
to the centrifuge tube and mix vigorously to obtain a uniform solution; 
and,
    (ii) Calculate the milligrams of griseofulvin per tablet as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.274
    
where:
Ru=Area of the griseofulvin sample peak (at a retention time 
equal to that observed for the griseofulvin standard)/Area of the 
internal standard peak;
Rs=Area of the griseofulvin working standard peak/Area of the 
internal standard peak;
Ws=Weight of the griseofulvin working standard in milligrams;
f=Potency of the griseofulvin working standard in micrograms per 
milligram;
Wa=Average tablet weight in milligrams;
Wu=Weight of the ground tablet powder sample in milligrams;
Vu=Volume of the dissolved ground tablet powder sample in 
milliliters.

    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.

[[Page 876]]

    (3) Solubility characteristic test. Proceed as directed in 
Sec. 436.317 of this chapter.
[40 FR 41523, Sept. 8, 1975, as amended at 43 FR 22676, May 26, 1978; 46 
FR 7275, Jan. 23, 1981; 46 FR 21361, Apr. 10, 1981; 46 FR 46313, Sept. 
18, 1981; 47 FR 34132, Aug. 6, 1982; 50 FR 19920, May 13, 1985]



Sec. 449.150  Nystatin oral dosage forms.



Sec. 449.150a  Nystatin tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nystatin tablets are tablets composed of 
nystatin and suitable and harmless buffer substances, diluents, binders, 
lubricants, colorings, and flavorings. Each tablet contains 500,000 
units of nystatin. Its potency is satisfactory if it is not less than 90 
percent and not more than 130 percent of the number of units of nystatin 
that it is represented to contain. The loss on drying is not more than 8 
percent. The tablets shall disintegrate within 2 hours. The nystatin 
used conforms to the standards prescribed by Sec. 449.50(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency, loss on drying, and disintegration time.
    (ii) Samples required:
    (a) The nystatin used in making the batch: 10 packages, each 
consisting of not less than 300 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Blend a representative number of tablets for 3 to 5 minutes in a high-
speed glass blender with sufficient dimethylformamide to give a 
convenient concentration. Dilute an aliquot with sufficient 
dimethylformamide to give a stock solution containing 400 units of 
nystatin per milliliter. Further dilute an aliquot with 10 percent 
potassium phosphate buffer, pH 6.0 (solution 6), to the reference 
concentration of 20 units of nystatin per milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter.
[39 FR 19134, May 30, 1974, as amended at 50 FR 19920, May 13, 1985; 50 
FR 52772, Dec. 26, 1985]



Sec. 449.150b  Nystatin oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nystatin oral suspension is a suspension 
containing nystatin and one or more suitable preservatives, suspending 
agents, surfactants, flavorings, and colorings in purified water. Each 
milliliter contains 100,000 units of nystatin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of units of nystatin that it is represented to 
contain. Its pH is not less than 4.5 and not more than 6.0; except, if 
the product contains glycerin, its pH is not less than 6.0 and not more 
than 7.5. The nystatin used conforms to the standards prescribed by 
Sec. 449.50(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The nystatin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place an accurately measured representative aliquot of the sample into a 
high-speed glass blender jar containing sufficient dimethylformamide to 
give a convenient concentration. Blend for 3 to 5

[[Page 877]]

minutes. Dilute an aliquot with sufficient dimethylformamide to give a 
stock solution containing 400 units of nystatin per milliliter 
(estimated). Remove and dilute with 10 percent potassium phosphate 
buffer, pH 6.0 (solution 6), to the reference concentration of 20 units 
of nystatin per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted suspension.
[39 FR 19134, May 30, 1974, as amended at 50 FR 19920, May 13, 1985]



Sec. 449.150c  Nystatin for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nystatin for oral suspension is a dry 
powder consisting of nystatin, and suitable and harmless suspending 
substances, preservatives, diluents, colorings, and flavorings. When the 
suspension is prepared as directed in its labeling, each milliliter 
contains 100,000 units of nystatin. Its potency is satisfactory if it is 
not less than 90 percent and not more than 140 percent of aliquot of the 
stock solution and further the number of units of nystatin that it is 
represented to contain. The pH of the reconstituted drug is not less 
than 4.9 and not more than 5.5. Its moisture content is not more than 
7.0 percent. The nystatin used conforms to the standards prescribed by 
Sec. 449.50(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency, moisture and pH.
    (ii) Samples required:
    (a) The nystatin used in making the batch: 10 packages, each 
consisting of 300 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute the drug as directed in the labeling. Blend an appropriate 
aliquot in a high-speed glass blender for 3 to 5 minutes, using 
sufficient dimethylformamide to give a convenient concentration. Dilute 
an aliquot with sufficient dimethylformamide to give a stock solution 
containing 400 units of nystatin per milliliter. Further dilute an 
aliquot with 10 percent potassium phosphate buffer, pH 6.0 (solution 6), 
to the reference concentration of 20 units of nystatin per milliliter 
(estimated).
    (2) Moisture. Using the dry powder, proceed as directed in 
Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the suspension after reconstituting as directed in the labeling.
[39 FR 19134, May 30, 1974, as amended at 50 FR 19920, May 13, 1985]



Sec. 449.150d  Nystatin pastilles.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nystatin pastilles are composed of 
nystatin with suitable diluents, binders, buffers, colorings, and 
flavorings. Each pastille contains nystatin equivalent to 200,000 units 
of nystatin. Its potency is satisfactory if it contains not less than 90 
percent and not more than 125 percent of the number of units of nystatin 
that it is represented to contain. The pH in an aqueous solution is not 
less than 5.0 and not more than 7.5. It disintegrates within 90 minutes. 
The nystatin used conforms to the standards prescribed by 
Sec. 449.50(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency, pH, and disintegration time.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:

[[Page 878]]

    (a) The nystatin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 36 pastilles.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of pastilles into a high-speed glass 
blender jar containing 100 milliliters of sterile distilled water. Blend 
for 18 to 20 minutes. Add 400 milliliters of dimethylformamide and 
continue blending for an additional 10 minutes. Remove an aliquot and 
add sufficient 80 percent dimethylformamide so that upon final dilution 
with 10 percent potassium phosphate buffer, pH 6.0 (solution 6), to the 
reference concentration of 20 units of nystatin per milliliter, the 
concentration of dimethylformamide will be 4 percent.
    (2) pH. Dissolve 1 pastille in 100 milliliters of distilled water at 
37  deg.C, cool, and proceed as directed in Sec. 436.202 of this 
chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the method described in paragraph (e)(4) of that section.
[52 FR 4617, Feb. 13, 1987; 52 FR 7741, Mar. 12, 1987, as amended at 55 
FR 11584, Mar. 29, 1990]



                   Subpart C--Injectable Dosage Forms



Sec. 449.204  Amphotericin B for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amphotericin B for injection is a dry 
mixture containing in each immediate container 50 milligrams of 
amphotericin B, 41 milligrams of sodium desoxycholate, and suitable 
buffering substances. Its potency is satisfactory if it is not less than 
90 percent and not more than 120 percent of the number of milligrams of 
amphotericin B that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its loss on drying is not more than 8.0 percent. Its pH in 
an aqueous solution containing 10 milligrams of amphotericin B per 
milliliter is not less than 7.2 and not more than 8.0. The amphotericin 
B used conforms to the standards prescribed by Sec. 449.4a(a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, each package shall bear on its label and 
labeling the following statement: ``For intravenous infusion in 
hospitals only''.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The amphotericin B used in making the batch for potency, 
amphotericin A content, loss on drying, pH, residue on ignition, and 
identity.
    (b) The batch for potency, sterility, pyrogens, loss on drying, and 
pH.
    (ii) Samples required:
    (a) Amphotericin B used in making the batch: 10 packages, each 
containing approximately equal portions of not less than 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Then using a suitable syringe 
and hypodermic needle, remove all of the withdrawable contents if the 
container is represented as a single-dose container; or, if the labeling 
specifies the amount of potency in a given volume of the resultant 
preparation, remove an accurately measured representative portion from 
each container. Dilute with sufficient dimethylsulfoxide to give a stock 
solution of convenient concentration. Further dilute an aliquot of the 
stock solution with dimethylsulfoxide to a concentration of 20 
micrograms of amphotericin B per milliliter (estimated). Remove an 
aliquot of this solution and dilute with 0.2M potassium phosphate 
buffer, pH 10.5 (solution 10), to the reference concentration of 1.0 
microgram of amphotericin B per milliliter (estimated).

[[Page 879]]

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use 50 milligrams in lieu of 300 milligrams.
    (3) [Reserved]
    (4) Pyrogens. Proceed as directed in Sec. 436.32(e) of this chapter, 
using a solution containing 2 milligrams of amphotericin B per 
milliliter, except in lieu of paragraph (a)(3), if no rabbit shows an 
individual rise in temperature of 1.1 deg. C. or more above its 
respective control temperature, and if the sum of the three temperature 
rises does not exceed 3 deg. C., the sample meets the requirements for 
absence of pyrogen. If one or two rabbits show a temperature rise of 
1.1 deg. C. or more, or if the sum of temperature rises exceeds 3 deg. 
C., repeat the test using five other rabbits. If not more than three of 
the eight rabbits show a temperature rise of 1.1 deg. C. or more, and if 
the sum of the temperature rises does not exceed 8 deg. C. the sample 
meets the requirements for absence of pyrogens.
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter using an 
aqueous solution containing 10 milligrams of amphotericin B per 
milliliter.
[39 FR 19134, May 30, 1974, as amended at 45 FR 16472, Mar. 14, 1980; 50 
FR 19920, May 13, 1985]



                   Subpart D--Ophthalmic Dosage Forms



Sec. 449.340  Natamycin ophthalmic suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Natamycin ophthalmic suspension contains 
natamycin with one or more suitable and harmless preservatives in a 
suitable and harmless aqueous vehicle. Each milliliter contains 50 
milligrams of natamycin. Its potency is satisfactory if it is not less 
than 90 percent and not more than 125 percent of the number of 
milligrams of natamycin that it is represented to contain. It is 
sterile. Its pH is not less than 5.0 and not more than 7.5. The 
natamycin used conforms to the standards prescribed by 
Sec. 449.40(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The natamycin used in making the batch for potency, moisture, 
pH, identity, and crystallinity.
    (b) The batch for potency, sterility, and pH.
    (ii) Samples required:
    (a) The natamycin used in making the batch: 10 packages, each 
containing not less than 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of five immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Dilute solutions of natamycin are 
very sensitive to light and should be kept in the dark as much as 
possible or substantial decomposition will take place.
    (1) Potency. Proceed as directed in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Dilute an accurately measured 
representative portion of the sample with sufficient dimethylsulfoxide 
to give a stock solution of convenient concentration. Further dilute an 
aliquot of the stock solution with dimethylsulfoxide to a concentration 
of 100 micrograms of natamycin per milliliter (estimated). Further 
dilute an aliquot with 0.2M potassium phosphate buffer, pH 10.5 
(solution 10), to the reference concentration of 5.0 micrograms of 
natamycin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section, except 
use 0.25 milliliter of sample in lieu of 1.0 milliliter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted suspension.
[43 FR 55384, Nov. 28, 1978, as amended at 48 FR 51293, Nov. 8, 1983]

[[Page 880]]



                          Subpart E  [Reserved]



                  Subpart F--Dermatologic Dosage Forms



Sec. 449.504  Amphotericin B dermatologic dosage forms.



Sec. 449.504a  Amphotericin B ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amphotericin B ointment is composed of 
amphotericin B in a suitable and harmless ointment base. It may contain 
suitable and harmless coloring agents and protectants. It contains 30 
milligrams of amphotericin B in each gram. Its potency is satisfactory 
if it is not less than 90 percent and not more than 125 percent of the 
number of milligrams of amphotericin B that it is represented to 
contain. Its moisture content is not more than 1.0 percent. The 
amphotericin B used conforms to the standards prescribed by 
Sec. 449.4(a)(1) (i), (ii), (v), (vi), and (vii).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The amphotericin B used in making the batch for potency, 
amphotericin A content, pH, residue on ignition, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) Amphotericin B used in making the batch: 10 packages, each 
containing not less than 500 milligrams.
    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the sample 
(usually 1 gram) into an appropriate-sized Erlenmeyer flask with 10 
milliliters of ethyl ether. Allow to dissolve for 1 hour with the 
intermittent manual shaking. Add a measured amount of dimethylsulfoxide 
to the flask and place on a shaker for 10 minutes. Further dilute with 
dimethylsulfoxide to a concentration of 20 micrograms of amphotericin B 
per milliliter (estimated). Remove an aliquot and dilute with 0.2M 
potassium phosphate buffer, pH 10.5 (solution 10), to the reference 
concentration of 1.0 microgram of amphotericin B per milliliter 
(estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.



Sec. 449.504b  Amphotericin B cream.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amphotericin B cream is composed of 
amphotericin B, with or without one or more suitable and harmless 
emollients, perfumes, dispersants, and preservatives, in a suitable and 
harmless cream base. It contains 30 milligrams of amphotericin B in each 
gram. Its potency is satisfactory if it is not less than 90 percent and 
not more than 125 percent of the number of milligrams of amphotericin B 
per gram that it is represented to contain. The amphotericin B used 
conforms to the standards prescribed by Sec. 449.4(a)(1) (i), (ii), (v), 
(vi), and (vii).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The amphotericin B used in making the batch for potency, 
amphotericin A content, pH, residue on ignition, and identity.
    (b) The batch for potency.
    (ii) Samples required:
    (a) Amphotericin B used in making the batch: 10 packages, each 
containing not less than 500 milligrams.
    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay; potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
With the aid of a high-speed glass blender, dissolve an accurately 
weighed sample in sufficient dimethylsulfoxide to give a stock solution 
of convenient concentration. Further dilute with dimethylsulfoxide to a 
concentration of 20 micrograms of

[[Page 881]]

amphotericin B per milliliter (estimated). Remove an aliquot and dilute 
with 0.2M potassium phosphate buffer, pH 10.5 (solution 10), to the 
reference concentration of 1.0 microgram of amphotericin B per 
milliliter (estimated).



Sec. 449.504c  Amphotericin B lotion.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Amphotericin B lotion is composed of 
amphotericin B in a suitable and harmless lotion vehicle. It contains 
suitable and harmless emollients, emulsifiers, coloring agents, 
diluents, preservatives, and perfumes. It contains 30 milligrams of 
amphotericin B per milliliter. Its potency is satisfactory if it is not 
less than 90 percent and not more than 125 percent of the number of 
milligrams of amphotericin B per milliliter that it is represented to 
contain. Its pH is not less than 5.0 and not more than 7.0. The 
amphotericin B used conforms to the standards prescribed by 
Sec. 449.4(a)(1) (i), (ii), (v), (vi), and (vii).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The amphotericin B used in making the batch for potency, 
amphotericin A content, pH, residue on ignition, and identity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The amphotericin B used in making the batch: 10 packages, each 
containing not less than 500 milligrams.
    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an aliquot in sufficient dimethylsulfoxide to give a stock 
solution of convenient concentration. Further dilute the stock solution 
with dimethylsulfoxide to a concentration of 20 micrograms of 
amphotericin B per milliliter (estimated). Remove an aliquot and dilute 
with 0.2M potassium phosphate buffer, pH 10.5 (solution 10), to the 
reference concentration of 1.0 microgram of amphotericin B per 
milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted lotion.



Sec. 449.550  Nystatin dermatologic dosage forms.



Sec. 449.550a  Nystatin ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nystatin ointment is composed of nystatin 
and a suitable and harmless ointment base. Each gram contains 100,000 
units of nystatin. Its potency is satisfactory if it is not less than 90 
percent and not more than 130 percent of the number of units of nystatin 
that it is represented to contain. The moisture content is not more than 
0.5 percent. The nystatin used conforms to the standards prescribed by 
Sec. 449.50(a)(1) (i), (iii), (iv), and (v).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The nystatin used in making the batch: 10 containers, each 
consisting of 300 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Using sufficient dimethylformamide to give a concentration of 400 units 
of nystatin (estimated) per milliliter, blend an accurately weighed 
representative portion in a high-speed glass blender for 3 to 5 minutes. 
Further dilute with 10 percent potassium phosphate buffer, pH 6 
(solution 6), to the reference concentration of 20 units of nystatin per 
milliliter (estimated).

[[Page 882]]

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.



Sec. 449.550b  Nystatin-iodochlorhydroxyquin ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nystatin-iodochlorhydroxyquin ointment is 
composed of nystatin and iodochlorhydroxyquin in a suitable and harmless 
ointment base. Each gram contains 100,000 units of nystatin and 10 
milligrams of iodochlorhydroxyquin. Its nystatin content is satisfactory 
if it is not less than 90 percent and not more than 140 percent of the 
number of units of nystatin that it is represented to contain. Its 
iodochlorhydroxyquin content is satisfactory if it is not less than 90 
percent and not more than 110 percent of the number of milligrams of 
iodochlorhydroxyquin that it is represented to contain. It passes the 
identity test for iodochlorhydroxyquin. Its moisture content is not more 
than 0.5 percent. The nystatin used conforms to the standards prescribed 
by Sec. 449.50(a)(1). The iodochlorhydroxyquin used conforms to the 
standards prescribed by U.S.P. XVIII.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The iodochlorhydroxyquin used in making the batch for all U.S.P. 
XVIII specifications.
    (c) The batch for nystatin content, iodochlorhydroxyquin content, 
iodochlorhydroxyquin identity, and moisture.
    (ii) Samples required:
    (a) The nystatin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of seven immediate containers.
    (b) Tests and methods of assay--(1) Nystatin content. Proceed as 
directed in Sec. 436.105 of this chapter, preparing the sample for assay 
as follows: Place an accurately weighed representative portion of the 
sample into a high-speed glass blender jar containing sufficient 
dimethylformamide to give a convenient concentration. Blend for 3 to 5 
minutes. Remove an aliquot and dilute with sufficient dimethylformamide 
to yield a stock solution containing 400 units of nystatin per 
milliliter (estimated). Further dilute an aliquot of the stock solution 
with 10 percent potassium phosphate buffer, pH 6.0 (solution 6), to the 
reference concentration of 20 units of nystatin per milliliter 
(estimated).
    (2) Iodochlorhydroxyquin content--(i) Reagents. (a) Ferric chloride 
reagent. Dissolve 1.0 gram of ferric chloride 
(FeCl36H2O) in a mixture of 1.0 
milliliter of concentrated hydrochloric acid and sufficient distilled 
water to make 1 liter.
    (b) Acetone, reagent grade.
    (c) 2-Methoxyethanol, reagent grade.
    (ii) Preparation of standard solution. Dissolve an accurately 
weighed portion of iodochlorhydroxyquin U.S.P. reference standard in 
sufficient 2-methoxyethanol to make a solution containing 1.0 milligram 
of iodochlorhydroxyquin per milliliter. Transfer 5.0 milliliters of this 
standard solution to a 50-milliliter volumetric flask.
    (iii) Preparation of sample solution. Accurately weigh a portion of 
the sample equivalent to 50 milligrams of iodochlorhydroxyquin into a 
125-milliliter Erlenmeyer flask. Add 50 milliliters of acetone, warm on 
a steam bath, and shake gently. Cool to room temperature and filter 
contents through a pledget of glass wool into a 100-milliliter 
volumetric flask. Wash the Erlenmeyer flask with two 20-milliliter 
portions of acetone and filter the washings into the volumetric flask. 
Dilute to volume with acetone and mix thoroughly. Transfer a 10-
milliliter aliquot of the acetone solution to a 50-milliliter volumetric 
flask and evaporate on a steam bath. To the residue, add 20 milliliters 
of 2-methoxyethanol and swirl to dissolve the iodochlorhydroxyquin.
    (iv) Procedure. To each flask containing standard solution and 
sample solution, respectively, add 2.0 milliliters of ferric chloride 
reagent and dilute to

[[Page 883]]

volume with 2-methoxyethanol. Mix thoroughly. Using a suitable 
spectrophotometer equipped with 1.0-centimeter cells and a blank 
prepared by diluting 2.0 milliliters of ferric chloride reagent to 50 
milliliters with 2-methoxyethanol, determine the absorbance of the 
sample and standard solutions at 650 nanometers. Set the instrument to 
100-percent transmission with the blank.
    (v) Calculation.

Milligrams of iodochlorhydroxyquin per gram of sample=(Absorbance of 
          sample  x  50)/(Absorbance of standard  x  Weight of sample in 
          grams)
    (3) Iodochlorhydroxyquin identity. Proceed as directed in 
Sec. 436.400 of this chapter, preparing the sample solution as follows: 
Accurately weigh a portion of the sample equivalent to 50 milligrams of 
iodochlorhydroxyquin into a 125-milliliter Erlenmeyer flask. Add 50 
milliliters of acetone, warm on a steam bath, and shake gently. Cool to 
room temperature and filter contents through a pledget of glass wool 
into a 100-milliliter volumetric flask. Wash the Erlenmeyer flask with 
two 20-milliliter portions of acetone and filter the washings into the 
volumetric flask. Dilute to volume with acetone and mix thoroughly.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19134, May 30, 1974, as amended at 50 FR 19920, May 13, 1985]



Sec. 449.550c  Nystatin-neomycin sulfate-gramicidin-triamcinolone acetonide ointment; nystatin-neomycin sulfate-gramicidin-fludrocortisone acetate ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. The drug is nystatin, neomycin sulfate, 
gramicidin, and either triamcinolone acetonide or fludrocortisone 
acetate in a suitable ointment base. Each gram contains 100,000 units of 
nystatin, 2.5 milligrams of neomycin, 0.25 milligram of gramicidin, and 
either 1.0 milligram of triamcinolone acetonide or 1.0 milligram of 
fludrocortisone acetate. Its nystatin content is satisfactory if it is 
not less than 90 percent and not more than 140 percent of the number of 
units of nystatin that it is represented to contain. Its neomycin 
content is satisfactory if it is not less than 90 percent and not more 
than 140 percent of the number of milligrams of neomycin that it is 
represented to contain. Its gramicidin content is satisfactory if it is 
not less than 90 percent and not more than 140 percent of the number of 
milligrams of gramicidin that it is represented to contain. Its moisture 
content is not more than 0.5 percent. The nystatin used conforms to the 
standards prescribed by Sec. 449.50(a)(1) (i), (iii), (iv), and (v). The 
neomycin sulfate used conforms to the standards prescribed by 
Sec. 444.42a(a)(1) of this chapter. The gramicidin used conforms to the 
standards prescribed by Sec. 448.25(a)(1) (i), (iii), (iv), (v), and 
(vi) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (c) The gramicidin used in making the batch for potency, loss on 
drying, residue on ignition, melting point, crystallinity, and identity.
    (d) The batch for potency and moisture.
    (ii) Samples required:
    (a) The nystatin used in making the batch: 10 packages, each 
consisting of 300 milligrams.
    (b) The neomycin sulfate used in making the batch: 10 packages, each 
consisting of 300 milligrams.
    (c) The gramicidin used in making the batch: 10 packages, each 
consisting of 500 milligrams.
    (d) The batch: A minimum of seven immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Nystatin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Blend an accurately weighed representative 
portion in a high-speed glass blender for 3 to 5 minutes with sufficient 
dimethylformamide to give a

[[Page 884]]

concentration of 400 units of nystatin per milliliter (estimated). 
Further dilute with 10 percent potassium phosphate buffer, pH 6.0 
(solution 6), to the reference concentration of 20 units of nystatin per 
milliliter (estimated).
    (ii) Neomycin content. Proceed as directed in Sec. 436.105 of this 
chapter, preparing the sample for assay as follows: Place an accurately 
weighed representative portion of the ointment into a separatory funnel 
containing 50 milliliters of peroxide-free ether. Shake the sample and 
ether until homogenous. Add 20 to 25 milliliters of 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), and shake well. Allow the layers 
to separate. Remove the buffer layer and repeat the extraction with new 
portions of the buffer at least three times and any additional times 
necessary to insure complete extraction of the antibiotic. Combine the 
extractives and adjust to an appropriate volume to give a stock solution 
of convenient concentration. Further dilute an aliquot of the stock 
solution with solution 3 to the reference concentration of 1.0 microgram 
of neomycin per milliliter (estimated).
    (iii) Gramicidin content. Proceed as directed in Sec. 436.106 of 
this chapter, preparing the sample for assay as follows: Accurately 
weigh and dissolve a representative portion of the sample in 
approximately 50 milliliters of petroleum ether in a separatory funnel. 
Extract with 20 milliliters of 80 percent alcohol prepared from alcohol 
U.S.P. XX. Repeat the extraction three times. Combine the extractives in 
a suitable volumetric flask, bring to volume with alcohol U.S.P. XX, and 
mix well. Further dilute with alcohol U.S.P. XX to the reference 
concentration of 0.04 microgram of gramicidin per milliliter 
(estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19134, May 30, 1974, as amended at 47 FR 23710, June 1, 1982; 50 
FR 19920, May 13, 1985]



Sec. 449.550d  Nystatin cream.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nystatin cream is composed of nystatin 
and suitable and harmless emulsifiers, perfumes, buffers, preservatives, 
and a protectant in a suitable and harmless cream base. Each gram 
contains 100,000 units of nystatin. Its potency is satisfactory if it is 
not less than 90 percent and not more than 130 percent of the number of 
units of nystatin that it is represented to contain. The nystatin used 
conforms to the standards prescribed by Sec. 449.50(a)(1) (i), (iii), 
(iv), and (v).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency.
    (ii) Samples required:
    (a) The nystatin used in making the batch: 10 containers, each 
consisting of 300 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay; potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Using sufficient dimethylformamide to give an estimated concentration of 
400 units of nystatin per milliliter, blend an accurately weighed 
representative portion in a high-speed blender for 3 to 5 minutes. 
Further dilute with 10 percent potassium phosphate buffer, pH 6.0 
(solution 6), to the reference concentration of 20 units of nystatin per 
milliliter (estimated).



Sec. 449.550e  Nystatin-neomycin sulfate-gramicidin-triamcinolone acetonide cream.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nystatin-neomycin sulfate-gramicidin-
triamcinolone acetonide cream is composed of nystatin, neomycin sulfate, 
gramicidin, triamcinolone acetonide, and suitable and harmless 
emulsifiers, solvents, perfumes, buffers, preservatives, and a 
protectant in a suitable cream base. Each gram contains 100,000 units of 
nystatin, 2.5 milligrams of neomycin, 0.25 milligram of gramicidin, and 
1 milligram of

[[Page 885]]

triamcinolone acetonide. Its nystatin content is satisfactory if it is 
not less than 90 percent and not more than 140 percent of the number of 
units of nystatin that it is represented to contain. Its neomycin 
content is satisfactory if it is not less than 90 percent and not more 
than 140 percent of the number of milligrams of neomycin that it is 
represented to contain. Its gramicidin content is satisfactory if it is 
not less than 90 percent and not more than 140 percent of the number of 
milligrams of gramicidin that it is represented to contain. The nystatin 
used conforms to the standards prescribed by Sec. 449.50(a)(1) (i), 
(iii), (iv), and (v). The neomycin sulfate used conforms to the 
standards prescribed by Sec. 444.42(a)(1) of this chapter. The 
gramicidin used conforms to the standards prescribed by 
Sec. 448.25(a)(1) (i), (iii), (iv), (v), and (vi) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (c) The gramicidin used in making the batch for potency, loss on 
drying, residue on ignition, melting point, crystallinity, and identity.
    (d) The batch for nystatin content, neomycin content, and gramicidin 
content.
    (ii) Samples required:
    (a) The nystatin used in making the batch: 10 packages, each 
consisting of 300 milligrams.
    (b) The neomycin sulfate used in making the batch: 10 packages, each 
consisting of 300 milligrams.
    (c) The gramicidin used in making the batch: 10 packages, each 
consisting of 500 milligrams.
    (d) The batch: A minimum of seven immediate containers.
    (b) Tests and methods of assay--(1) Potency--(i) Nystatin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Using sufficient dimethylformamide to give 
a concentration of 400 units of nystatin (estimated) per milliliter, 
blend an accurately weighed representative portion in a high-speed glass 
blender for 3 to 5 minutes. Further dilute with 10 percent potassium 
phosphate buffer, pH 6.0 (solution 6), to the reference concentration of 
20 units of nystatin per milliliter (estimated).
    (ii) Neomycin content. Proceed as directed in Sec. 436.105 of this 
chapter, preparing the sample for assay as follows: Place an accurately 
weighed representative portion of the cream in a separatory funnel 
containing 50 milliliters of peroxide-free ether. Shake the sample and 
either until homogeneous. Add 20 to 25 milliliters of 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), and shake well. Allow the layers 
to separate. Remove the buffer layer and repeat the extraction with new 
portions of the buffer at least three times and any additional times 
necessary to ensure complete extraction of the antibiotic. Combine the 
extractives and adjust to an appropriate volume to give a stock solution 
of convenient concentration. Further dilute an aliquot of the stock 
solution with solution 3 to the reference concentration of 1.0 microgram 
of neomycin per milliliter (estimated).
    (iii) Gramicidin content. Proceed as directed in Sec. 436.106 of 
this chapter, preparing the sample for assay as follows: Accurately 
weigh a representative portion of the sample and dissolve in 
approximately 50 milliliters of petroleum ether in a separatory funnel. 
Extract with 20 milliliters of 80 percent alcohol prepared from alcohol 
U.S.P. XX. Repeat the extraction three times. Combine the extractives in 
a suitable volumetric flask, bring to volume with alcohol U.S.P. XX, and 
mix well. Further dilute with alcohol U.S.P. XX to the reference 
concentration of 0.04 microgram of gramicidin per milliliter 
(estimated).
[39 FR 19134, May 30, 1974, as amended at 47 FR 23710, June 1, 1982; 50 
FR 19920, May 13, 1985]



Sec. 449.550f  Nystatin topical powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality,

[[Page 886]]

and purity. Nystatin topical powder is a dry powder composed of nystatin 
and talc. Each gram contains 100,000 units of nystatin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of units of nystatin that it is represented to 
contain. Its loss on drying is not more than 2.0 percent. The nystatin 
used conforms to the standards prescribed by Sec. 449.50(a)(1) (i), 
(iii), (iv), and (v).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:
    (a) The nystatin used in making the batch: 10 packages, each 
containing 300 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Blend an accurately weighed representative sample for 3 to 5 minutes in 
a high-speed glass blender with sufficient dimethylformamide to give a 
convenient concentration. Dilute with sufficient dimethylformamide to 
yield a stock solution containing 400 units of nystatin per milliliter 
(estimated). Further dilute with 10 percent potassium phosphate buffer, 
pH 6.0 (solution 6), to the reference concentration of 20 units of 
nystatin per milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.



Sec. 449.550g  Nystatin-neomycin sulfate-gramicidin topical powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nystatin-neomycin sulfate-gramicidin 
topical powder is a dry powder composed of nystatin, neomycin sulfate, 
gramicidin, and talc. Each gram contains 100,000 units of nystatin, 2.5 
milligrams of neomycin and 0.25 milligram of gramicidin. Its nystatin 
content is satisfactory if it is not less than 90 percent and not more 
than 140 percent of the number of units of nystatin that it is 
represented to contain. Its neomycin content is satisfactory if it is 
not less than 90 percent and not more than 140 percent of the number of 
milligrams of neomycin that it is represented to contain. Its gramicidin 
content is satisfactory if it is not less than 90 percent and not more 
than 140 percent of the number of milligrams of gramicidin that it is 
represented to contain. Its loss on drying is not more than 2.0 percent. 
The nystatin used conforms to the standards prescribed by 
Sec. 449.50(a)(1) (i), (iii), (iv), and (v). The neomycin sulfate used 
conforms to the standards prescribed by Sec. 444.42a(a)(1) of this 
chapter. The gramicidin used conforms to the standards prescribed by 
Sec. 448.25(a)(1) (i), (iii), (iv), (v) and (vi) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The neomycin sulfate used in making the batch for potency, loss 
on drying, pH, and identity.
    (c) The gramicidin used in making the batch for potency, loss on 
drying, residue on ignition, melting point, crystallinity, and identity.
    (d) The batch for nystatin content, neomycin content, gramicidin 
content, and loss on drying.
    (ii) Samples required:
    (a) The nystatin used in making the batch: 10 packages each 
consisting of 300 milligrams.
    (b) The neomycin sulfate used in making the batch: 10 packages, each 
consisting of 300 milligrams.
    (c) The gramicidin used in making the batch: 10 packages, each 
consisting of 500 milligrams.
    (d) The batch: A minimum of seven immediate containers.

[[Page 887]]

    (b) Tests and methods of assay--(1) Potency--(i) Nystatin content. 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Blend the entire contents of an accurately 
weighed representative portion of the sample for 3 to 5 minutes in a 
high-speed glass blender with sufficient dimethylformamide to give a 
convenient concentration. Dilute with sufficient dimethylformamide to 
yield a stock solution containing 400 units of nystatin per milliliter 
(estimated). Further dilute with 10 percent potassium phosphate buffer, 
pH 6.0 (solution 6), to the reference concentration of 20 units of 
nystatin per milliliter (estimated).
    (ii) Neomycin content. Proceed as directed in Sec. 436.105 of this 
chapter, preparing the sample for assay as follows: Blend an accurately 
weighed representative sample for 3 to 5 minutes in sufficient 0.1M 
potassium phosphate buffer, pH 8 (solution 3), to give a convenient 
concentration. Further dilute an aliquot with solution 3 to the 
reference concentration of 1 microgram of neomycin per milliliter 
(estimated).
    (iii) Gramicidin content. Proceed as directed in Sec. 436.106 of 
this chapter, preparing the sample for assay as follows: Dissolve an 
accurately weighed representative sample in alcohol U.S.P. XX and 
filter. Collect the filtrate and dilute a portion with alcohol U.S.P. XX 
to the reference concentration of 0.04 microgram of gramicidin per 
milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[39 FR 19134, May 30, 1974, as amended at 41 FR 10886, Mar. 15, 1976; 47 
FR 23710, June 1, 1982; 50 FR 19920, May 13, 1985]



Sec. 449.550h  Nystatin lotion.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nystatin lotion is composed of nystatin 
with one or more suitable and harmless suspending agents, emulsifiers, 
surfactants, and preservatives in a suitable and harmless vehicle. Each 
milliliter contains 100,000 units of nystatin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 140 
percent of the number of units of nystatin that it is represented to 
contain. Its pH is not less than 5.5 and not more than 7.5. The nystatin 
used conforms to the standards prescribed by Sec. 449.50(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The nystatin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place an accurately measured representative portion of the sample into a 
high-speed glass blender jar containing sufficient dimethylformamide to 
give a convenient concentration. Blend for 3 to 5 minutes. Remove an 
aliquot and dilute with sufficient dimethylformamide to yield a stock 
solution containing 400 units of nystatin per milliliter (estimated). 
Further dilute with 10 percent potassium phosphate buffer, pH 6.0 
(solution 6), to the reference concentration of 20 units of nystatin per 
milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[40 FR 3766, Jan. 24, 1975, as amended at 50 FR 19920, May 13, 1985]



                     Subpart G--Vaginal Dosage Forms



Sec. 449.610  Candicidin vaginal dosage forms.



Sec. 449.610a  Candicidin vaginal ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Candicidin vaginal ointment is composed 
of candicidin and a suitable ointment base. It contains 0.6 milligram of 
candicidin per gram. Its

[[Page 888]]

potency is satisfactory if it is not less than 90 percent and not more 
than 140 percent of the number of milligrams of candicidin that it is 
represented to contain. Its moisture content is not more than 0.1 
percent. The candicidin used conforms to the requirements of 
Sec. 449.10(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The candicidin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The candicidin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the sample into a 
separatory funnel containing approximately 50 milliliters of n- hexane 
(containing 0.1 percent butylated hydroxyanisole). Shake the sample and 
n- hexane until homogeneous. Add 15 milliliters of dimethylsulfoxide 
(containing 0.1 percent butylated hydroxyanisole) and shake well. Allow 
the layers to separate. Remove the bottom layer and repeat the 
extraction procedure with a second 15-milliliter portion of 
dimethylsulfoxide (containing 0.1 percent butylated hydroxyanisole). 
Combine the extractives in a suitable volumetric flask and fill to 
volume with sterile distilled water. Further dilute an aliquot with 
sterile distilled water to the reference concentration of 0.06 microgram 
of candicidin per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19134, May 30, 1974, as amended at 40 FR 15089, Apr. 4, 1975]



Sec. 449.610b  Candicidin vaginal tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Candicidin vaginal tablets are tablets 
composed of candicidin with suitable binders, diluents, and lubricants. 
Each tablet contains 3 milligrams of candicidin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 150 
percent of the number of milligrams of candicidin that it is represented 
to contain, except that for the issuance of a certificate for each 
batch, the candicidin content must be not less than 115 percent and not 
more than 150 percent of the number of milligrams of candicidin that it 
is represented to contain. The tablets shall disintegrate within 30 
minutes. The loss on drying is not more than 1 percent. The candicidin 
used in making the batch conforms to the standards of Sec. 449.10(a)(1).
    (2) Labeling. The drug shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The candicidin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency, loss on drying, and disintegration time.
    (ii) Samples required. (a) The candicidin used in making the batch: 
10 packages, each consisting of approximately 300 milligrams.
    (b) The batch: A minimum of 56 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Weigh a pool of five tablets and grind in a mortar to a very fine 
powder. Suspend an accurately weighed aliquot (of approximately 2 grams) 
in 10 milliliters of dimethylsulfoxide. Centrifuge for 5 minutes at 
2,000 revolutions per minute. Carefully decant the supernatant solution 
into a sterile 250-milliliter volumetric flask. Wash the residue three 
times with 5-milliliter portions of dimethylsulfoxide, centrifuging each 
time. Add the washes to the 250-milliliter volumetric flask

[[Page 889]]

and fill to volume with sterile distilled water. Using sterile distilled 
water, further dilute to the reference concentration of 0.06 microgram 
of candicidin per milliliter (estimated).
    (2) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the method described in paragraph (e)(1) of that section, 
except use distilled water as the immersion fluid.
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.



Sec. 449.610c  Candicidin vaginal capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Candicidin vaginal capsules are gelatin 
capsules containing 3 milligrams of candicidin in a suitable and 
harmless ointment. The candicidin content is satisfactory if it is not 
less than 90 percent and not more than 150 percent of the number of 
milligrams of candicidin that it is represented to contain. The moisture 
content is not more than 0.1 percent. The candicidin used conforms to 
the requirements of Sec. 449.10(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The candicidin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The candicidin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 20 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Remove the tips from two capsules and express the ointment from each 
capsule into a separatory funnel containing approximately 50 milliliters 
of n- hexane (containing 0.1 percent butylated hydroxyanisole). Wash out 
the capsules at least two times with 2- to 3-milliliter portions of warm 
(approximately 50 deg. C) n- hexane (containing 0.1 percent butylated 
hydroxyanisole). Add the washes to the separatory funnel. Shake the 
sample and n- hexane until homogeneous. Add 15 milliliters of 
dimethylsulfoxide (containing 0.1 percent butylated hydroxyanisole) and 
shake well. Allow the layers to separate. Remove the bottom layer and 
repeat the extraction procedure with a second 15-milliliter portion of 
dimethylsulfoxide (containing 0.1 percent butylated hydroxyanisole). 
Combine the extractives in a suitable volumetric flask and fill to 
volume with sterile distilled water. Further dilute an aliquot with 
sterile distilled water to the reference concentration of 0.06 microgram 
of candicidin per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19134, May 30, 1974, as amended at 40 FR 15089, Apr. 4, 1975]



Sec. 449.650  Nystatin vaginal dosage forms.



Sec. 449.650a  Nystatin vaginal tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nystatin vaginal tablets are tablets 
composed of nystatin and suitable and harmless diluents, binders, and 
lubricants. Each tablet contains 100,000 units of nystatin. Its potency 
is satisfactory if it is not less than 90 percent and not more than 140 
percent of the number of units of nystatin that it is represented to 
contain. The loss on drying is not more than 5 percent. The 
disintegration time is not more than 1 hour. The nystatin used conforms 
to the standards prescribed therefor by Sec. 449.50(a)(1) (i), (iii), 
(iv), and (v).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for nystatin content, loss on drying, and 
disintegration time.
    (ii) Samples required:

[[Page 890]]

    (a) The nystatin used in making the batch: 10 immediate containers 
of approximately 300 milligrams each.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Blend a representative number of tablets for 3 to 5 minutes in a high-
speed glass blender with sufficient dimethylformamide to give a 
convenient concentration. Dilute an aliquot with sufficient 
dimethylformamide to give a stock solution containing 400 units of 
nystatin per milliliter (estimated). Further dilute the stock solution 
with 10 percent potassium phosphate buffer, pH 6.0 (solution 6), to the 
reference concentration of 20 units of nystatin per milliliter 
(estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure described in paragraph (e)(1) of that 
section, except use distilled water in lieu of gastric fluid.
[39 FR 19134, May 30, 1974. Redesignated at 43 FR 43458, Sept. 26, 1978]



Sec. 449.650b  Nystatin vaginal suppositories.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Nystatin vaginal suppositories contain in 
each suppository 100,000 units of nystatin in a suitable and harmless 
water soluble base. Its potency is satisfactory if it is not less than 
90 percent and not more than 130 percent of the number of units of 
nystatin that it is represented to contain. Its moisture content is not 
more than 1.5 percent. The nystatin used conforms to the standards 
prescribed by Sec. 449.50(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The nystatin used in making the batch for potency, loss on 
drying, pH, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The nystatin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch: A minimum of 30 suppositories.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of suppositories into a high-speed glass 
blender jar containing sufficient dimethylformamide to give a convenient 
concentration. Blend for 3 to 5 minutes. Dilute an aliquot with 
sufficient dimethylformamide to obtain a concentration of 400 units of 
nystatin per milliliter (estimated). Further dilute an aliquot with 10 
percent potassium phosphate buffer, pH 6.0 (solution 6), to the 
reference concentration of 20 units of nystatin per milliliter 
(estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[43 FR 43458, Sept. 26, 1978, as amended at 50 FR 19920, May 13, 1985]



PART 450--ANTITUMOR ANTIBIOTIC DRUGS--Table of Contents




                          Subpart A--Bulk Drugs

Sec.
450.10a  Sterile bleomycin sulfate.
450.20  Dactinomycin.
450.22  Daunorubicin hydrochloride.
450.24  Doxorubicin hydrochloride.
450.30  Idarubicin hydrochloride.
450.40  Plicamycin.
450.45  Mitomycin.

                          Subpart B  [Reserved]

                   Subpart C--Injectable Dosage Forms

450.210  Sterile bleomycin sulfate.
450.220  Dactinomycin for injection.
450.222  Daunorubicin hydrochloride for injection.
450.224  Doxorubicin hydrochloride injectable dosage forms.
450.224a  Doxorubicin hydrochloride for injection.
450.224b  Doxorubicin hydrochloride injection.
450.230  Idarubicin hydrochloride for injection.
450.240  Plicamycin for injection.
450.245  Mitomycin for injection.


[[Page 891]]


    Authority:  21 U.S.C. 357.



                          Subpart A--Bulk Drugs



Sec. 450.10a  Sterile bleomycin sulfate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile bleomycin sulfate is the 
amorphous sulfate salt of bleomycin. Bleomycin has been separated into 
several similar glyco-peptide molecules. It is a cream-colored powder 
that is so purified and dried that:
    (i) Its potency is not less than 1.5 units and not more than 2.0 
units of bleomycin per milligram. If it is packaged for dispensing, the 
content of the ampoule or vial is not less than 90 percent and not more 
than 120 percent of the number of units of bleomycin that it is 
represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) It contains no depressor substances.
    (vi) Its loss on drying is not more than 6.0 percent.
    (vii) Its pH in an aqueous solution containing 10 units per 
milliliter is not less than 4.5 and not more than 6.0.
    (viii) Its copper content is not greater than 0.1 percent.
    (ix) Its content of various bleomycins is as follows: Bleomycin 
A2 is not less than 55 percent and not more than 70 percent; 
bleomycin B2 is not less than 25 percent and not more than 32 
percent; bleomycin B4 is not more than 1 percent. Bleomycins 
A2 and B2 should comprise not less than 85 percent 
of the total bleomycins.
    (x) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, depressor substances, loss on drying, pH, copper, content of 
various bleomycins, and identity.
    (ii) Samples required:
    (a) For all tests except sterility: A minimum of 20 immediate 
containers.
    (b) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1M potassium 
phosphate buffer, pH 7.0 (solution 16), to provide a stock solution of 
convenient concentration; if it is packaged for dispensing, reconstitute 
as directed in the labeling. Then, using a suitable hypodermic needle 
and syringe, remove all of the withdrawable contents. Dilute the sample 
thus obtained with solution 16 to provide a stock solution of convenient 
concentration. Further dilute an aliquot of the stock solution with 
solution 16 to the reference concentration of 0.04 unit of activity per 
milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use the entire contents of each of the immediate containers tested.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 0.5 unit of bleomycin per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter, using the total contents of 2 or 3 vials.
    (7) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 units per milliliter.
    (8) Copper content--(i) Reagents. Dissolve 10 milligrams of zinc 
dibenzyldithiocarbamate in 100 milliliters of carbon tetrachloride.
    (ii) Preparation of standard copper solution. Accurately weigh 1.965 
grams of cupric sulfate pentahydrate and transfer to a 1-liter 
volumetric flask. Dissolve the material in 0.1N hydrochloric acid, 
dilute to volume with 0.1N hydrochloric acid and mix well. Transfer 3 
milliliters of this stock solution to a 1-liter volumetric flask, dilute 
to volume with 0.1N hydrochloric acid, and mix well. This standard 
copper solution contains 0.0015 milligram of copper per

[[Page 892]]

milliliter. Transfer 10 milliliters of the standard copper solution to a 
60-milliliter separatory funnel.
    (iii) Preparation of the sample. Accurately weigh approximately 15 
milligrams of sample into a 60-milliliter separatory funnel. Dissolve 
the sample in 10 milliliters of 0.1N hydrochloric acid.
    (iv) Procedure. To the separatory funnels containing the sample 
solution and standard copper solution, add 10 milliliters of the zinc 
dibenzyldithiocarbamate solution and shake the funnels vigorously for 1 
minute. Allow the phases to separate. Filter the carbon tetrachloride 
phase (lower phase) through 1 gram of anhydrous sodium sulfate to remove 
excess water. Using a suitable spectrophotometer equipped with 1-
centimeter cells, and carbon tetrachloride as a blank, measure the 
absorbance of the standard copper solution and the sample solution at 
435 nanometers. Calculate the percent copper as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.275

    (9) Content of various bleomycin fractions. Proceed as directed in 
Sec. 436.339 of this chapter.
    (10) Identity test. Proceed as directed in Sec. 436.211 of this 
chapter, using the method described in paragraph (b)(1) of that section, 
using a 1 percent mixture.
[40 FR 52005, Nov. 7, 1975; 40 FR 53998, Nov. 20, 1975, as amended at 46 
FR 60568, Dec. 11, 1981; 48 FR 51913, Nov. 15, 1983; 50 FR 19920, May 
13, 1985]



Sec. 450.20  Dactinomycin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Dactinomycin is a bright-red compound 
that is so purified and dried that:
    (i) Its dactinomycin content is not less than 900 micrograms of 
dactinomycin per milligram, calculated on an anhydrous basis.
    (ii) Its loss on drying is not more than 15 percent.
    (iii) Its absorptivity at 445 nanometers is not less than 0.95 and 
not more than 1.03 times that of the dactinomycin working standard at 
the same wavelength. Its absorbance at 240 nanometers is not less than 
1.3 and not more than 1.5 times its absorbance at 445 nanometers.
    (iv) It is crystalline.
    (v) It passes the identity test for dactinomycin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter, and in addition each 
package shall bear on its label the statement ``Protect from light and 
excessive heat.''
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for dactinomycin 
content, loss on drying, absorptivity, crystallinity, and identity.
    (ii) Samples required: 16 packages, each containing approximately 40 
milligrams.
    (b) Tests and methods of assay. Dactinomycin is toxic and corrosive. 
It must be handled with care in the laboratory. Transfer all dry powders 
in a suitable hood, while wearing rubber gloves. Avoid inhaling fine 
particles of the powder. Do not pipette by mouth. If any of the 
substance contacts the skin, wash copiously with soap and water. Dispose 
of all waste material by dilution with large volumes of trisodium 
phosphate solution.
    (1) Dactinomycin content. Proceed as directed in Sec. 436.331 of 
this chapter, preparing the sample and calculating the dactinomycin 
content as follows:
    (i) Preparation of sample solution. Accurately weigh a sufficient 
amount of the sample to obtain a solution containing approximately 0.25 
milligram per milliliter of dactinomycin in mobile phase.

[[Page 893]]

    (ii) Calculations. Calculate the micrograms of dactinomycin per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.276

where:
Au = Area of the dactinomycin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As = Area of the dactinomycin peak in the chromatogram of the 
          dactinomycin working standard;
Ps = Dactinomycin activity in the dactinomycin working 
          standard solution in micrograms per milliliter;
Cu = Milligrams of sample per milliliter of sample solution; 
          and
m = Percent moisture content of the sample.

    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Absorptivity--(i) Procedure. Accurately weigh approximately 15 
milligrams of the sample ``as is'' and 15 milligrams of the working 
standard dried as directed in Sec. 436.200(a) of this chapter. Transfer 
each weighing to separate 100-milliliter volumetric flasks. Dissolve the 
material and bring to volume with spectrophotometric-grade methyl 
alcohol. Mix well. Pipette 5.0 milliliters of each solution into 
separate 25-milliliter volumetric flasks, dilute to volume with 
spectrophotometric-grade methyl alcohol. Mix well. Using a suitable 
spectrophotometer and 1-centimeter absorption cells, determine the 
absorbance of the sample solution at the 240-nanometer and at the 445-
nanometer absorption peaks (the exact position of the peaks should be 
determined for the particular instrument used). Determine the absorbance 
of the standard at the 445-nanometer absorption peak.
    (ii) Calculations. Calculate the relative absorptivity and the ratio 
for the absorbances of the sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.277

[GRAPHIC] [TIFF OMITTED] TC01AP94.068

where:

A1=Absorbance at 240 nanometers for the sample;
A2=Absorbance at 445 nanometers for the sample;
A3=Absorbance at 445 nanometers for the standard;
M=Percent moisture in the sample.

    (4) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (5) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the dactinomycin working standard.
[49 FR 6092, Feb. 17, 1984, as amended at 49 FR 24018, June 11, 1984; 50 
FR 19675, May 10, 1985]



Sec. 450.22  Daunorubicin hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Daunorubicin hydrochloride is the 
monohydrochloride salt of (1s,3s)-3-acetyl-1,2,3,4,6,11-hexahydro-
3,5,12-trihydroxy-10-methoxy-6,11-dioxo-1-naphthacenyl-3-amino-2,3,6-
trideoxy--L-lyxo-hexopyranoside. It is a red-orange, 
hygroscopic powder. It is so purified and dried that:
    (i) Its potency is not less than 842 micrograms and not more than 
1,030 micrograms of daunorubicin per milligram.
    (ii) Its moisture content is not more than 3.0 percent.
    (iii) Its pH in an aqueous solution containing 5 milligrams per 
milliliter is not less than 4.5 and not more than 6.5.
    (iv) It is crystalline.
    (v) It passes the identity test for daunorubicin.

[[Page 894]]

    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, crystallinity, and identity.
    (ii) Samples required: 14 packages, each containing approximately 40 
milligrams.
    (b) Tests and methods of assay. Daunorubicin hydrochloride is toxic. 
It must be handled with care in the laboratory. Transfer all dry powders 
in a suitable hood. Wear rubber gloves, protective gowns, head 
coverings, and protective eye goggles when handling dry powders. Avoid 
inhaling fine particles of powder. Solutions should not be pipetted by 
mouth. If the substance contacts the skin, promptly wash with soap and 
water. Dispose of all waste material by dilution with large volumes of 
sodium hypochlorite solution.
    (1) Potency. Use either of the following methods; however, the 
results obtained from the high-pressure liquid chromatography shall be 
conclusive.
    (i) High-pressure liquid chromatography. Proceed as directed in 
Sec. 436.322 of this chapter, except in lieu of the mobile phase and pH 
described in paragraph (b)(2) of that section, use a mixture of water: 
acetonitrile (62:38) adjusted to pH 2.2 plus-minus 0.2 with 
phosphoric acid. Prepare the sample and standard solutions and calculate 
the daunorubicin content as follows:
    (a) Preparation of sample and working standard solutions. Accurately 
weigh approximately 25 milligrams of the sample and of the daunorubicin 
working standard and dissolve each in 25 milliliters of the internal 
standard solution prepared as directed in Sec. 436.322(b)(3) of this 
chapter.
    (b) Calculations. Calculate the daunorubicin content as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.069
    
where:
Ru=Area of the daunorubicin sample peak/Area of the internal 
          standard peak;
Rs=Area of the daunorubicin standard peak/Area of the 
          internal standard peak;
Ws=Weight of the daunorubicin working standard in milligrams;
Wu=Weight of the sample in milligrams;
M=Moisture content of the sample in percent;
P=Potency of the daunorubicin working standard in micrograms per 
          milligram.

    (ii) Microbiological turbidimetric assay for daunorubicin--(a) 
Preparation of working standard stock solutions and standard response 
line concentrations. Dissolve an accurately weighed portion of the 
working standard with sufficient 0.054M sodium phosphate buffer, pH 6.9 
(solution 18), as described in Sec. 436.101(a)(18) of this chapter, to 
obtain a stock solution containing 1 milligram of daunorubicin activity 
per milliliter. The working standard stock solution may be stored under 
refrigeration for 1 week. Further dilute an aliquot of the stock 
solution with solution 18 to obtain standard response line 
concentrations of 4, 8, and 16 micrograms of daunorubicin activity per 
milliliter. The 8-micrograms-per-milliliter concentration is the 
reference concentration of the assay.
    (b) Preparation of sample solution. Dissolve an accurately weighed 
portion of the sample with sufficient 0.054M sodium phosphate buffer, pH 
6.9 (solution 18), as described in Sec. 436.101(a)(18) of this chapter, 
to obtain a stock solution containing 1 milligram of daunorubicin 
activity per milliliter (estimated). Further dilute an aliquot of the 
stock solution with solution 18 to the reference concentration of 8 
micrograms of daunorubicin activity per milliliter (estimated).
    (c) Procedure for assay. Place 1.0 milliliter of each concentration 
of the standard response line and of the sample solution in each set of 
replicate tubes (as described in Sec. 436.100(b)(1) of this chapter). 
Eighteen tubes are used for the three-point standard response line and 
six for each sample. To each tube, add 9 milliliters of medium 3 (as 
listed in Sec. 436.102(b)(3) of this chapter), inoculated with 2 
milliliters of a suspension of test organism I per liter of medium 3. 
The suspension of test organism I is prepared as described in 
Sec. 436.103 of this chapter, except incubate the slants and Roux bottle 
for 16 to 18 hours at 37 deg. C. Place the inoculated tubes immediately 
in a water bath at 37 deg. C for approximately 3 hours.

[[Page 895]]

The absorbance value for the growth control should be approximately 
0.70-0.75 and the absorbance values for the 16 and 4 micrograms per 
milliliter standard doses should be approximately 0.25-0.35 and 0.55-
0.65, respectively. An adjustment of the inoculum may be necessary in 
order to obtain absorbance values to these approximate levels in a 3-
hour time period. Remove the tubes from the water bath and add 0.5 
milliliter of a 12-percent formaldehyde solution to each tube. Determine 
the absorbance value of each tube in a suitable spectrophotometer, at a 
wavelength of 530 nanometers. Set the instrument at zero absorbance with 
an uninoculated blank composed of the same amounts of medium 3, solution 
18, and formaldehyde used in the assay.
    (d) Estimation of potency. Estimate the potency of the sample as 
follows: Using the three x values and the three corresponding y values, 
calculate x, x\2\,(x)\2\, y and 
xy. Calculate b, the slope (regression coefficient), and a, the 
Y-intercept of the standard response line by the following equations:
[GRAPHIC] [TIFF OMITTED] TC01AP94.070

where:

n=Number of standard doses;
x=Logarithm of the concentration in micrograms per milliliter of each 
          dose of the standard curve;
y=Mean response of the six absorbance values for each dose of the 
          standard.

    Calculate the concentration of the sample solution X corresponding 
to the observed mean response of the sample solution Y by the following 
equation:
[GRAPHIC] [TIFF OMITTED] TR01JA93.280

where:

X=The concentration of the sample solution in micrograms per milliliter;
Y=The mean response of the six absorbance values for reference 
          concentration sample solutions.

    Calculate the potency of the daunorubicin sample as follows:
    [GRAPHIC] [TIFF OMITTED] TC01AP94.071
    
where:

F=125, the appropriate dilution factor of the daunorubicin sample;
W=Weight of sample in milligrams.

    The following example illustrates the mathematical calculations of 
the potency of a sample solution:

                                                                                                                
                                                                                                                
                                                                                                                
Standard doses (micrograms per milliliter)..  16.0       8.0       4.0             n =  3                       
Log doses (x)...............................   1.20412   0.90309   0.60206  x  2.70927                 
                                                                                     =  7.34014                 
                                                                            (                          
                                                                             x)  \2\ =                          
x  \2\......................................   1.4499    0.81557   0.36248  x  2.62795                 
                                                                                 \2\ =                          
Absorbance readings.........................   0.247     0.483     0.583    ..........  ........................
                                               0.236     0.414     0.584    ..........  ........................
                                               0.241     0.446     0.574    ..........  ........................
                                               0.236     0.423     0.555    ..........  ........................
                                               0.233     0.416     0.578    ..........  ........................
                                               0.243     0.413     0.559    ..........  ........................
Mean responses (y)..........................   0.239     0.433     0.572    y  1.244                   
                                                                                     =                          
xy..........................................   0.28778   0.39104   0.34438  x  1.0232                  
                                                                                   y =                          
                                                                                                                

                                                                                        [GRAPHIC] [TIFF OMITTED] TR01JA93.278
                                                                                        
Mean response, Y, of sample solution=0.405.

[[Page 896]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.279


    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 5 milligrams per milliliter.
    (4) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (5) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraph (b)(1)(i) of this section compares 
qualitatively to that of the daunorubicin working standard.
[45 FR 75195, Nov. 14, 1980]



Sec. 450.24  Doxorubicin hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Doxorubicin hydrochloride is the 
monohydrochloride salt of (8S, 10S)-10-[(3-amino-2,3,6-trideoxy-
-L-lyxo- hexopyranosyl)oxy]-8-glycoloyl-7,8,9,10-tetrahydro-
6,8,11-trihydroxy-1-methoxy-5,12-naphthacenedione. It is a red-orange, 
almost completely odorless, hygroscopic powder. It is so purified and 
dried that:
    (i) Its doxorubicin hydrochloride content is not less than 970 
micrograms and not more than 1,020 micrograms of doxorubicin 
hydrochloride per milligram on the anhydrous and solvent free basis.
    (ii) Its total solvent residue (as acetone and alcohol) is not more 
than 2.5 percent.
    (iii) It contains no depressor substances.
    (iv) Its moisture content is not more than 4.0 percent.
    (v) The pH of an aqueous solution containing 5 milligrams per 
milliliter is not less than 4.0 and not more than 5.5.
    (vi) It is crystalline.
    (vii) It passes the identity test for doxorubicin.
    (viii) The total of any impurities detected by high-pressure liquid 
chromatography assay is not more than 3.0 percent.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each request shall 
contain:
    (i) Results of tests and assays on the batch for doxorubicin 
hydrochloride content, solvent residue, depressor substances, moisture, 
pH, crystallinity, identity, and total impurities.
    (ii) Samples required: 14 packages, each containing approximately 40 
milligrams.
    (b) Tests and methods of assay. Doxorubicin hydrochloride is toxic. 
It must be handled with care in the laboratory. Transfer all dry powders 
in a suitable hood while wearing rubber gloves. Avoid inhaling fine 
particles of powder. Solutions should not be pipetted by mouth. If the 
substance contacts the skin, wash with soap and water. Dispose of all 
waste material by dilution with large volumes of dilute sodium 
hypochlorite (bleach) solution.
    (1) Doxorubicin hydrochloride content (high-performance liquid 
chromatography). Proceed as directed in Sec. 436.216 of this chapter, 
using ambient temperature, an ultraviolet detection system operating at 
a wavelength of 254 nanometers, a 4.6-millimeter X 25-centimeter column 
packed with microparticulate (5 to 10 micrometers in diameter) packing 
material, such as trimethylsilane chemically bonded to porous silica, a 
flow rate of not more than 2.0 milliliters per minute, and a known 
injection volume of between 10 and 20 microliters. Mobile phase, working 
standard and sample solutions, resolution test solution, system 
suitability requirements, and calculations are as follows:
    (i) Mobile phase. Prepare a suitable mixture of water, acetonitrile, 
methanol, and phosphoric acid (540:290:170:2). Dissolve 1 gram of sodium 
lauryl sulfate in 1,000 milliliters of this solution, adjust with 2N 
sodium hydroxide to a pH of 3.60.1. Filter through a 
suitable

[[Page 897]]

filter capable of removing particulate matter to 0.5 micron in diameter. 
Degas the mobile phase just prior to its introduction into the 
chromatograph.
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(A) Working standard solution. Dissolve an accurately weighed 
quantity of doxorubicin hydrochloride working standard in mobile phase 
to obtain a solution having a known concentration of 0.1 milligram of 
doxorubicin hydrochloride per milliliter.
    (B) Sample solution. Transfer approximately 20 milligrams of sample, 
accurately weighed, to a 200-milliliter volumetric flask, add mobile 
phase to volume, and mix. This yields a solution containing 0.1 
milligram of doxorubicin hydrochloride per milliliter (estimated).
    (C) Resolution test solution. Use either of the following 
preparation methods:
    (1) To 2 milliliters of a 1.0 milligram per milliliter solution of 
doxorubicin hydrochloride, add 20 microliters of 1N hydrochloric acid. 
Hold for 30 minutes at 95  deg.C in an oil bath.
    (2) Dissolve about 10 milligrams of doxorubicin hydrochloride in 5 
milliliters of water, add 5 milliliters of phosphoric acid, and allow to 
stand for about 30 minutes. Adjust with 2N sodium hydroxide (about 37 
milliliters) to a pH of 2.60.1, add 15 milliliters of 
acetonitrile and 10 milliliters of methanol, mix, and filter. (Note: 
Portions of this solution may be frozen until needed, then thawed and 
mixed before use.)
    (3) The procedures in paragraphs (b)(1)(ii)(C)(1) and 
(b)(1)(ii)(C)(2) of this section generate doxorubicinone, the aglycone 
of doxorubicin. Use this solution to determine the resolution 
requirement for the chromatographic system.
    (iii) System suitability requirements--(A) Asymmetry factor. The 
asymmetry factor (AS (for the doxorubicin peak measured at a 
point 5 percent of the peak height is not less than 0.7 and not more 
than 1.2.
    (B) Efficiency of the column. The absolute column efficiency 
(hr (is satisfactory if it is not greater than 10.0, 
equivalent to 2,500 theoretical plates for a 25-centimeter column of 10-
micrometer particles.
    (C) Resolution. The resolution (R) between the peaks of doxorubicin 
and doxorubicinone (generated in situ) is satisfactory if it is not less 
than 5.5.
    (D) Capacity factor. The capacity factor (k) for doxorubicin is 
satisfactory if it is in the range between 1.0 and 5.0.
    (E) Coefficient of variation. The coefficient of variation (relative 
standard of deviation in percent) of 5 replicate injections is 
satisfactory if it is not more than 1.0 percent. If the system 
suitability parameters have been met, then proceed as described in 
Sec. 436.216(b) of this chapter.
    (iv) Calculations. Calculate the micrograms of doxorubicin 
hydrochloride per milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.281

where:
AU = Area of the doxorubicin hydrochloride peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
AS = Area of the doxorubicin hydrochloride peak in the 
          chromatogram of the doxorubicin hydrochloride working 
          standard;
PS = Doxorubicin hydrochloride activity in the doxorubicin 
          hydrochloride working standard solution in micrograms per 
          milliliter;
CU = Milligrams of the sample per milliliter of sample 
          solution;
m = Percent moisture content of the sample; and
X = Percent solvent residue determined as directed in paragraph (b)(2) 
          of this section.

    (2) Residue solvent (as acetone and alcohol)--(i) Standard 
preparation. Transfer to a 100-milliliter volumetric flask about 200 
milligrams of acetone, 300 milligrams of dehydrated alcohol, and 1,000 
milligrams of dioxane, each accurately weighed, and mix. Dilute with 
water to volume, and mix. Transfer 5.0 milliliters of the resulting 
solution to a 50-milliliter volumetric flask, dilute with water to 
volume, and mix. This solution contains about 0.2 milligram of acetone, 
0.3 milligram of alcohol, and 1 milligram of dioxane per milliliter.

[[Page 898]]

    (ii) Solvent. Transfer about 100 milligrams of dioxane, accurately 
weighed to a 100-milliliter volumetric flask, dilute with water to 
volume, and mix.
    (iii) Test preparation. Dissolve about 200 milligrams of doxorubicin 
hydrochloride sample in 3.0 milliliters of solvent.
    (iv) Chromatographic system (see United States Pharmacopeia (U.S.P.) 
Chromatography (621)). The gas chromatograph is equipped with a flame-
ionization detector and a 4-millimeter X 2-meter column packed with 8-
percent liquid phase G16 (see U.S.P. Chromatographic Reagents--Phases) 
on 100- to 120-mesh support S1AB (potassium hydroxide-washed) (see 
U.S.P. Chromatographic Reagents--Supports). The column is maintained at 
about 60  deg.C, and helium is used as the carrier gas. Adjust the 
column temperature and carrier gas flow rate so that dioxane elutes in 
about 6 minutes. Chromatograph the standard preparation, and record the 
peak responses as directed under procedure; the resolution (R) between 
adjacent peaks is not less than 2.0; the relative standard deviations of 
the ratios of the peak responses of the acetone and dioxane peaks and of 
the alcohol and dioxane peaks for replicate injections is not more than 
4.0 percent; and the tailing factor for the alcohol peak is not more 
than 1.5.
    (v) Procedure. (Note: Use peak areas where peak responses are 
indicated.) Separately inject equal volumes (about 1 microliter) of the 
standard preparation and the test preparation into the chromatograph, 
record the chromatograms, and measure the responses for the major peaks. 
The relative retention times are about 0.2 for acetone, 0.5 for alcohol, 
and 1.0 for dioxane. Calculate the percentage, by weight, of acetone and 
alcohol, respectively, in the sample as follows:

X = Percent acetone or alcohol = 100(CA/
          CD)(DU/WU)(RU/
          RS)

where:
CA = Concentration of acetone or alcohol in the standard 
          preparation in milligrams per milliliter;
CD = Concentration of dioxane in the standard preparation in 
          milligrams per milliliter;
DU = Total quantity of dioxane in the test preparation, in 
          milligrams;
WU = Quantity of doxorubicin hydrochloride taken to prepare 
          the test preparation, in milligrams;
RU = Response ratio of the analyte peak (acetone or alcohol) 
          to the dioxane peak obtained from the test preparation; and
RS = Response ratio of the analyte peak (acetoneoralcohol) to 
          the dioxane peak obtained from the standard preparation.


The total of acetone and alcohol is not greater than 2.5 percent. Use 
the result obtained to calculate the doxorubicin hydrochloride content 
of the sample on the solvent-free basis.
    (3) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 5 milligrams per milliliter.
    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (7) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the doxorubicin hydrochloride working standard.
    (8) Chromatographic purity. Proceed as directed in paragraph (b)(1) 
of this section, except prepare the sample solution by dissolving the 
sample to be tested in mobile phase to obtain a solution containing 
approximately 0.5 milligram of doxorubicin hydrochloride per milliliter. 
Calculate the percentage of impurities as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.282

where:
S = The sum of the responses of the minor component peaks; and
r = The response of the major doxorubicin hydrochloride peak.


The total related impurities detected is not more than 2.0 percent.
[41 FR 14184, Apr. 2, 1976; 41 FR 15844, Apr. 15, 1976, as amended at 42 
FR 43063, Aug. 26, 1977; 43 FR 44836, Sept. 29, 1978; 47 FR 9396, Mar. 
5, 1982; 47 FR 23710, June 1, 1982; 50 FR 19676, May 10, 1985; 53 FR 
37292, Sept. 26, 1988; 59 FR 9639, Mar. 1, 1994]



Sec. 450.30  Idarubicin hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality,

[[Page 899]]

and purity. Idarubicin hydrochloride is the monohydrochloride salt of 
5,12-Naphthacenedione,9-acetyl-7-[(3-amino-2,3,6-trideoxy--L-
lyxo-hexopyranosyl)oxy]-7,8,9,10-tetrahydro-6,9,11-trihydroxy-(7S-cis). 
It is an orange-red powder. It is so purified and dried that:
    (i) Its idarubicin hydrochloride content is not less than 960 
micrograms and not more than 1,030 micrograms of idarubicin 
hydrochloride per milligram on the anhydrous basis.
    (ii) Its moisture content is not more than 5.0 percent.
    (iii) The pH of an aqueous solution containing 5 milligrams per 
milliliter is not less than 5.0 and not more than 6.5.
    (iv) It is crystalline.
    (v) The level of any individual impurity detected by high-
performance liquid chromatography (HPLC) assay is not more than 1.0 
percent.
    (vi) The total of all detected impurities is not more than 3.0 
percent.
    (vii) It passes the identity test for idarubicin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for idarubicin 
hydrochloride content, solvent residues, moisture, pH, crystallinity, 
related individual thin-layer chromatography and HPLC impurities, total 
impurities, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 14 packages, each containing approximately 40 
milligrams.
    (b) Tests and methods of assay. Idarubicin hydrochloride is toxic. 
It must be handled with care in the laboratory. Transfer all dry powders 
into a suitable hood while wearing rubber gloves. Avoid inhaling fine 
particles of powder. Solutions should not be pipetted by mouth. If the 
substance contacts the skin, wash with soap and water. Dispose of all 
waste material by dilution with large volumes of dilute sodium 
hypochlorite (bleach) solution.
    (1) Potency (HPLC). Proceed as directed in Sec. 436.216 of this 
chapter, using ambient temperature, an ultraviolet detection system 
operating at a wavelength of 254 nanometers, a 4.6-millimeter by 25-
centimeter column packed with microparticulate (5 to 10 micrometers in 
diameter) packing material such as trimethylsilane chemically bonded to 
porous silica, a flow rate of not more than 2.0 milliliters per minute, 
and a known injection volume of between 10 and 20 microliters. The 
retention time for idarubicin hydrochloride is between 14 and 16 
minutes. The retention time for the resolution compound 4-
demethoxydaunorubicinone (generated in situ) is between 6 and 9 minutes. 
Mobile phase, diluent, working standard and sample solutions, resolution 
test solution, system suitability requirements, and calculations are as 
follows:
    (i) Mobile phase. Prepare a suitably sized quantity of a mixture of 
water, acetonitrile, and methanol (540:290:170). Dissolve 1 gram of 
sodium lauryl sulfate and 2 milliliters of 85 percent phosphoric acid 
per liter of this solution. Adjust with 2 N sodium hydroxide to a pH of 
3.60.1. Filter through a suitable filter capable of removing 
particulate matter to 0.5 micron in diameter. Degas the mobile phase 
just prior to its introduction into the chromatograph.
    (ii) Diluent. Prepare as mobile phase, excluding the sodium lauryl 
sulfate.
    (iii) Preparation of working standard solution. Dissolve an 
accurately weighed quantity of idarubicin hydrochloride working standard 
in diluent to obtain a solution having a known concentration of 0.5 
milligram of idarubicin hydrochloride per milliliter.
    (iv) Sample solution. Transfer approximately 50 milligrams of 
sample, accurately weighed, to a 100-milliliter volumetric flask, add 
diluent to volume, and mix. This yields a solution containing 0.5 
milligram of idarubicin hydrochloride per milliliter (estimated).
    (v) Resolution test solution. To 2 milliliters of a 1.0 milligram 
per milliliter aqueous solution of idarubicin hydrochloride, add 20 
microliters of 1 N hydrochloric acid. Hold for 30 minutes at 95 C in an 
oil bath. This procedure generates the aglycone of idarubicin, 4-
demethoxydaunorubicinone. Transfer 1.0 milliliter of this solution to a 
10-milliliter volumetric flask, add diluent

[[Page 900]]

to volume, and mix. Use this solution to determine the resolution 
requirement for the chromatographic system.
    (vi) System suitability requirements--(A) Asymmetry factor. The 
asymmetry factor (AS), measure data point 5 percent of the 
peak height from the baseline, is satisfactory if it is not less than 
0.85 and not more than 1.1.
    (B) Efficiency of the column. The absolute efficiency 
(hr) is satisfactory if it is not more than 10.0 for the 
idarubicin hydrochloride peak, equivalent to 4,500 theoretical plates 
for a 25-centimeter column of 6-micrometer particles.
    (C) Resolution factor. The resolution factor (RS) between 
the peak for idarubicin and 4-demethoxydaunorubicinone (generated 
insitu) is satisfactory if it is not less than 9.5.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SRin percent of 5 replicate 
injections) is satisfactory if it is not more than 2.0 percent.
    (E) Capacity factor. The capacity factor (k') for idarubicin 
hydrochloride is satisfactory if it is not less than 5 and not more than 
15. If the system suitability parameters have been met, proceed as 
described in Sec. 436.216(b) of this chapter.
    (vii) Calculations. Calculate the micrograms of idarubicin 
hydrochloride per milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.072

where:

    AU=Area of the idarubicin hydrochloride peak in the 
chromatogram of the sample (at a retention time equal to that observed 
for the standard);
    AS=Area of the idarubicin hydrochloride peak in the 
chromatogram of the idarubicin hydrochloride working standard;
    PS=Idarubicin hydrochloride activity in the idarubicin 
hydrochloride working standard solution in micrograms per milliliter;
    CU=Milligrams of idarubicin hydrochloride sample per 
milliliter of sample solution;
    m=Percent moisture content of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 5 milligrams per milliliter.
    (4) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (5) HPLC impurities. Proceed as directed in paragraph (b)(1) of this 
section. Calculate the percentage of impurities as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.073

where:
    Ai=Area of the individual impurity peak;
    A=The sum of areas of all peaks minus the area due to the idarubicin 
hydrochloride peak and solvent peak; and
    At=The sum of areas of all peaks in the chromatogram 
excluding the solvent peak.

    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 1.0 percent potassium bromide disc prepared as directed in 
Sec. 436.211(b)(1).
[58 FR 26664, May 4, 1993]



Sec. 450.40  Plicamycin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Plicamycin is a yellow compound and is so 
purified and dried that:
    (i) Its plicamycin content is not less than 900 micrograms of 
plicamycin per milligram calculated on an anhydrous basis.
    (ii) Its loss on drying is not more than 8 percent.
    (iii) Its pH in an aqueous solution containing 0.5 milligram per 
milliliter is not less than 4.5 nor more than 5.5.
    (iv) Its absorptivity on the anhydrous basis at the absorption 
maximum of 278 millimicrons is 100plus-minus5 percent of that 
of the plicamycin standard similarly treated.
    (v) It gives a positive result to the identity tests for plicamycin.
    (vi) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. In addition, each package 
shall bear on its label the statement ``Store below 10 deg. C. (50 deg. 
F.)''.

[[Page 901]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for plicamycin content, 
loss on drying, pH, absorptivity, identity, and crystallinity.
    (ii) Samples required on the batch: 2 packages, each containing not 
less than 100 milligrams; and 3 packages, each containing not less than 
50 milligrams.
    (b) Tests and methods of assay. Plicamycin is more toxic than the 
average drug and must be handled with care in the laboratory. Avoid 
inhaling fine particles of powder. If the substance contacts the skin, 
wash with soap and water. Solutions should not be pipetted by mouth. 
Plicamycin is hygroscopic and care should be exercised during storage 
and weighing samples. Samples should be stored at 10 deg. C. or less in 
a sealed, light-resistant container with a desiccant. Dispose of all 
waste material by dilution with larger volumes of trisodium phosphate 
solution.
    (1) Plicamycin content. Proceed as directed in Sec. 436.341 of this 
chapter, preparing the sample and calculating the plicamycin content as 
follows:
    (i) Preparation of sample solution. Place approximately 5 milligrams 
of the sample, accurately weighed, into a 50-milliliter, amber 
volumetric flask and dilute to volume with mobile phase and mix.
    (ii) Calculations. Calculate the micrograms of plicamycin per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.283

where:

Au = Area of the plicamycin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As = Area of the plicamycin peak in the chromatogram of the 
          plicamycin working standard;
Ps = Plicamycin activity in the plicamycin working standard 
          solution in micrograms per milliliter;
Cu = Milligrams of sample per milliliter of sample solution; 
          and
m = Percent moisture content of the sample.

    (2) Loss on drying. Proceed as directed in Sec. 436.200(g) of this 
chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 0.5 milligram of plicamycin per 
milliliter. Allow the solution to remain in contact with the electrodes 
until a steady reading is obtained or for 5 minutes.
    (4) Absorptivity. Determine the absorbance of the sample and 
standard solutions in the following manner: Dissolve approximately 10 
milligrams each of the sample and standard (dried as described in 
Sec. 436.200(g) of this chapter), accurately weighed, in 50 milliliters 
of absolute methanol. Transfer 5-milliliter portions into 100-milliliter 
volumetric flasks and dilute to volume with 0.01N hydrochloric acid in 
methanol prepared by diluting 20 milliliters of 0.5N aqueous 
hydrochloric acid to 1 liter with absolute methanol. Using a suitable 
spectrophotometer and 0.01N hydrochloric acid in methanol as the blank, 
scan the absorption spectrum between the wavelengths of 220 millimicrons 
and 400 millimicrons. Determine the absorbance of each solution at the 
absorption maximum near 278 millimicrons. Determine the percent 
absorptivity of the sample relative to the absorptivity of the standard 
using the following calculation:

[GRAPHIC] [TIFF OMITTED] TR01JA93.284


[[Page 902]]


    (5) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the plicamycin working standard.
    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19145, May 30, 1974, as amended at 49 FR 5097, Feb. 10, 1984; 49 
FR 24018, June 11, 1984]



Sec. 450.45  Mitomycin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Mitomycin is 7-amino-9a-methoxymitosane. 
It is a blue-violet compound that is soluble in water, methanol, 
acetone, butyl acetate, and cyclohexanone. It is so purified and dried 
that:
    (i) Its potency is not less than 900 micrograms per milligram.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 5 percent.
    (iv) Its pH in a solution containing 5 milligrams per milliliter is 
not less than 6.0 and not more than 8.0.
    (v) When calculated on the anhydrous basis, its absorptivity at 357 
nanometers is not less than 95 percent and not more than 105 percent of 
that of the mitomycin working standard similarly treated.
    (vi) It gives a positive identity test for mitomycin.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, absorptivity, identity, and crystallinity.
    (ii) Samples required: Five packages, each containing approximately 
100 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 1 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to obtain a stock solution of 
convenient concentration. Further dilute an aliquot of the stock 
solution with solution 1 to the reference concentration of 1.0 microgram 
of mitomycin per milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 5 milligrams per milliliter.
    (5) Absorptivity. Determine the absorbance of the sample and 
standard solution in the following manner: Place an accurately weighed 
portion of approximately 25 milligrams of mitomycin into a 50-milliliter 
volumetric flask. Dissolve and dilute to volume with absolute methanol. 
Further dilute an aliquot with absolute methanol to 0.005 milligram of 
mitomycin per milliliter. Using a suitable spectrophotometer equipped 
with a 1-centimeter quartz cell and absolute methanol as the blank, 
determine the absorbance of the sample and standard solutions at 357 
nanometers. Calculate the percent relative absorptivity as follows:

[GRAPHIC] [TIFF OMITTED] TR01JA93.285

where:
m=percent moisture in the sample.

    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation method described in paragraph (b)(2) of 
that section.

[[Page 903]]

    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19145, May 30, 1974, as amended at 50 FR 19920, May 13, 1985]



                          Subpart B  [Reserved]



                   Subpart C--Injectable Dosage Forms



Sec. 450.210  Sterile bleomycin sulfate.

    The requirements for certification and the tests and methods of 
assay for sterile bleomycin sulfate packaged for dispensing are 
described in Sec. 450.10a.
[40 FR 52006, Nov. 7, 1975]



Sec. 450.220  Dactinomycin for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Dactinomycin for injection is a dry 
mixture of dactinomycin and mannitol. Each container contains 0.5 
milligram of dactinomycin. Its dactinomycin content is satisfactory if 
it is not less than 90 percent and not more than 120 percent of the 
number of milligrams of dactinomycin that it is represented to contain. 
It is sterile. It is nonpyrogenic. Its loss on drying is not more than 
4.0 percent. Its pH is not less than 5.5 and not more than 7.5. The 
dactinomycin used conforms to the standards prescribed by 
Sec. 450.20(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter, and in addition each package 
shall bear on its label or labeling, as hereinafter indicated, the 
following:
    (i) On the outside wrapper or container the statement ``Protect from 
light and excessive heat''.
    (ii) On the outside wrapper or container and the immediate container 
the statement ``For hospitalized patients only''.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The dactinomycin used in making the batch for dactinomycin 
content, loss on drying, absorptivity, crystallinity, and identity.
    (b) The batch for dactinomycin content, sterility, pyrogens, loss on 
drying, and pH.
    (ii) Samples required:
    (a) The dactinomycin used in making the batch: 10 containers each 
containing not less than 40 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers.
    (b) Tests and methods of assay. Dactinomycin is toxic and corrosive. 
It must be handled with care in the laboratory. Transfer all dry powders 
in a suitable hood, while wearing rubber gloves. Avoid inhaling fine 
particles of the powder. Do not pipette by mouth. If any of the 
substance contacts the skin, wash copiously with soap and water. Dispose 
of all waste material by dilution with large volumes of trisodium 
phosphate solution.
    (1) Dactinomycin content. Proceed as directed in Sec. 436.331 of 
this chapter, except prepare the sample solution and calculate the 
dactinomycin content as follows:
    (i) Sample solution. Reconstitute the vial with 2.0 milliliters of 
mobile phase. Shake well and filter if necessary.
    (ii) Calculations. Calculate the dactinomycin content of the vial as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.286

where:
Au=Area of the dactinomycin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the dactinomycin peak in the chromatogram of the 
          dactinomycin working standard;
Ps=Dactinomycin activity in the dactinomycin working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use the entire contents of each of the immediate containers tested.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
preparing the

[[Page 904]]

sample for test as follows: Use a sufficient number of containers to 
yield 3 milligrams of dactinomycin. Reconstitute by adding 1.1 
milliliters of sterile water for injection to each container. 
Aseptically pool the resultant solutions from each container. Dilute an 
accurately measured portion with sufficient diluent 1 to give a 
concentration of 0.2 milligram of dactinomycin per milliliter.
    (4) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (5) pH. Reconstitute as directed in the labeling and proceed as 
directed in Sec. 436.202 of this chapter.
[39 FR 19145, May 30, 1974, as amended at 44 FR 10379, Feb. 20, 1979; 46 
FR 16685, Mar. 13, 1981; 46 FR 46313, Sept. 18, 1981; 49 FR 6093, Feb. 
17, 1984; 49 FR 15074, Apr. 17, 1984; 49 FR 24018, June 11, 1984; 50 FR 
1504, Jan. 11, 1985; 50 FR 19676, May 10, 1985]



Sec. 450.222  Daunorubicin hydrochloride for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Daunorubicin hydrochloride for injection 
is a freeze-dried powder whose components are daunorubicin hydrochloride 
and mannitol. Its potency is satisfactory if it is not less than 90 
percent and not more than 115 percent of the number of milligrams of 
daunorubicin that it is represented to contain. It is sterile. It is 
nonpyrogenic. It contains no depressor substances. Its moisture content 
is not more than 3.0 percent. When reconstituted as directed in the 
labeling, its pH is not less than 4.5 and not more than 6.5. It passes 
the identity test. The daunorubicin hydrochloride used conforms to the 
standards prescribed by Sec. 450.22(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The daunorubicin hydrochloride used in making the batch for 
potency, moisture, pH, crystallinity, and identity.
    (b) The batch for potency, sterility, pyrogens, depressor 
substances, moisture, pH, and identity.
    (ii) Samples required:
    (a) The daunorubicin hydrochloride used in making the batch: 14 
packages, each containing approximately 40 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 34 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Daunorubicin hydrochloride is toxic. 
It must be handled with care in the laboratory. Solutions should not be 
pipetted by mouth. Transfer all dry powders in a suitable hood. Wear 
rubber gloves, protective gowns, head coverings, and protective eye 
goggles when handling dry powders. If the substance contacts the skin, 
wash with soap and water. Dispose of all waste material by dilution with 
larger volumes of sodium hypochlorite solution.
    (1) Daunorubicin content (high-pressure liquid chromatography). 
Proceed as directed in Sec. 436.322 of this chapter, preparing the 
sample and standard solutions and calculating the daunorubicin content 
as follows:
    (i) Preparation of working standard solution. Accurately weigh 
approximately 25 milligrams of the daunorubicin working standard and 
dissolve in 25 milliliters of the internal standard solution prepared as 
directed in Sec. 436.322(b)(3) of this chapter.
    (ii) Preparation of sample solution. Prepare the sample solution by 
rinsing the contents of the vial into an appropriate-sized volumetric 
flask with a sufficient amount of internal standard solution prepared as 
directed in Sec. 436.322(b)(3) of this chapter, to obtain a 
concentration of 1.0 milligram of daunorubicin per milliliter.
    (iii) Calculations. Calculate the daunorubicin content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.287
    
where:

[[Page 905]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.288


Ws = Weight of the daunorubicin working standard in 
          milligrams;
V = Volume in milliliters of the internal standard solution added to the 
          vials;
P = Potency of the daunorubicin working standard in micrograms per 
          milligram.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 2.25 milligrams of daunorubicin per 
milliliter.
    (4) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the sample preparation method described in paragraph (d)(4) of 
that section.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the sample obtained after reconstituting the drug as directed in the 
labeling.
    (7) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the daunorubicin working standard.
[45 FR 75198, Nov. 14, 1980, as amended at 50 FR 47214, Nov. 15, 1985]



Sec. 450.224  Doxorubicin hydrochloride injectable dosage forms.



Sec. 450.224a  Doxorubicin hydrochloride for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Doxorubicin hydrochloride for injection 
is a freeze-dried powder whose components are doxorubicin hydrochloride 
and lactose. It may also contain methylparaben. Its doxorubicin 
hydrochloride content is satisfactory if it is not less than 90 percent 
and not more than 115 percent of the number of milligrams of doxorubicin 
hydrochloride that it is represented to contain. It is sterile. It 
contains not more than 2.2 U.S.P. endotoxin units per milligram of 
doxorubicin hydrochloride. Its moisture content is not more than 4.0 
percent. When reconstituted as directed in the labeling, its pH is not 
less than 4.5 and not more than 6.5. It passes the identity test. The 
doxorubicin hydrochloride used conforms to the standards prescribed by 
Sec. 450.24(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The doxorubicin hydrochloride used in making the batch for 
doxorubicin hydrochloride content, residue solvents, depressor 
substances, moisture, pH, crystallinity, identity, and total related 
impurities.
    (b) The batch for doxorubicin hydrochloride content, sterility, 
bacterial endotoxins, moisture, pH, and identity.
    (ii) Samples required:
    (a) The doxorubicin hydrochloride used in making the batch: 14 
packages, each containing approximately 40 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 34 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Doxorubicin hydrochloride is toxic. 
It must be handled with care in the laboratory. Solutions should not be 
pipetted by mouth. Transfer all dry powders in a suitable hood while 
wearing rubber gloves. If the substance contacts the skin, wash with 
soap and water. Dispose of all waste material by dilution with large 
volumes of sodium hypochlorite (bleach) solution.

[[Page 906]]

    (1) Doxorubicin hydrochloride content (high-performance liquid 
chromatography). Proceed as directed in Sec. 450.24(b)(1), preparing the 
sample solution and calculating the doxorubicin hydrochloride content as 
follows:
    (i) Sample solution. Prepare the sample solution by rinsing the 
contents of the vial into an appropriate sized volumetric flask with 
sufficient mobile phase to obtain a concentration of 0.1 milligram of 
doxorubicin hydrochloride per milliliter (estimated).
    (ii) Calculations. Calculate the doxorubicin hydrochloride content 
per vial as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.289

where:

AU = Area of the doxorubicin hydrochloride peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
AS = Area of the doxorubicin hydrochloride peak in the 
          chromatogram of the doxorubicin hydrochloride working 
          standard;
PS = Doxorubicin hydrochloride activity in the doxorubicin 
          hydrochloride working standard solution in micrograms per 
          milliliter; and
d = Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Bacterial endotoxins. Proceed as directed in the United States 
Pharmacopeia (U.S.P.) Bacterial Endotoxin Test, using a solution of 
doxorubicin hydrochloride for injection containing 1.1 milligrams of 
doxorubicin hydrochloride per milliliter. The specimen under test 
contains not more than 2.2 U.S.P. endotoxin units per milligram of 
doxorubicin hydrochloride.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the sample preparation method described in paragraph (d)(4) of 
that section.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the sample obtained after reconstituting the drug as directed in the 
labeling, except in lieu of saline use distilled water.
    (7) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section, compares 
qualitatively to that of the doxorubicin hydrochloride working standard.
[41 FR 14185, Apr. 2, 1976, as amended at 43 FR 44837, Sept. 29, 1978; 
46 FR 60568, Dec. 11, 1981; 50 FR 19676, May 10, 1985. Redesignated at 
53 FR 37292, Sept. 26, 1988; 59 FR 9641, Mar. 1, 1994]



Sec. 450.224b  Doxorubicin hydrochloride injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Doxorubicin hydrochloride injection is an 
aqueous solution of doxorubicin hydrochloride in an isosmotic diluent. 
Each milliliter contains doxorubicin hydrochloride equivalent to 2 
milligrams of doxorubicin hydrochloride. Its doxorubicin hydrochloride 
content is satisfactory if it is not less than 90 percent and not more 
than 115 percent of the number of milligrams it is represented to 
contain. It is sterile. It contains not more than 2.2 U.S.P. endotoxin 
units per milligram of doxorubicin hydrochloride. Its pH is not less 
than 2.5 and not more than 3.5. It passes the identity test. The 
doxorubicin hydrochloride used conforms to the standards prescribed by 
Sec. 450.24(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The doxorubicin hydrochloride used in making the batch for 
doxorubicin hydrochloride content, residue solvents, depressor 
substances, moisture, pH, crystallinity, identity, and total related 
impurities.
    (B) The batch for doxorubicin hydrochloride content, sterility, 
bacterial endotoxins, pH, and identity.
    (ii) Samples required:
    (A) The doxorubicin hydrochloride used in making the batch: 14 
packages, each containing approximately 40 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 34 immediate 
containers.

[[Page 907]]

    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Doxorubicin hydrochloride is toxic. 
It must be handled with care in the laboratory. Solutions should not be 
pipetted by mouth. Transfer all dry powders in a suitable hood while 
wearing rubber gloves. If the substance contacts the skin, wash with 
soap and water. Dispose of all waste material by dilution with large 
volumes of sodium hypochlorite (bleach) solution.
    (1) Doxorubicin hydrochloride content (high-performance liquid 
chromatography). Proceed as directed in Sec. 450.24(b)(1), preparing the 
sample solution and calculating the doxorubicin hydrochloride content as 
follows:
    (i) Sample solution. Dilute an accurately measured volume of sample 
equivalent to not less than 2 milligrams of doxorubicin hydrochloride, 
quantitatively with mobile phase to obtain a solution containing 0.1 
milligram of doxorubicin hydrochloride per milliliter (estimated).
    (ii) Calculations. Calculate the milligrams of doxorubicin 
hydrochloride per milliliter of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.290

where:
AU = Area of the doxorubicin hydrochloride peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
AS = Area of the doxorubicin hydrochloride peak in the 
          chromatogram of the doxorubicin hydrochloride working 
          standard;
PS = Doxorubicin hydrochloride activity in the doxorubicin 
          hydrochloride working standard solution in micrograms per 
          milliliter; and
d = Dilution factor of the sample.

    (2) [Reserved]
    (3) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (4) Bacterial endotoxins. Proceed as directed in the United States 
Pharmacopeia (U.S.P.) Bacterial Endotoxin Test, using a test solution 
prepared by diluting doxorubicin hydrochloride injection with sterile 
water for injection to obtain a concentration of 1.1 milligrams of 
doxorubicin hydrochloride per milliliter. The specimen under test 
contains not more than 2.2 U.S.P. endotoxin units per milligram of 
doxorubicin hydrochloride.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (6) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section, compares 
qualitatively to that of the doxorubicin hydrochloride working standard.
[53 FR 37292, Sept. 26, 1988, as amended at 59 FR 9641, Mar. 1, 1994]



Sec. 450.230  Idarubicin hydrochloride for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Idarubicin hydrochloride for injection is 
a lyophilized mixture of idarubicin hydrochloride and lactose. Its 
idarubicin hydrochloride content is satisfactory if it is not less than 
90 percent and not more than 110 percent of the number of milligrams of 
idarubicin hydrochloride that it is represented to contain. It is 
sterile. It contains not more than 8.93 U.S.P. endotoxin units per 
milligram of idarubicin hydrochloride. Its moisture content is not more 
than 4.0 percent. When reconstituted as directed in the labeling, its pH 
is not less than 5.0 and not more than 7.0. It passes the identity test. 
The idarubicin hydrochloride used conforms to the standards prescribed 
by Sec. 450.30(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The idarubicin hydrochloride used in making the batch for 
idarubicin hydrochloride content, solvent residues, moisture, pH, 
crystallinity, related individual thin layer chromatography and high-
performance liquid chromatography (HPLC) impurities, total impurities, 
and identity.

[[Page 908]]

    (B) The batch for idarubicin hydrochloride content, sterility, 
bacterial endotoxins, moisture, pH, and identity.
    (ii) Samples required if requested by the Director, Center for Drug 
Evaluation and Research:
    (A) The idarubicin hydrochloride used in making the batch: 14 
packages, each containing approximately 40 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 34 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Idarubicin hydrochloride is toxic. 
It must be handled with care in the laboratory. Transfer all dry powders 
into a suitable hood while wearing rubber gloves. Avoid inhaling fine 
particles of powder. Solutions should not be pipetted by mouth. If the 
substance contacts the skin, wash with soap and water. Dispose of all 
waste material by dilution with large volumes of dilute sodium 
hypochlorite (bleach) solution.
    (1) Idarubicin hydrochloride content (HPLC). Proceed as directed in 
Sec. 450.30(b)(1), preparing the sample solution and calculating the 
idarubicin hydrochloride as follows:
    (i) Sample solution. Prepare the sample solution by rinsing the 
contents of the vial into an appropriate-sized volumetric flask with 
sufficient diluent to obtain a concentration of 0.5 milligram of 
idarubicin hydrochloride per milliliter (estimated).
    (ii) Calculations. Calculate the idarubicin hydrochloride content 
per vial as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.291

where:

    AU=Area of the idarubicin hydrochloride peak in the 
chromatogram of the sample (at a retention time equal to that observed 
for the standard);
    AS=Area of the idarubicin hydrochloride peak in the 
chromatogram of the idarubicin hydrochloride working standard;
    PS=Idarubicin hydrochloride activity in the idarubicin 
hydrochloride working standard solution in micrograms per milliliter; 
and
    d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in Sec. 436.20(e)(1).
    (3) Bacterial endotoxins. Proceed as directed in the U.S.P. Bacteria 
endotoxin test. The specimen under test contains not more than 8.93 
U.S.P. endotoxin units per milligram of idarubicin hydrochloride.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the sample preparation method described in Sec. 436.201(d)(4).
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the sample obtained after reconstituting the drug as directed in the 
labeling, except use distilled water instead of saline.
    (6) Identity. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the idarubicin hydrochloride working standard.
[58 FR 26665, May 4, 1993]



Sec. 450.240  Plicamycin for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Plicamycin for injection is a dry mixture 
of plicamycin and mannitol with or without a suitable buffer substance. 
Each immediate container contains 2.5 milligrams of plicamycin. Its 
plicamycin content is satisfactory if it contains not less than 90 
percent and not more than 110 percent of the number of milligrams of 
plicamycin that it is representated to contain. It is sterile. It is 
nonpyrogenic. Its moisture content is not more than 2.0 percent. It 
contains no depressor substances. Its pH when reconstituted as directed 
in the labeling is not less than 5.0 and not more than 7.5. It passes 
the identity test for plicamycin. The plicamycin used conforms to the 
standards prescribed by Sec. 450.40(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. In addition, each package 
shall bear on its label or labeling the following as indicated:
    (i) On the outside wrapper or container the statement ``Store below 
10 deg. C. (50 deg. F.)''.

[[Page 909]]

    (ii) On the outside wrapper or container and on the immediate 
container the statement ``Mandatory: Before using read enclosed 
professional information carefully for dosage instructions and 
warnings''.
    (iii) On the outside wrapper or container the statement ``Warning: 
For hospital use only. To be used under direct supervision of a 
physician''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The plicamycin used in making the batch for plicamycin content, 
loss on drying, absorptivity, pH, identity, and crystallinity.
    (b) The batch for plicamycin content, sterility, pyrogens, moisture, 
Ph, depressor substances, and identity.
    (ii) Samples required:
    (a) The plicamycin used in making the batch: 3 packages, each 
containing not less than 50 milligrams; and 2 packages, each containing 
not less than 100 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 21 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Plicamycin is more toxic than the 
average drug and must be handled with care in the laboratory. Avoid 
inhaling fine particles of powder. If the substance contacts the skin, 
wash with soap and water. Plicamycin is hygroscopic and care should be 
exercised during storage and weighing of samples. Dispose of all waste 
materials by dilution with larger volumes of trisodium phosphate 
solution. The samples should be stored at 10 deg. C. or less in a sealed 
light-resistant container with a dessicant. Solutions should not be 
pipetted by mouth.
    (1) Plicamycin content. Proceed as directed in Sec. 436.341 of this 
chapter, except prepare the sample solution and calculate the plicamycin 
content as follows:
    (i) Preparation of sample solution. Place approximately 5 milligrams 
of the sample, accurately weighed, into a 50-milliliter, amber 
volumetric flash and dilute to volume with mobile phase and mix.
    (ii) Calculations. Calculate the plicamycin content of the vial as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.292

where:

Au=Area of the plicamycin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the plicamycin peak in the chromatogram of the 
          plicamycin working standard;
Ps=Plicamycin activity in the plicamycin working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use the entire contents of each of the immediate containers tested.
    (3) Pyrogens. Reconstitute the sample as directed in the labeling 
and proceed as directed in Sec. 436.32(b) of this chapter, using a 
solution containing 50 micrograms of plicamycin per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the total contents of three to five vials.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling. Allow the solution 
to remain in contact with the electrodes until a steady reading is 
obtained or for 5 minutes.
    (6) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (7) Thin layer chromatography identity test for plicamycin--(i) 
Equipment--(a) Plates. Use 20 by 20 centimeter or 15 by 20 centimeter 
thin layer chromatographic plates coated with Silica Gel Mixture, 
Chromatographic, U.S.P., to a thickness of 250 microns. Activate the 
plates by heating at 110 deg. C. for 75 minutes. Place the plates in a 
desiccator until cooled to room temperature. Plates may be stored in a 
desiccator for 7 days.
    (b) Chamber (chromatographic). A suitable chamber, equipped for thin 
layer chromatography.

[[Page 910]]

    (ii) Preparations of solutions--(a) Solvent. Mix reagent grade 
chloroform with reagent grade absolute methanol in volumetric 
proportions of 1:1.
    (b) Spray A. Mix 50 milliliters of freshly prepared 1.0 percent 
ferric chloride in water (weight per volume), just before spraying, with 
50 milliliters of freshly prepared 1.0 percent potassium ferricyanide in 
water (weight per volume).
    (c) Spray B. Dissolve 2.28 grams of periodic acid in 100 milliliters 
of water. Dilute one volume of this periodic solution with 10 volumes of 
acetone.
    (d) Spray C. Dissolve 184 milligrams of benzidine in a solution of 
0.6 milliliter of acetic acid, 4.4 milliliters of water, and 95 
milliliters of acetone.
    (iii) Preparation of spotting solutions--(a) Plicamycin standard 
solution. Weigh 5 milligrams of plicamycin working standard and dissolve 
in 10 milliliters of methanol. Use the solution the same day it is 
prepared.
    (b) Plicamycin for injection sample solution. Dilute with methanol 
to a concentration of 0.5 milligram of plicamycin per milliliter. 
Centrifuge and use the supernatant for spotting.
    (c) Mannitol reference solution. Suspend 100 milligrams of mannitol 
in 5 milliliters of methanol. Centrifuge and use the supernatant for 
spotting.
    (iv) Procedure. Fill the chamber to a depth of 0.6 centimeter with 
freshly prepared solvent. Spot duplicate plates as follows: On a line 
2.5 centimeters from the base of the silica gel plate, and at intervals 
of 2.0 centimeters, spot 100 microliters (in four 25-microliter 
aliquots) of the standard solution, the sample solution, and the 
mannitol reference solution. Allow each aliquot to dry before applying 
subsequent volumes. After all spots are thoroughly dry, place the silica 
gel plates in the chromatographic chamber and develop by the ascending 
technique for approximately 60 minutes. Allow several minutes for the 
plates to air dry. On one plate, locate and record the position of 
fluorescent spots by examining under long wave ultraviolet light. Apply 
spray A and record the position of blue spots on the yellow-green 
background. On the other plate, locate the mannitol by first applying 
spray B, followed by spray C. The spots appearing white are mannitol. 
Measure the distance the solvent front traveled from the starting line 
and the distance the fluorescent spots are from the starting line. 
Calculate the Rf value by dividing the latter by the former. 
The plicamycin standard should have an Rf value of 0.7. If 
the standard has an Rf value greater than 0.8, the mobility 
of the standard may be decreased by increasing the ratio of the 
chloroform to methanol in the solvent to 3:2 or 3:1. Plicamycin appears 
as a single major component with the same Rf value as the 
plicamycin standard. It may show trace components at Rf 
values of about 0.5 and 0.4, and at the origin, which shall not be more 
intense than those shown by the plicamycin standard.
[39 FR 19145, May 30, 1974, as amended at 40 FR 1512, Jan. 8, 1975; 46 
FR 60568, Dec. 11, 1981; 47 FR 9396, Mar. 5, 1982; 48 FR 11427, Mar. 18, 
1983; 49 FR 5097, Feb. 10, 1984; 49 FR 24019, June 11, 1984; 50 FR 
19676, May 10, 1985]



Sec. 450.245  Mitomycin for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Mitomycin for injection is a dry mixture 
of mitomycin and mannitol. Its potency is satisfactory if it contains 
not less than 90 percent and not more than 120 percent of the number of 
milligrams of mitomycin that it is represented to contain. It is 
sterile. It is nonpyrogenic. It contains no depressor substances. Its 
moisture content is not more than 5 percent. Its pH, when reconstituted 
as directed in the labeling, is not less than 6.0 and not more than 8.0. 
It passes the identity test for mitomycin. The mitomycin used conforms 
to the standards prescribed by Sec. 450.45(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The mitomycin used in making the batch for potency, moisture, 
pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, depressor 
substances, moisture, pH, and identity.

[[Page 911]]

    (ii) Samples required:
    (a) The mitomycin used in making the batch: Five packages, each 
containing approximately 100 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 25 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Using a suitable hypodermic 
needle and syringe, remove all of the withdrawable contents from each 
container if it is represented as a single dose container; or if the 
labeling specifies the amount of potency in a given volume of the 
resultant preparation, remove an accurately measured representative 
portion from each container. Dilute the solution thus obtained with 
sufficient 1 percent potassium phosphate buffer, pH 6.0 (solution 1), to 
give a stock solution of convenient concentration. Further dilute the 
stock solution with solution 1 to the reference concentration of 1 
microgram of mitomycin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 0.5 milligram of mitomycin per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this chapter using 
the drug reconstituted as directed in the labeling.
    (8) Identity. Proceed as directed in Sec. 436.310 of this chapter.
[39 FR 19145, May 30, 1974, as amended at 46 FR 60568, Dec. 11, 1981; 50 
FR 19920, May 13, 1985]



PART 452--MACROLIDE ANTIBIOTIC DRUGS--Table of Contents




                          Subpart A--Bulk Drugs

Sec.
452.10  Erythromycin.
452.15  Erythromycin estolate.
452.25  Erythromycin ethylsuccinate.
452.25a  Sterile erythromycin ethylsuccinate.
452.30a  Sterile erythromycin gluceptate.
452.32a  Sterile erythromycin lactobionate.
452.35  Erythromycin stearate.
452.50  Clarithromycin.
452.60  Azithromycin.
452.75  Troleandomycin.

                      Subpart B--Oral Dosage Forms

452.110  Erythromycin oral dosage forms.
452.110a  Erythromycin tablets.
452.110b  Erythromycin enteric-coated tablets.
452.110c  Erythromycin capsules.
452.110d  Erythromycin particles in tablets.
452.115  Erythromycin estolate oral dosage forms.
452.115a  Erythromycin estolate tablets.
452.115b  Erythromycin estolate capsules.
452.115c  Erythromycin estolate oral suspension.
452.115d  Erythromycin estolate for oral suspension.
452.115e  Erythromycin estolate for pediatric drops.
452.115f  Erythromycin estolate chewable tablets.
452.115g  Erythromycin estolate and sulfisoxazole acetyl oral 
          suspension.
452.125  Erythromycin ethylsuccinate oral dosage forms.
452.125a  Erythromycin ethylsuccinate chewable tablets.
452.125b  Erythromycin ethylsuccinate oral suspension.
452.125c  Erythromycin ethylsuccinate for oral suspension.
452.125d  Erythromycin ethylsuccinate tablets.
452.125e  Erythromycin ethylsuccinate-sulfisoxazole acetyl for oral 
          suspension.
452.135  Erythromycin stearate oral dosage forms.
452.135a  Erythromycin stearate tablets.
452.135b  Erythromycin stearate oral suspension.
452.135c  Erythromycin stearate for oral suspension.
452.150  Clarithromycin oral dosage forms.
452.150a  Clarithromycin tablets.
452.150b  Clarithromycin granules for oral suspension.

[[Page 912]]

452.160  Azithromycin oral dosage forms.
452.160a  Azithromycin capsules.
452.160b  Azithromycin for oral suspension.
452.175  Troleandomycin oral dosage forms.
452.175a  Troleandomycin capsules.
452.175b  Troleandomycin oral suspension.
452.175c  Troleandomycin for oral suspension.
452.175d  Troleandomycin chewable tablets.

                   Subpart C--Injectable Dosage Forms

452.225  Erythromycin ethylsuccinate injection.
452.230  Sterile erythromycin gluceptate.
452.232  Erythromycin lactobionate injectable dosage forms.
452.232a  Erythromycin lactobionate for injection.
452.232b  Sterile erythromycin lactobionate.

                   Subpart D--Ophthalmic Dosage Forms

452.310  Erythromycin ophthalmic ointment.

                          Subpart E  [Reserved]

                  Subpart F--Dermatologic Dosage Forms

452.510  Erythromycin dermatologic dosage forms.
452.510a  Erythromycin ointment.
452.510b  Erythromycin topical solution.
452.510d  Erythromycin-benzoyl peroxide topical gel.
452.510e  Erythromycin topical gel.

                          Subpart G  [Reserved]

                     Subpart H--Rectal Dosage Forms

452.710  Erythromycin suppositories.

                          Subpart I  [Reserved]

                  Subpart J--Certain Other Dosage Forms

452.910  Erythromycin for prescription compounding.

    Authority:  21 U.S.C. 357.



                          Subpart A--Bulk Drugs



Sec. 452.10  Erythromycin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin is the odorless, white to 
grayish-white or slightly yellow compound of a kind of erythromycin or a 
mixture of two or more such compounds. It is so purified and dried that:
    (i) It contains not less than 850 micrograms of erythromycin per 
milligram calculated on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 10 percent.
    (iv) Its pH is not less than 8.0 or more than 10.5.
    (v) Its residue on ignition is not more than 2.0 percent.
    (vi) Its heavy metals content is not more than 50 parts per million.
    (vii) It gives a positive identity test for erythromycin.
    (viii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
residue on ignition, heavy metals, pH, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing not less than 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient methyl alcohol to 
give a concentration of 10 milligrams of erythromycin base per 
milliliter (estimated). Dilute this solution further with sufficient 
0.1M potassium phosphate buffer, pH 8.0 (solution 3), to give a stock 
solution containing 1.0 milligram of erythromycin base per milliliter 
(estimated). Further dilute an aliquot of the stock solution with 
solution 3 to the reference concentration of 1.0 microgram of 
erythromycin base per milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, except 
standardize the pH meter with pH 7.0 and pH 10.0 buffers and prepare the 
sample as follows: Dissolve 200 milligrams of sample in 5 milliliters of 
reagent grade methyl alcohol. Add 95 milliliters of water and mix. 
Record the pH when an equilibrium value has been reached.
    (5) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (6) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.

[[Page 913]]

    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (8) Identity test. Proceed as directed in Sec. 436.211 of this 
chapter, using the sample preparation method described in paragraph 
(b)(3) of that section.
[39 FR 19149, May 30, 1974, as amended at 42 FR 38564, July 29, 1977; 43 
FR 9801, Mar. 10, 1978; 50 FR 19920, May 13, 1985]



Sec. 452.15  Erythromycin estolate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin estolate is the lauryl 
sulfate salt of the propionyl ester of a kind of erythromycin or a 
mixture of two or more such salts. It occurs as a white powder. It is 
soluble in alcohol, methyl alcohol, acetone, and chloroform, but is 
practically insoluble in water. It is so purified and dried that:
    (i) It contains not less than 600 micrograms of erythromycin per 
milligram, calculated on an anhydrous basis.
    (ii) Its free erythromycin content is not more than 3.0 percent.
    (iii) Its moisture content is not more than 4.0 percent.
    (iv) Its pH is not less than 4.5 nor more than 7.0.
    (v) It gives positive identity tests for erythromycin estolate.
    (vi) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, free 
erythromycin content, moisture, pH, crystallinity, and identity.
    (ii) Samples of the batch: A minimum of 10 containers, each 
containing not less than 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient methyl alcohol to 
give a concentration of 1.0 milligram of erythromycin base per 
milliliter (estimated). Immediately dilute this solution further with 
0.1M potassium phosphate buffer, pH 8.0 (solution 3), to give a 
concentration of 0.1 milligram of erythromycin per milliliter 
(estimated). Hydrolyze this solution in a 60 deg. C. constant 
temperature water bath for 2 hours or at room temperature for 16 to 18 
hours. Further dilute with solution 3 to the reference concentration of 
1.0 microgram of erythromycin base per milliliter (estimated).
    (2) Free erythromycin content. Proceed as directed in Sec. 436.362 
of this chapter.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous suspension containing 10 milligrams per milliliter.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (6) Identity test. Proceed as directed in Sec. 436.211 of this 
chapter, preparing the sample as described in paragraph (b)(1) of that 
section.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19920, May 13, 1985; 53 
FR 1920, Jan. 25, 1988]



Sec. 452.25  Erythromycin ethylsuccinate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin ethylsuccinate is the white, 
odorless, ethylsuccinate ester of erythromycin. It is so purified and 
dried that:
    (i) It contains not less than 765 micrograms of erythromycin per 
milligram, calculated on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 3.0 percent.
    (iv) Its pH is not less than 6.0 and not more than 8.5.
    (v) Its residue on ignition is not more than 1.0 percent.
    (vi) It gives a positive identity test for erythromycin 
ethylsuccinate.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:

[[Page 914]]

    (i) Results of tests and assays on the batch for potency, moisture, 
pH, residue on ignition, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient methyl alcohol to 
give a concentration of 1 milligram of erythromycin base per milliliter 
(estimated). Further dilute with 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to the reference concentration of 1.0 microgram of 
erythromycin base per milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
1.0 percent suspension in water.
    (5) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample prepared as described in paragraph (b)(3) of that 
section.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19920, May 13, 1985]



Sec. 452.25a  Sterile erythromycin ethylsuccinate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin ethylsuccinate is the white, 
odorless, ethylsuccinate ester of erythromycin. It is so purified and 
dried that:
    (i) It contains not less than 765 micrograms of erythromycin per 
milligram, calculated on an anhydrous basis.
    (ii) It is sterile.
    (iii) [Reserved]
    (iv) Its moisture content is not more than 3.0 percent.
    (v) Its pH is not less than 6.0 and not more than 8.5.
    (vi) Its residue on ignition is not more than 1.0 percent.
    (vii) It gives a positive identity test for erythromycin 
ethylsuccinate.
    (viii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
moisture, pH, residue on ignition, identity, and crystallinity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 600 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient methyl alcohol to 
give a concentration of 1 milligram of erythromycin base per milliliter 
(estimated). Further dilute with 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to the reference concentration of 1.0 microgram of 
erythromycin base per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section.
    (3) [Reserved]
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
1.0 percent suspension in water.
    (6) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation method described in paragraph (b)(3) of 
that section.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19920, May 13, 1985]



Sec. 452.30a  Sterile erythromycin gluceptate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin gluceptate is the white 
powder of the glucoheptonic

[[Page 915]]

acid salt of erythromycin or a mixture of two or more such salts. It is 
freely soluble in water, alcohol, and methyl alcohol. It is slightly 
soluble in acetone and chloroform, but is practically insoluble in 
ether. It is so purified and dried that:
    (i) It contains not less than 600 micrograms of erythromycin per 
milligram, calculated on an anhydrous basis. If it is packaged for 
dispensing, its potency is satisfactory if it is not less than 90 
percent and not more than 115 percent of the number of milligrams of 
erythromycin that it is represented to contain.
    (ii) It is sterile.
    (iii) [Reserved]
    (iv) It is nonpyrogenic.
    (v) Its moisture content is not more than 5.0 percent.
    (vi) Its pH in an aqueous solution containing 25 milligrams per 
milliliter is not less than 6.0 nor more than 8.0.
    (vii) It gives a positive identity test for erythromycin gluceptate.
    (2) [Reserved]
    (3) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (4) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, and identity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use as an 
ingredient in the manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing not 
less than 300 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 12 immediate 
containers of the batch.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
If the batch is packaged for repacking or for use in manufacturing 
another drug, dissolve an accurately weighed sample in sufficient methyl 
alcohol to give a concentration of 10 milligrams of erythromycin base 
per milliliter (estimated). Dilute this solution further with sufficient 
0.1M potassium phosphate buffer, pH 8.0 (solution 3), to give a stock 
solution containing 1.0 milligram of erythromycin base per milliliter 
(estimated). If it is packaged for dispensing, reconstitute as directed 
in the labeling. Then using a suitable hypodermic needle and syringe, 
remove all of the withdrawable contents if it is represented as a single 
dose container; or if the labeling specifies the amount of potency in a 
given volume of the resultant preparation, remove an accurately measured 
representative portion from each container. Dilute with solution 3 to 
give a stock solution of convenient concentration. Further dilute the 
stock solution with solution 3 to the reference concentration of 1.0 
microgram of erythromycin base per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 30 milligrams of erythromycin per 
milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
concentration of 25 milligrams per milliliter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation method described in paragraph (b)(2) of 
that section.
[39 FR 19149, May 30, 1974, as amended at 46 FR 16685, Mar. 13, 1981; 50 
FR 19920, 19921, May 13, 1985]



Sec. 452.32a  Sterile erythromycin lactobionate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin lactobionate is the white to 
off-white powder of the lactobionate salt of erythromycin or a mixture 
of two or more such salts. It is so purified and dried that:

[[Page 916]]

    (i) If the erythromycin lactobionate is not packaged for dispensing, 
its erythromycin potency is not less than 525 micrograms of erythromycin 
per milligram on an anhydrous basis. If the erythromycin lactobionate is 
packaged for dispensing, its erythromycin potency is not less than 525 
micrograms of erythromycin per milligram on an anhydrous basis and also, 
each container contains not less than 90 percent and not more than 120 
percent of the number of milligrams of erythromycin that it is 
represented to contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 5.0 percent.
    (v) Its pH is not less than 6.5 and not more than 7.5.
    (vi) Its residue on ignition is not more than 2.0 percent.
    (vii) Its heavy metals content is not more than 50 parts per 
million.
    (viii) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, residue on ignition, heavy metals, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) If the batch is packaged for repacking or for use as an 
ingredient in the manufacture of another drug:
    (1) For all tests except sterility: A minimum of 12 immediate 
containers.
    (2) For sterility testing: 20 packages, each containing equal 
portions of approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 12 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows:
    (i) Product not packaged for dispensing (micrograms of erythromycin 
per milligram). Dissolve an accurately weighed sample with sufficient 
methyl alcohol to obtain a concentration of 10 milligrams of 
erythromycin base per milliliter (estimated). Further dilute an aliquot 
of this solution with 0.1M potassium phosphate buffer, pH 8.0 (solution 
3), to the reference concentration of 1.0 microgram of erythromycin base 
per milliliter (estimated).
    (ii) Product packaged for dispensing. Determine both micrograms of 
erythromycin per milligram of sample and milligrams of erythromycin per 
container. Use separate containers for preparation of each sample 
solution as described in paragraph (b)(1)(ii)(a) and (b) of this 
section.
    (a) Micrograms of erythromycin per milligram. Dissolve an accurately 
weighed sample with sufficient methyl alcohol to obtain a concentration 
of 10 milligrams of erythromycin base per milliliter (estimated). 
Further dilute an aliquot of this solution with 0.1M potassium phosphate 
buffer, pH 8.0 (solution 3), to the reference concentration of 1.0 
microgram of erythromycin base per milliliter (estimated).
    (b) Milligrams of erythromycin per container. Reconstitute the 
sample as directed in the labeling. Then using a suitable hypodermic 
needle and syringe, remove all of the withdrawable contents if it is 
represented as a single-dose container; or, if the labeling specifies 
the amount of potency in a given volume of the resultant preparation, 
remove an accurately measured representative portion from each 
container. Dilute an aliquot of the solution thus obtained with sterile 
distilled water to obtain a concentration of 10 milligrams of 
erythromycin base per milliliter (estimated). Further dilute an aliquot 
of this solution with 0.1M potassium phosphate buffer, pH 8.0 (solution 
3), to the reference concentration of 1.0 microgram of erythromycin base 
per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.

[[Page 917]]

    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 30 milligrams of erythromycin per 
milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
concentration of 50 milligrams of erythromycin per milliliter.
    (6) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (7) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
    (8) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation method described in paragraph (b)(3) of 
that section.
[51 FR 35215, Oct. 2, 1986, as amended at 55 FR 11584, Mar. 29, 1990]



Sec. 452.35  Erythromycin stearate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin stearate is the odorless, 
white or slightly yellow powder of the stearic acid salt of 
erythromycin. It is practically insoluble in water but is soluble in 
alcohol, methyl alcohol, chloroform, and ether. It is so purified and 
dried that:
    (i) It contains not less than 550 micrograms of erythromycin per 
milligram, calculated on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 4.0 percent.
    (iv) Its pH is not less than 6.0 and not more than 11.0.
    (v) Its residue on ignition is not more than 1.0 percent.
    (vi) It gives positive identity tests for erythromycin stearate.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH residue on ignition, identity, and crystallinity.
    (ii) Samples required: A minimum of 10 containers, each consisting 
of 500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient methyl alcohol to 
give a concentration of 1 milligram of erythromycin base per milliliter 
(estimated). Further dilute with 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to the reference concentration of 1.0 microgram of 
erythromycin base per milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
1 percent slurry of erythromycin stearate in water.
    (5) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation method described in paragraph (b)(2) of 
that section.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19920, 19921, May 13, 
1985]



Sec. 452.50  Clarithromycin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clarithromycin is 6-O-methylerythromycin 
A. It is so purified and dried that:
    (i) Its potency is not less than 960 micrograms of clarithromycin 
activity per milligram, on an anhydrous basis.
    (ii) Its moisture content is not more than 2.0 percent.
    (iii) The pH of a 0.2 percent (weight per volume) slurry in aqueous 
methanol (95:5) is not less than 7.5 and not more than 10.0.
    (iv) Its residue on ignition is not more than 0.3 percent.
    (v) Its heavy metals content is not more than 20 parts per million.
    (vi) Its specific rotation in chloroform containing 10 milligrams of 
clarithromycin per milliliter at 20  deg.C is between -89 deg. and -
95 deg., calculated on an anhydrous basis.
    (vii) It gives a positive identity test.
    (viii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 918]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for clarithromycin 
potency, moisture, pH, residue on ignition, heavy metals, specific 
rotation, identity, and crystallinity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using a constant column temperature of 50 
deg.C, a suitable ultraviolet detection system operating at 210 
nanometers, an analytical column 3 to 30 centimeters long packed with a 
reversed phase packing material such as octadecyl hydrocarbon bonded 
silicas (3 to 10 micrometers in diameter), the inlet of this column is 
connected to a guard column 1 to 5 centimeters in length packed with the 
same material of 5- to 30-micrometer particle size, a constant flow rate 
of 0.7 to 1.0 milliliters per minute, and a known injection volume of 
between 10 and 20 microliters. The retention time for clarithromycin is 
between 5 and 6 minutes and the retention time for 6,11-Di-O-
methylerythromycin A (resolution compound) is between 7 and 8 minutes. 
Mobile phase, system suitability solution, working standard and sample 
solutions, system suitability requirements, and calculations are as 
follows:
    (i) Mobile phase. Add 650 milliliters of methanol and 350 
milliliters of 0.067 M potassium phosphate (monobasic) to a suitable 
container, mix well, and adjust the pH to 4.0 with phosphoric acid. 
Filter through a suitable filter capable of removing particulate matter 
to 0.5 micron in diameter. Degas the mobile phase just prior to its 
introduction into the chromatograph.
    (ii) Preparation of system suitability solution. Prepare a methanol 
solution containing approximately 625 micrograms per milliliter each of 
clarithromycin and 6,11-Di-O-methylerythromycin A. Quantitatively 
transfer an aliquot of this solution to a suitable volumetric flask and 
dilute it to volume with mobile phase to obtain a solution containing 
approximately 125 micrograms each of clarithromycin and 6,11-Di-O-
methylerythromycin A.
    (iii) Preparation of working standard solution. Dissolve (by shaking 
or sonication) an accurately weighed portion of the clarithromycin 
working standard in sufficient methanol to obtain a known solution 
containing about 625 micrograms of clarithromycin activity per 
milliliter. Quantitatively transfer an aliquot of this solution to a 
suitable volumetric flask and dilute to volume with mobile phase and mix 
to obtain a known solution containing approximately 125 micrograms of 
clarithromycin activity per milliliter. Filter through a suitable filter 
capable of removing particulate matter to 0.5 micron in diameter.
    (iv) Sample solution. Dissolve (by shaking or sonication) an 
accurately weighed portion of the sample in sufficient methanol to 
obtain a solution containing 625 micrograms of clarithromycin activity 
per milliliter (estimated). Quantitatively transfer an aliquot of this 
solution to a suitable volumetric flask and dilute to volume with mobile 
phase and mix to obtain a known solution containing approximately 125 
micrograms of clarithromycin activity per milliliter (estimated). Filter 
through a suitable filter capable of removing particulate matter to 0.5 
micron in diameter.
    (v) System suitability requirements--(A) Asymmetry factor. The 
asymmetry factor (AS) is satisfactory if it is not less than 
0.9 and not more than 1.5 for the clarithromycin peak.
    (B) Efficiency of the column. The absolute efficiency 
(hr) is satisfactory if it is not more than 40.0 for the 
clarithromycin peak.
    (C) Resolution factor. The resolution factor (R) between the peak 
for clarithromycin and the peak for 6,11-Di-O-methylerythromycin A is 
satisfactory if it is not less than 2.0.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent of 5 replicate 
injections) is satisfactory if it is not more than 2.0 percent.
    (E) Capacity factor. Calculate the clarithromycin capacity factor 
(k') as follows:


[[Page 919]]



                  k' = (tr/t0)-1

where:

tr = Retention time of the clarithromycin peak; and
t0 = Void volume time.


The capacity factor is satisfactory if it is not less than 1.3 and not 
more than 4.0. If the system suitability parameters have been met, then 
proceed as described in Sec. 436.216(b) of this chapter.
    (vi) Calculations. Calculate the micrograms of clarithromycin per 
milligram of sample on an anhydrous basis as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.293

where:

AU = Area of the clarithromycin peak (at a retention time 
          equal to that observed for the clarithromycin standard) in the 
          chromatogram of the sample;
AS = Area of the clarithromycin peak in the chromatogram of 
          the clarithromycin working standard;
PS = Clarithromycin activity in the clarithromycin working 
          standard solution in micrograms per milliliter;
CU = Milligrams of sample per milliliter of sample solution; 
          and
m = Percent moisture content of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the sample preparation described in paragraph (d)(1) of that 
section and the titration procedure described in paragraph (e)(3) of 
that section, except that instead of adding 20 milliliters of solvent A 
before starting the titration, add a sufficient volume of solvent C to 
cover the electrodes in the dry titrating vessel.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, except 
standardize the pH meter with pH 7.0 and pH 10.0 buffers and prepare the 
sample as follows: Transfer 200 milligrams of the sample to a 150-
milliliter beaker. Add 5 milliliters of methanol and then 95 milliliters 
of distilled water. Place the pH electrodes in the slurry and stir at 
the slowest speed possible to ensure mixing but no vortex. After 10 
minutes, while still stirring, determine the pH.
    (4) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (5) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
    (6) Specific rotation. Dilute an accurately weighed sample with 
sufficient chloroform to give a concentration of approximately 10 
milligrams of clarithromycin per milliliter. Proceed as directed in 
Sec. 436.210 of this chapter, using a 1.0-decimeter polarimeter tube, 
maintaining the solution at 20  deg.C, and calculate the specific 
rotation on an anhydrous basis.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
preparing the sample as follows: Prepare a 5-percent solution of the 
sample in chloroform and use 0.1 millimeter matched absorption cells.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[58 FR 26653, May 4, 1993]



Sec. 452.60  Azithromycin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Azithromycin is the dihydrate form of 
(2R,3S,4R,5R,8R,10R,11R,12S,13S,14R)-13-[(2,6-dideoxy-3-C-methyl-3-O-
methyl--L-ribo-hexopyranosyl)oxy]-2-ethyl-3,4,10-trihydroxy-
3,5,6,8,10,12,14-heptamethyl-11-[[3,4,6-trideoxy-3-(dimethylamino)-
-D-xylo-hexopyranosyl]oxy]-1-oxa-6-azacyclopentadecan-15-one. 
It is so purified and dried that:
    (i) Its potency is not less than 945 micrograms and not more than 
1,030 micrograms of azithromycin activity per milligram, on the 
anhydrous basis.
    (ii) Its moisture content is not less than 4.0 percent and not more 
than 5.0 percent.
    (iii) The pH of an aqueous methanol (1:1) solution containing 2 
milligrams per milliliter is not less than 9 and not more than 11.
    (iv) Its residue on ignition is not more than 0.3 percent.
    (v) Its heavy metals content is not more than 25 parts per million.
    (vi) The specific rotation in absolute ethanol containing 20 
milligrams of azithromycin per milliliter at 20  deg.C is between -
45 deg. to -49 deg., calculated on an anhydrous basis.
    (vii) It is crystalline.
    (viii) It gives a positive identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 920]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for azithromycin 
potency, moisture, pH, residue on ignition, heavy metals, specific 
rotation, crystallinity, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an amperometric 
electrochemical detection system with dual glassy carbon electrodes 
operated in the oxidative screen mode with electrode 1 set at +0.70 
volt0.05 volt and electrode 2 at +0.82 volt0.05 
volt. (The 0.05-volt variance allows for optimization of the 
background current to 70 to 100 nanoamperes.) Detection of azithromycin 
occurs at electrode 2 where the voltage is sufficiently high to oxidize 
the amine functional groups on the molecules. Use a 15-centimeters by 
4.6-millimeters (inside diameter) column packed with alumina-based 
polybutadiene 5 micrometer spherical particles with 80-angstrom pore 
size (e.g., ES Industries RP1/p). The inlet of this column is 
connected to a guard column 5 centimeters by 4.6 millimeters (inside 
diameter) packed with the same material. The flow rate is 1.5 
milliliters per minute. Use a fixed volume loop injector or equivalent 
device to inject a volume of 50 microliters into the system. The 
retention time for azithromycin is between 10 and 13 minutes. Mobile 
phase, working standard, sample, and resolution solutions, system 
suitability requirements, and calculations are as follows:
    (i) Mobile phase. Dissolve 5.8 grams of potassium phosphate 
monobasic in 2,130 milliliters of ultrapure deionized or high-
performance liquid chromatographic-grade water. Add 870 milliliters of 
acetonitrile and mix. The mobile phase is 0.02 M potassium phosphate 
monobasic: acetonitrile (71:29). Adjust the pH of the mobile phase to pH 
11.00.1 with 10 M potassium hydroxide (about 6 milliliters). 
Filter the mobile phase through a suitable filter capable of removing 
particulate matter 0.5 micron in diameter and degas it just prior to its 
introduction into the chromatograph.
    (ii) Preparation of working standard solution. Accurately weigh 
approximately 16.5 milligrams of the azithromycin working standard into 
a 100-milliliter volumetric flask. Dissolve the material, aided by brief 
sonication, in 10 milliliters of acetonitrile and dilute to volume with 
acetonitrile. Pipet 2.0 milliliters of this solution into a 100-
milliliter volumetric flask and dilute to volume with mobile phase. This 
solution contains approximately 0.003 milligram per milliliter of 
azithromycin.
    (iii) Sample solution. Accurately weigh approximately 16.5 
milligrams of sample into a 100-milliliter volumetric flask. Dissolve 
the sample, aided by brief sonication, in 10 milliliters of acetonitrile 
and dilute to volume with acetonitrile. Pipet 2.0 milliliters of this 
solution into a 100-milliliter volumetric flask and dilute to volume 
with mobile phase.
    (iv) Resolution test solution. Weigh approximately 16.5 milligrams 
each of azithromycin working standard and azaerythromycin A reference 
standard into a 100-milliliter volumetric flask. Dissolve the materials 
aided by brief sonication in 10 milliliters of acetonitrile and dilute 
to volume with acetonitrile. Pipet 2.0 milliliters of this solution into 
a 100-milliliter volumetric flask and dilute to volume with mobile 
phase.
    (v) System suitability requirements. Using the resolution test 
solution, determine the:
    (A) Asymmetry factor. The asymmetry factor (AS) is 
satisfactory if it is not less than 0.9 and not more than 1.5 for the 
azithromycin peak.
    (B) Efficiency of the column. The absolute efficiency 
(hr) is satisfactory if it is not more than 40.0 for the 
azithromycin peak.
    (C) Resolution factor. The resolution factor (R) between the peak 
from azithromycin and the peak for azaerythromycin A is satisfactory if 
it is not less than 2.5.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent of 5 replicate 
injections) is satisfactory if it is not

[[Page 921]]

more than 2.0 percent. If the system suitability parameters have been 
met, then proceed as described in Sec. 436.216(b) of this chapter.
    (vi) Calculations. Calculate the micrograms of azithromycin per 
milligram of sample on an anhydrous basis as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.294

where:

AU = Area of the azithromcin peak (at a retention time equal 
          to that observed for the azithromycin standard) in the 
          chromatogram of the sample;
AS = Area of the azithromcin peak in the chromatogram of the 
          azithromycin working standard;
PSAzithromycin activity in the azithromycin working standard 
          solution in micrograms per milliliter;
CU = Milligrams of sample per milliliter of sample solution; 
          and
m = Percent moisture content of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous methanol (1:1) solution containing 2 milligrams per 
milliliter, prepared by diluting a methanol solution containing 4 
milligrams of azithromycin dihydrate 1:1 with distilled water.
    (4) Residue on ignition. Proceed as directed in Sec. 436.207(b) of 
this chapter, except use a temperature of 800  deg.C instead of a 
temperature range of 500 to 600  deg.C.
    (5) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
    (6) Specific rotation. Dissolve an accurately weighed sample with 
sufficient absolute ethanol to give a concentration of approximately 20 
milligrams per milliliter. Proceed as directed in Sec. 436.210 of this 
chapter, except dilute and maintain the test solution at 20  deg.C 
instead of 25  deg.C. Use a 1.0-decimeter polarimeter tube and calculate 
the specific rotation on an anhydrous basis.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (8) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 0.5 percent potassium bromide disc prepared as described in 
paragraph (b)(1) of that section.
[58 FR 26657, May 4, 1993]



Sec. 452.75  Troleandomycin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Troleandomycin is the triacetyl ester of 
oleandomycin base or a mixture of two or more such esters. It is a white 
powder. It is so purified that:
    (i) Its potency is not less than 750 micrograms of troleandomycin 
per milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 1.0 percent.
    (iv) Its pH in an aqueous alcohol solution containing 100 milligrams 
of troleandomycin per milliliter is not less than 7.0 and not more than 
8.5.
    (v) Its residue on ignition is not more than 0.1 percent.
    (vi) It gives a positive identity test for oleandomycin.
    (vii) Its Rf value by paper chromatography is 
approximately 0.85. If more than one spot appears on the paper 
chromatogram, determine its acetyl value, which is not less than 15.3 
percent and not more than 16.0 percent.
    (viii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, residue on ignition, identity, Rf value, acetyl 
value (only if more than one spot is present in the determination of 
Rf value), and crystallinity.
    (ii) Samples of the batch: 10 packages, nine containing 
approximately equal portions of not less than 500 milligrams, and one 
containing not less than 2.0 grams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological turbidimetric assay shall be conclusive.
    (i) Chemical method--(a) Reagents and equipment. (1) Methyl orange 
reagent: Shake 0.5M boric acid solution for 12 hours (to ensure 
saturation) with an excess of methyl orange indicator. An alternative 
method is to heat the mixture to about 50 deg. C. and shake for about

[[Page 922]]

an hour. Then allow to cool. Filter the saturated dye solution and wash 
three times with chloroform. Store the dye solution over chloroform.
    (2) Acid-alcohol solution: Add 2 milliliters of concentrated 
sulfuric acid to 98 milliliters of absolute methyl alcohol.
    (3) Glycerin: Reagent grade.
    (4) Chloroform.
    (5) Glacial acetic acid.
    (6) Centrifuge tubes: 40 milliliters, glass-stoppered.
    (b) Procedure. Using the troleandomycin working standard which has 
been dried for 3 hours at 60 deg. C. and a pressure of 5 millimeters or 
less, prepare a standard solution in chloroform containing 50.0 
milligrams of oleandomycin base in 200 milliliters. Transfer 10.0 
milliliters of the solution to a 100-milliliter volumetric flask and 
dilute to volume with chloroform. Transfer 2.0, 4.0, 6.0, and 8.0 
milliliters of this solution to glass-stoppered centrifuge tubes (40-
milliliter size) and dilute to a total volume of 20.0 milliliters each 
with chloroform. To the 20 milliliters of the solution present in each 
40-milliliter size centrifuge tube, add 0.2 milliliter of glacial acetic 
acid, 0.2 milliliter of glycerin, and 0.4 milliliter of methyl orange 
reagent. Shake for 5 minutes and centrifuge for 3 minutes. Immediately 
transfer to another tube a 10.0-milliliter aliquot from the chloroform 
(lower) layer. Care must be exercised to see that no portion of the dye-
glycerin phase is included with the chloroform aliquot. Add 1.0 
milliliter of acid-alcohol solution to this chloroform aliquot, mix 
well, and read the absorbancy at 535 nanometers, using a 1-centimeter 
cell and a suitable photometer and using chloroform, similarly treated, 
as a blank. Prepare a standard curve, plotting the absorbance values of 
the standard solution against the concentration expressed in micrograms 
of oleandomycin base per aliquot. Accurately weigh the sample to be 
tested to give 50 milligrams (estimated) of oleandomycin base. Dissolve 
in chloroform and make to 200 milliliters with chloroform. Transfer 10.0 
milliliters to a 100-milliliter volumetric flask and make to volume with 
chloroform. Transfer 5.0 milliliters to a glass-stoppered centrifuge 
tube and proceed as above. Determine the potency of the sample from the 
standard curve.
    (ii) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 80 percent isopropyl 
alcohol solution (solution 15) to obtain a stock solution containing 
1,000 micrograms per milliliter. Further dilute an aliquot of the stock 
solution with distilled water to the reference concentration of 25 
micrograms of troleandomycin per milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
saturated solution prepared by adding 100 milligrams of troleandomycin 
per milliliter of water-ethyl alcohol (1:1) diluent.
    (5) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter, except use a silica crucible.
    (6) Identity. Dissolve about 10 milligrams in 5 milliliters of 
hydrochloric acid and heat the solution in a boiling water bath; a 
greenish yellow color is produced.
    (7) Rf value--(i) Apparatus and reagents. (a) Chromatographic 
chamber (cylinder, glass-stoppered museum jar, 11.5 inches x 3.5 
inches).
    (b) Chromatographic paper (8 inches x 8 inches, Whatman No. 1).
    (c) 0.1N hydrochloric acid.
    (d) Resolving solvent: Butyl acetate, benzene, nitromethane, 
pyridine (5:5:5:1 by volume).
    (e) Spray developing reagent: Place 1.0 milliliter of 10 percent 
platinic chloride solution and 25.0 milliliters of 4 percent potassium 
iodide solution in a 250-milliliter volumetric flask. Fill to mark with 
distilled water and mix well.
    (ii) Procedure. Dissolve the sample in chloroform to give a solution 
containing 10 to 20 milligrams of oleandomycin base equivalent per 
milliliter. Prepare a sheet of chromatographic paper by drawing a line 
of origin parallel to and 1 inch from the edge of the paper. Wet the 
paper thoroughly with the 0.1N hydrochloric acid and blot it firmly 
between

[[Page 923]]

sheets of absorbent paper. Starting 2 inches in from the edge and at 1-
inch intervals, apply 3 to 5 microliters of the sample solutions to the 
starting line. Allow a few minutes for the paper to dry partially. While 
it is still damp, form a cylinder by bringing the outer edges together, 
allowing about 1-inch overlap, and secure with a paper clip. Stand the 
paper in the chromatographic chamber, which has been filled to a depth 
of one-half of an inch with the resolving solvent. After the solvent 
front rises to a height of 4 to 5 inches above the origin, remove the 
paper from the tank and hang it up to air dry. Spray the dried paper 
with the developing reagent. Hang the paper in a 100 deg. C. oven for 3 
minutes. A purple spot becomes visible for troleandomycin at an Rf 
value of about 0.85. The approximate Rf values for 
diacetyloleandomycin, monoacetyloleandomycin, and oleandomycin are, 
respectively, 0.72, 0.27, and 0.13.
    (8) Acetyl determination--(i) Apparatus and reagents. (a) One 3-
necked Pyrex flask of approximately 45 milliliters capacity, pear-shaped 
with T-joints, agar inlet tube, glass-stoppered funnel, glass condenser, 
and bubble counter.
    (b) 50-milliliter Pyrex Erlenmeyer flask.
    (c) 10-milliliter buret, calibrated to 0.02 milliliter.
    (d) Anhydrous methyl alcohol, reagent grade.
    (e) 2N sodium hydroxide solution.
    (f) Sulfuric acid solution prepared by adding 100 milliliters of 
concentrated H2SO4 to 200 milliliters of water.
    (g) 1N barium chloride solution.
    (h) Phenolphthalein solution (1 percent in ethyl alcohol).
    (i) Water-pumped nitrogen.
    (j) NaOH solution 0.015N.
    (ii) Procedure. Weigh accurately (to 0.01 milligram) approximately 
30 milligrams of the sample into the three-necked acetyl flask. Add 2.0 
milliliters of methyl alcohol to dissolve the sample; then add slowly, 
with gentle swirling, 1.0 milliliter of NaOH solution. Connect the gas 
inlet tube with bubble counter attached and adjust nitrogen flow to 
about two bubbles a second. Put glass-stoppered funnel in centerneck of 
acetyl flask and put about 5 milliliters of H2O in the 
funnel. Add a boiling chip to the solution and attach condenser in the 
refluxing position with water cooling. Adjust burner flame under acetyl 
flask to reflux solution gently. Reflux for 30 minutes. Cool assembly 
slightly; then rinse down condenser (still in reflux position) with a 
few milliliters of H2O. Reassemble condenser to the 
distillation position and add water through the funnel to make a total 
of approximately 5 milliliters of H2O added to acetyl flask. 
Adjust burner flame so that about 5 milliliters of H2O and 
methyl alcohol is distilled over in approximately 10 minutes. Discard 
this distillate. Cool acetyl flask slightly. Acidify solution in flask 
by adding 1 milliliter of the sulfuric acid solution through the funnel. 
Adjust burner flame and distill over approximately 20 milliliters of 
distillate into an Erlenmeyer flask in about 20 minutes, adding water 
through the funnel as necessary. It is important to keep the liquid 
volume in the acetyl flask around 2 to 3 milliliters in order to obtain 
a quantitative recovery of the acetic acid. Collect a second fraction of 
distillate, about 10 milliliters in volume. As the second fraction is 
distilling, process the first fraction. Heat the first fraction and boil 
gently about 20 seconds. Add a few drops of BaCl2 solution to 
check if any sulfate was distilled over. If the sulfate is present, 
discard and repeat the whole determination. If the sulfate is absent, 
immediately titrate the solution with the 0.015N NaOH solution to a 
faint-pink endpoint, using one drop of phenolphthalein solution as the 
indicator. Repeat the above procedure with the second fraction. If the 
second fraction requires less than 0.10 milliliter of the 0.015N NaOH 
solution and all the acetic acid has been distilled over, the 
determination is completed. If greater than this, collect a third 
fraction of approximately 10 milliliters and titrate this as before. 
Total volumes of NaOH used and calculate results as follows:

(Milliliters of NaOH x N NaOH x 0.043 x 100)/(Weight sample in 
          grams)=Percent acetyl.


[[Page 924]]


    (9) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19149, May 30, 1974, as amended at 48 FR 3960, Jan. 28, 1983; 50 
FR 19920, 19921, May 13, 1985]



                      Subpart B--Oral Dosage Forms



Sec. 452.110  Erythromycin oral dosage forms.



Sec. 452.110a  Erythromycin tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin tablets are erythromycin 
with suitable and harmless buffer substances, diluents, binders, 
lubricants, colorings, flavorings, and suitable preservatives. The 
potency of each tablet is 250 milligrams or 500 milligrams of 
erythromycin. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
erythromycin that it is represented to contain. Tablets shall 
disintegrate within 1 hour. The loss on drying is not more than 5.0 
percent. The erythromycin used in making the batch conforms to the 
standards prescribed by Sec. 452.10(a)(1), except heavy metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin used in making the batch for potency, pH, 
moisture, residue on ignition, crystallinity, and identity.
    (b) The batch for potency, disintegration time, and loss on drying.
    (ii) Samples required:
    (a) The erythromycin used in making the batch: 10 packages, each 
containing 500 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Blend a representative number of tablets in a high-speed glass blender 
for 2 to 3 minutes with 200 milliliters of methyl alcohol. Add 300 
milliliters of 0.1M potassium phosphate buffer, pH 8.0 (solution 3), and 
blend again for 2 to 3 minutes. Further dilute with solution 3 to the 
reference concentration of 1.0 microgram of erythromycin base per 
milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure described in paragraph (e)(2) of that 
section.
[39 FR 19149, May 30, 1974, as amended at 42 FR 59068, Nov. 15, 1977; 50 
FR 19921, May 13, 1985]



Sec. 452.110b  Erythromycin enteric-coated tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromyin enteric-coated tablets are 
enteric-coated tablets composed of erythromycin, suitable and harmless 
buffer substances, diluents, binders, lubricants, colorings, and 
flavorings. Each tablet contains 100, 250, 333, or 500 milligrams of 
erythromycin. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
erythromycin that it is represented to contain. The tablets shall 
disintegrate within 2 hours. The moisture content is not more than 6 
percent. The erythromycin base used in making the batch conforms to the 
standards of Sec. 452.10(a)(1) (i), (iii), (iv), (v), (vii), and (viii).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin used in making the batch for potency, moisture, 
pH, residue on ignition, crystallinity, and identity.
    (b) The batch for potency, moisture, and disintegration time.
    (ii) Samples required:
    (a) The erythromycin used in making the batch: 10 packages, each 
containing 500 milligrams.
    (b) The batch: A minimum of 36 tablets.

[[Page 925]]

    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Blend a representative number of tablets in a high-speed glass blender 
for 2 to 3 minutes with 200 milliliters of methyl alcohol. Add 300 
milliliters of 0.1M potassium phosphate buffer, pH 8.0 (solution 3), and 
blend again for 2 to 3 minutes. Further dilute with solution 3 to the 
reference concentration of 1.0 microgram of erythromycin base per 
milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure described in paragraph (e)(3) of that 
section.
[39 FR 14149, May 30, 1974, as amended at 44 FR 30332, May 25, 1979; 44 
FR 48190, Aug. 17, 1979; 46 FR 44442, Sept. 4, 1981; 47 FR 15326, Apr. 
9, 1982; 50 FR 19921, May 13, 1985]



Sec. 452.110c  Erythromycin capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin capsules are capsules 
containing enteric-coated erythromycin pellets, suitable and harmless 
buffer substances, diluents, binders, lubricants, and colorings. Each 
capsule contains either 125 milligrams or 250 milligrams of 
erythromycin. Its potency is satisfactory if it is not less than 90 
percent and not more than 115 percent of the number of milligrams of 
erythromycin that it is represented to contain. The moisture content is 
not more than 7.5 percent. It passes the acid resistance/dissolution 
test. The erythromycin used conforms to the standards prescribed by 
Sec. 452.10(a)(1), except heavy metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The erythromycin used in making the batch for potency, moisture, 
pH, residue on ignition, identity, and crystallinity.
    (b) The batch for potency, moisture, and acid resistance/
dissolution.
    (ii) Samples required:
    (a) The erythromycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch: A minimum of 100 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing 200 milliliters of methyl alcohol. Blend for 2 to 
3 minutes. Add 300 milliliters of 0.1M potassium phosphate buffer, pH 
8.0 (solution 3), and blend again for 2 to 3 minutes. Further dilute an 
aliquot with solution 3 to the reference concentration of 1.0 microgram 
of erythromycin base per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the sample preparation method described in paragraph (d)(1) of 
that section.
    (3) Acid resistance/dissolution. Proceed as directed in Sec. 436.542 
of this chapter. The quantity Q (the amount of erythromycin dissolved) 
is 85 percent at 45 minutes.
[46 FR 16678, Mar. 13, 1981; 46 FR 22359, Apr. 17, 1981, as amended at 
50 FR 19921, May 13, 1985; 50 FR 36992, Sept. 11, 1985; 50 FR 47214, 
Nov. 15, 1985]



Sec. 452.110d  Erythromycin particles in tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin particles in tablets are 
tablets containing erythromycin acid-resistant coated particles, 
suitable and harmless diluents, binders, lubricants, and colorings. Each 
tablet contains 333 milligrams of erythromycin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of erythromycin that it is 
represented to contain. The loss on drying is not more than 5.0 percent. 
It passes the dissolution test and the acid resistance test. The 
erythromycin used conforms to the standards prescribed by 
Sec. 452.10(a)(1), except heavy metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 926]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The erythromycin used in making the batch for potency, safety, 
moisture, pH, residue on ignition, identity, and crystallinity.
    (b) The batch for potency, loss on drying, dissolution, and acid 
resistance.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The erythromycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch: A minimum of 100 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar containing 200 milliliters of methyl alcohol. Blend for 2 to 3 
minutes. Add 300 milliliters of 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), and blend again for 2 to 3 minutes. Further dilute an 
aliquot with solution 3 to the reference concentration of 1.0 microgram 
of erythromycin base per milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q (the amount of erythromycin dissolved) is 75 
percent at 45 minutes.
    (4) Acid resistance. Proceed as directed in Sec. 436.545 of this 
chapter. The quantity of erythromycin dissolved is not more than 25 
percent at 60 minutes.
[51 FR 37723, Oct. 24, 1986, as amended at 55 FR 11584, Mar. 29, 1990]



Sec. 452.115  Erythromycin estolate oral dosage forms.



Sec. 452.115a  Erythromycin estolate tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin estolate tablets are 
composed of erythromycin estolate with one or more suitable and harmless 
diluents, binders, lubricants, and colorings. Each tablet contains 
erythromycin estolate equivalent to 500 milligrams of erythromycin. Its 
potency is satisfactory if it is not less than 90 percent and not more 
than 120 percent of the number of milligrams of erythromycin that it is 
represented to contain. The moisture content is not more than 5 percent. 
The tablets shall disintegrate within 30 minutes. The erythromycin 
estolate used conforms to the standards prescribed by Sec. 452.15(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The erythromycin estolate used in making the batch for potency, 
moisture, pH, identity, and crystallinity.
    (b) The batch for potency, moisture, and disintegration time.
    (ii) Samples required:
    (a) The erythromycin estolate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch. A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tables into a high-speed glass blender 
jar with 200 milliliters of methyl alcohol. Blend for 3 to 5 minutes. 
Add 300 milliliters of 0.1M potassium phosphate buffer, pH 8.0 (solution 
3), and blend again for 3 to 5 minutes. Hydrolyze a portion of this 
solution in a 60 deg. C. constant temperature water bath for 2 hours or 
at room temperature for 16 to 18 hours. Further dilute with solution 3 
to the reference concentration of 1.0 microgram of erythromycin base per 
milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure described in paragraph (e)(1) of that 
section.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]

[[Page 927]]



Sec. 452.115b  Erythromycin estolate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
quality, and purity. Erythromycin estolate capsules are capsules 
containing erythromycin estolate with suitable and harmless buffer 
substances and diluents enclosed in a gelatin capsule. The erythromycin 
estolate content of each capsule is equivalent to either 250 milligrams 
of erythromycin or 125 milligrams of erythromycin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of milligrams of erythromycin that it is 
represented to contain. The moisture content is not more than 5 percent. 
The erythromycin estolate used conforms to the standards prescribed 
therefor by Sec. 452.15(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain;
    (i) Results of tests and assays on:
    (a) The erythromycin estolate used in making the batch for potency, 
pH, moisture, crystallinity, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The erythromycin estolate used in making the batch: 10 packages, 
each containing not less than 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Test and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Blend a representative number of capsules in a high-speed glass blender 
with 200 milliliters of methyl alcohol for 2 to 3 minutes. Add 300 
milliliters of 0.1M potassium phosphate buffer, pH 8.0 (solution 3), and 
blend again for 2 to 3 minutes. Hydrolyze a portion of this solution in 
a 60 deg. C. constant temperature water bath for 2 hours or at room 
temperature for 16 to 18 hours. Further dilute with solution 3 to the 
reference concentration of 1.0 microgram of erythromycin base per 
milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 452.115c  Erythromycin estolate oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin estolate oral suspension is 
erythromycin estolate with suitable and harmless buffer substances, 
dispersing agents, diluents, coloring, and flavorings. Each milliliter 
contains erythromycin estolate equivalent to 25, 50, or 100 milligrams 
of erythromycin. Its potency is satisfactory if it is not less than 90 
percent and not more than 115 percent of the number of milligrams of 
erythromycin that it is represented to contain. Its pH is not less than 
3.5 and not more than 6.5. The erythromycin estolate used conforms to 
the standards prescribed by Sec. 452.15(a)(1).
    (2) Labeling. In addition to conforming with the requirements of 
Sec. 432.5 of this chapter, each package shall bear on its outside 
wrapper or container and the immediate container the statement 
``Refrigerate'' or ``Keep under refrigeration''.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin estolate used in making the batch for potency, 
moisture, pH, crystallinity, and identity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The erythromycin estolate used in making the batch: 10 
containers, each having not less than 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Remove an accurately measured representative volume of the suspension 
and dilute with sufficient methyl alcohol to give a concentration of 2.5 
milligrams per milliliter (estimated). Dilute the entire mixture with 
sufficient 0.1M potassium phosphate buffer, pH 8.0 (solution 3), to give 
a

[[Page 928]]

concentration of 1.0 milligram of erythromycin base per milliliter 
(estimated). Hydrolyze in a 60 deg. C. constant temperature water bath 
for 2 hours or at room temperature for 16 to 18 hours. Further dilute 
with solution 3 to the reference concentration of 1.0 microgram of 
erythromycin base per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug as it is prepared for dispensing.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 452.115d  Erythromycin estolate for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin estolate for oral suspension 
is a dry mixture of erythromycin estolate with suitable and harmless 
buffer substances, dispersing agents, diluents, colorings, and 
flavorings. The erythromycin estolate content is 25 milligrams of 
erythromycin per milliliter of the reconstituted suspension. Its potency 
is satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of milligrams of erythromycin that it is 
represented to contain. When reconstituted as directed in its labeling, 
its pH is not less than 5.0 and not more than 7.0. Its moisture content 
is not more than 2.0 percent. The erythromycin estolate used conforms to 
the standards of Sec. 452.15(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin estolate used in making the batch for potency, 
moisture, pH, crystallinity, and identity.
    (b) The batch: Potency, moisture, and pH.
    (ii) Samples required:
    (a) The erythromycin estolate used in making the batch: 10 immediate 
containers, each consisting of 300 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute the sample as directed in the labeling. Withdraw an 
accurately measured representative volume of the reconstituted 
suspension and add sufficient methyl alcohol to give a concentration of 
2.5 milligrams of erythromycin base per milliliter (estimated). Dilute 
this entire mixture with sufficient 0.1M potassium phosphate buffer, pH 
8.0 (solution 3), to give a concentration of 1.0 milligram of 
erythromycin base per milliliter (estimated). Hydrolyze in a 60 deg. C. 
constant temperature water bath for 2 hours or at room temperature for 
16 to 18 hours. Further dilute with solution 3 to the reference 
concentration of 1.0 microgram of erythromycin base per milliliter 
(estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the dry powder.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in its labeling.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 452.115e  Erythromycin estolate for pediatric drops.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin estolate for pediatric drops 
is a dry mixture of erythromycin estolate with suitable and harmless 
dispersing agents, buffer substances, diluents, colorings, and 
flavorings. When reconstituted as directed in the labeling, each 
milliliter contains the equivalent of 100 milligrams of erythromycin. 
Its potency is satisfactory if it is not less than 90 percent and not 
more than 115 percent of the number of milligrams of erythromycin that 
it is represented to contain. Its moisture content is not more than 2.0 
percent. Its pH is not less than 5.0 nor more than 5.5. The erythromycin 
estolate used conforms to the standards prescribed by Sec. 452.15(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of

[[Page 929]]

Sec. 431.1 of this chapter, each such request shall contain:
    (i) Results of tests and assays on:
    (a) The erythromycin estolate used in making the batch for potency, 
pH, moisture, crystallinity, and identity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The erythromycin estolate used in making the batch: 10 packages, 
each containing not less than 300 milligrams.
    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute the sample as directed in the labeling. Withdraw an 
accurately measured representative volume of the reconstituted 
suspension and add sufficient methyl alcohol to give a concentration of 
2.5 milligrams of erythromycin base per milliliter (estimated). Dilute 
this entire mixture with sufficient 0.1M potassium phosphate buffer, pH 
8 (solution 3), to give a concentration of 1.0 milligram of erythromycin 
base per milliliter (estimated). Hydrolyze in a 60 deg. C. constant 
temperature water bath for 2 hours or at room temperature for 16 to 18 
hours. Further dilute with solution 3 to the reference concentration of 
1.0 microgram of erythromycin base per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202(b) of this chapter, 
using the suspension prepared as directed in the labeling.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 452.115f  Erythromycin estolate chewable tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin estolate chewable tablets 
are tablets composed of erythromycin estolate and suitable and harmless 
diluents, binders, buffers, colorings, and flavorings. Each tablet 
contains erythromycin estolate equivalent to either 125 or 250 
milligrams of erythromycin. Its potency is satisfactory if it is not 
less than 90 percent and not more than 115 percent of the number of 
milligrams of erythromycin that it is represented to contain. The 
moisture content is not more than 4 percent. The erythromycin estolate 
used in making the batch conforms to the standards prescribed by 
Sec. 452.15(a)(1).
    (2) Labeling. It shall be labeled in accordance with Sec. 432.5 of 
this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin estolate used in making the batch for potency, 
moisture, pH, crystallinity, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The erythromycin estolate used in making the batch: 10 packages, 
each consisting of not less than 300 milligrams.
    (b) The batch: A minimum of 30 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Blend a representative number of tablets in a high-speed glass blender 
for 2 to 3 minutes in 200 milliliters of methyl alcohol. Add 300 
milliliters of 0.1M potassium phosphate buffer, pH 8.0 (solution 3), and 
blend again for 2 to 3 minutes. Hydrolyze this solution in a 60 deg. C. 
constant temperature water bath for 2 hours or at room temperature for 
16 to 18 hours. Further dilute with solution 3 to the reference 
concentration of 1.0 microgram of erythromycin base per milliliter 
(estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 452.115g  Erythromycin estolate and sulfisoxazole acetyl oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin estolate and sulfisoxazole 
acetyl oral suspension is erythromycin estolate and sulfisoxazole acetyl 
with suitable and harmless buffer substances, preservatives, solvents, 
stabilizers, emulsifiers, dispersing agents, colorings, and

[[Page 930]]

flavorings. Each milliliter contains erythromycin estolate equivalent to 
25 milligrams of erythromycin and 120 milligrams of sulfisoxazole. Its 
erythromycin content is satisfactory if it is not less than 90 percent 
and not more than 120 percent of the number of milligrams of 
erythromycin that it is represented to contain. Its sulfisoxazole acetyl 
content is satisfactory if it is not less than 90 percent and not more 
than 115 percent of the number of milligrams of sulfisoxazole that it is 
represented to contain. Its pH is not less than 3.5 and not more than 
6.5. The erythromycin estolate used conforms to the standards prescribed 
by Sec. 452.15(a)(1). The sulfisoxazole acetyl used conforms to the 
standards prescribed by the U.S.P. XXII.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The erythromycin estolate used in making the batch for potency, 
moisture, pH, crystallinity, and identity.
    (B) The sulfisoxazole acetyl used in making the batch for all U.S.P. 
XXII specifications.
    (C) The batch for erythromycin content, sulfisoxazole content, and 
pH.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research:
    (A) The erythromycin estolate used in making the batch: 10 packages, 
each containing not less than 500 milligrams.
    (B) The batch: a minimum of 15 immediate containers.
    (b) Tests and methods of assay--(1) Erythromycin content. Proceed as 
directed in Sec. 436.105 of this chapter, preparing the sample for assay 
as follows: Remove an accurately measured representative volume of the 
suspension and dilute with sufficient methyl alcohol to give a 
concentration of 2.5 milligrams per milliliter (estimated). Dilute the 
entire mixture with sufficient 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to give a concentration of 1.0 milligram of erythromycin 
base per milliliter (estimated). Hydrolyze in a 60  deg.C constant 
temperature water bath for 2 hours or at room temperature for 16 to 18 
hours. Further dilute with solution 3 to the reference concentration of 
1.0 microgram of erythromycin base per milliliter (estimated).
    (2) Sulfisoxazole content. Proceed as directed in Sec. 436.328 of 
this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug as it is prepared for dispensing.
[55 FR 280, Jan. 4, 1990]



Sec. 452.125  Erythromycin ethylsuccinate oral dosage forms.



Sec. 452.125a  Erythromycin ethylsuccinate chewable tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin ethylsuccinate chewable 
tablets are composed of erythromycin ethylsuccinate and suitable and 
harmless diluents, binders, buffers, colorings, and flavorings. Each 
tablet contains erythromycin ethylsuccinate equivalent to 200 milligrams 
of erythromycin. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
erythromycin that it is represented to contain. The moisture content is 
not more than 5 percent. The erythromycin ethylsuccinate used conforms 
to the standards prescribed by Sec. 452.25(a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``erythromycin 
ethylsuccinate tablets''.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin ethylsuccinate used in making the batch for 
potency, moisture, pH, residue on ignition, identity, and crystallinity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The erythromycin ethylsuccinate used in making the batch: 10 
packages, each consisting of 500 milligrams.
    (b) The batch: A minimum of 36 tablets.

[[Page 931]]

    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Blend a representative number of tablets in a high-speed glass blender 
for 2 to 3 minutes with 200 milliliters of methyl alcohol. Add 300 
milliliters of 0.1M potassium phosphate buffer, pH 8.0 (solution 3), and 
blend again for 2 to 3 minutes. Further dilute with solution 3 to the 
reference concentration of 1.0 microgram of erythromycin base per 
milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19149, May 30, 1974, as amended at 41 FR 51596, Nov. 23, 1976; 42 
FR 29858, June 10, 1977; 50 FR 19921, May 13, 1985]



Sec. 452.125b  Erythromycin ethylsuccinate oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin ethylsuccinate oral 
suspension is erythromycin ethylsuccinate with suitable and harmless 
buffer substances, dispersing agents, diluents, colorings, flavorings, 
and preservatives. Each milliliter contains erythromycin ethylsuccinate 
equivalent to 40 or 80 milligrams of erythromycin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of erythromycin that it is 
represented to contain. Its pH is not less than 6.5 and not more than 
8.5. The erythromycin ethylsuccinate used conforms to the standards 
prescribed by Sec. 452.25(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin ethylsuccinate used in making the batch for 
potency, moisture, pH, identity, residue on ignition, and crystallinity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The erythromycin ethylsuccinate used in making the batch: 10 
containers each consisting of 500 milligrams.
    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place an accurately measured representative volume of the suspension 
into a high-speed glass blender jar and add sufficient methyl alcohol to 
give a concentration of 1.0 milligram of erythromycin base per 
milliliter (estimated). Blend for 3 to 5 minutes. Further dilute with 
0.1M potassium phosphate buffer, pH 8.0 (solution 3), to the reference 
concentration of 1.0 microgram of erythromycin base per milliliter 
(estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted drug.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 452.125c  Erythromycin ethylsuccinate for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin ethylsuccinate for oral 
suspension is a dry mixture of erythromycin ethylsuccinate with suitable 
and harmless buffer substances, dispersing agents, diluents, colorings, 
and flavorings. It contains the equivalent of 40 milligrams or 80 
milligrams of erythromycin per milliliter of the reconstituted 
suspension. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
erythromycin that it is represented to contain. Its loss on drying is 
not more than 1 percent. When reconstituted as directed in the labeling, 
its pH is not less than 7.0 nor more than 9.0. The crystalline 
erythromycin ethylsuccinate used conforms to the standards prescribed by 
Sec. 452.25(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin ethylsuccinate used in making the batch for 
potency,

[[Page 932]]

moisture, pH, residue on ignition, identity, and crystallinity.
    (b) The batch, for potency, pH, and loss on drying.
    (ii) Samples required:
    (a) The erythromycin ethylsuccinate used in making the batch: 10 
containers each consisting of approximately 500 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Place an accurately measured 
representative portion of the sample into a high-speed glass blender jar 
containing sufficient methyl alcohol to give a final volume of 200 
milliliters. Blend for 3 to 5 minutes. Further dilute an aliquot with 
0.1M potassium phosphate buffer, pH 8.0 (solution 3), to the reference 
concentration of 1.0 microgram of erythromycin base per milliliter 
(estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the suspension prepared as directed in the labeling. If the suspension 
contains 80 milligrams per milliliter, equilibrium usually is reached in 
approximately 15 minutes.
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[39 FR 19149, May 30, 1974, as amended at 47 FR 21240, May 18, 1982; 50 
FR 19921, May 13, 1985]



Sec. 452.125d  Erythromycin ethylsuccinate tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin ethylsuccinate tablets are 
composed of erythromycin ethylsuccinate and suitable and harmless 
diluents, binders, buffers, and colorings. Each tablet contains 
erythromycin ethylsuccinate equivalent to 400 milligrams of 
erythromycin. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
erythromycin that it is represented to contain. The loss on drying is 
not more than 4.0 percent. The tablets shall disintegrate within 40 
minutes. The erythromycin ethylsuccinate used conforms to the standards 
prescribed by Sec. 452.25(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The erythromycin ethylsuccinate used in making the batch for 
potency, moisture, pH, residue on ignition, identity, and crystallinity.
    (b) The batch for potency, loss on drying, and disintegration time.
    (ii) Samples required:
    (a) The erythromycin ethylsuccinate used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar containing sufficient methyl alcohol to yield a concentration of 5 
milligrams of erythromycin activity or less per milliliter when blended. 
Blend for 3 to 5 minutes. Further dilute an aliquot of this solution 
with 0.1M potassium phosphate buffer, pH 8.0 (solution 3), to the 
reference concentration of 1.0 microgram of erythromycin base per 
milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter.
    (3) Disintegration time--(i) If the tablet is uncoated. Proceed as 
directed in Sec. 436.212 of this chapter, using the procedure described 
in paragraph (e)(1) of that section.
    (ii) If the tablet is plain-coated. Proceed as directed in 
Sec. 436.212 of this chapter, using the procedure described in paragraph 
(e)(2) of that section.
[41 FR 51596, Nov. 23, 1976, as amended at 50 FR 19921, May 13, 1985; 55 
FR 14091, Apr. 16, 1990]



Sec. 452.125e  Erythromycin ethylsuccinate-sulfisoxazole acetyl for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin ethylsuccinate-sulfisoxazole 
acetyl for

[[Page 933]]

oral suspension is a dry mixture of erythromycin ethylsuccinate and 
sulfisoxazole acetyl with suitable and harmless flavorings, buffers, 
surfactants, colorings, and suspending agents. When reconstituted as 
directed in the labeling, each milliliter will contain erythromycin 
ethylsuccinate equivalent to 40 milligrams of erythromycin and 
sulfisoxazole acetyl equivalent to 120 milligrams of sulfisoxazole. Its 
erythromycin ethylsuccinate content is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the number of 
milligrams of erythromycin that it is represented to contain. Its 
sulfisoxazole acetyl content is satisfactory if it is not less than 90 
percent and not more than 115 percent of the number of milligrams of 
sulfisoxazole that it is represented to contain. Its loss on drying is 
not more than 1.0 percent. When reconstituted as directed in the 
labeling, its pH is not less than 5.0 and not more than 7.0. The 
erythromycin ethylsuccinate used conforms to the standards prescribed by 
Sec. 452.25(a)(1). The sulfisoxazole acetyl used conforms to the 
standards prescribed by the U.S.P.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The erythromycin ethylsuccinate used in making the batch for 
potency, moisture, pH, residue on ignition, identity, and crystallinity.
    (b) The sulfisoxazole acetyl used in making the batch for all U.S.P. 
specifications.
    (c) The batch for erythromycin content, sulfisoxazole content, loss 
on drying, and pH.
    (ii) Samples required:
    (a) The erythromycin ethylsuccinate used in making the batch: 10 
packages each containing approximately 500 milligrams.
    (b) The batch: A minimum of 10 immediate containers.
    (b) Tests and methods of assay--(1) Erythromycin content. Proceed as 
directed in Sec. 436.105 of this chapter, preparing the sample for assay 
as follows: Reconstitute the sample as directed in the labeling. Allow 
to stand for 1 hour. Shake gently and transfer 5 milliliters of the 
well-shaken suspension into a high-speed glass blender jar containing 
195 milliliters of methyl alcohol. Blend for 3 to 5 minutes. Further 
dilute an aliquot with 0.1M potassium phosphate buffer, pH 8.0 (solution 
3), to the reference concentration of 1.0 microgram of erythromycin base 
per milliliter (estimated).
    (2) Sulfisoxazole acetyl content. Proceed as directed in 
Sec. 436.328 of this chapter.
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the suspension reconstituted as directed in the labeling.
[46 FR 2990, Jan. 13, 1981, as amended at 50 FR 19921, May 13, 1985]



Sec. 452.135  Erythromycin stearate oral dosage forms.



Sec. 452.135a  Erythromycin stearate tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin stearate tablets are tablets 
composed of erythromycin stearate with suitable and harmless buffer 
substances, diluents, binders, lubricants, colorings, and flavorings. 
Each tablet contains erythromycin stearate equivalent to 75, 100, 125, 
250, or 500 milligrams of erythromycin. Its potency is satisfactory if 
it is not less than 90 percent and not more than 120 percent of the 
number of milligrams of erythromycin that it is represented to contain. 
Tablets shall disintegrate within 1\1/2\ hours. The loss on drying is 
not more than 5.0 percent. The erythromycin stearate used in making the 
tablets conforms to the standards prescribed by Sec. 452.35(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin stearate used in making the batch for potency, 
pH,

[[Page 934]]

moisture, residue on ignition, crystallinity, and identity.
    (b) The batch for potency, loss on drying, and disintegration time.
    (ii) Samples required:
    (a) The erythromycin stearate used in making the batch: 10 
containers, each consisting of not less than 500 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Blend a representative number of tablets in a high-speed glass blender 
with 200 milliliters of methyl alcohol for 3 to 5 minutes. Add 300 
milliliters of 0.1M potassium phosphate buffer, pH 8.0 (solution 3), and 
blend again for 3 to 5 minutes. Further dilute with solution 3 to the 
reference concentration of 1.0 microgram of erythromycin base per 
milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the procedure described in paragraph (e)(2) of that 
section.
[39 FR 19149, May 30, 1974, as amended at 42 FR 59068, Nov. 15, 1977; 50 
FR 19921, May 13, 1985]



Sec. 452.135b  Erythromycin stearate oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin stearate oral suspension is 
erythromycin stearate with suitable and harmless buffer substances, 
dispersing agents, diluents, colorings, and flavorings. It contains the 
equivalent of 25 milligrams of erythromycin per milliliter. Its potency 
is satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of erythromycin that it is 
represented to contain. Its pH is not less than 7.0 and not more than 
8.5. The erythromycin stearate used conforms to the standards prescribed 
by Sec. 452.35(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The erythromycin stearate used in making the batch for potency, 
moisture, pH, residue on ignition, identity, and crystallinity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The erythromycin stearate used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place an accurately measured representative volume of the suspension 
into a high-speed glass blender jar. Add sufficient methyl alcohol to 
the jar to give a concentration of 1.25 milligrams of erythromycin base 
per milliliter (estimated). Blend for 2 to 3 minutes. Add sufficient 
0.1M potassium phosphate buffer, pH 8.0 (solution 3), to give a 
concentration of 0.5 milligrams of erythromycin base per milliliter 
(estimated) and blend again for 2 to 3 minutes. Further dilute with 
solution 3 to the reference concentration of 1.0 microgram of 
erythromycin base per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted suspension.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 452.135c  Erythromycin stearate for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin stearate for oral suspension 
is a dry mixture of erythromycin stearate with suitable and harmless 
buffer substances, dispersing agents, diluents, colorings, and 
flavorings. It contains the equivalent of 25 or 50 milligrams of 
erythromycin per milliliter of the reconstituted suspension. Its potency 
is satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of erythromycin that it is 
represented to contain. Its moisture content is not more than 2 percent. 
When reconstituted as directed in the

[[Page 935]]

labeling, its pH is not less than 6.0 and not more than 9.0. The 
erythromycin stearate used conforms to the standards prescribed by 
Sec. 452.35(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The erythromycin stearate used in making the batch for potency, 
moisture, pH, residue on ignition, identity, and crystallinity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The erythromycin stearate used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Place an accurately measured 
representative portion of the suspension into a high-speed glass blender 
jar with sufficient methyl alcohol to give a concentration of 1.0 
milligram of erythromycin base per milliliter (estimated). Blend for 3 
to 5 minutes. Further dilute with 0.1M potassium phosphate buffer, pH 
8.0 (solution 3), to the reference concentration of 1.0 microgram of 
erythromycin base per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the suspension obtained when the drug is reconstituted as directed in 
the labeling.
[40 FR 49083, Oct. 21, 1975, as amended at 41 FR 24884, June 21, 1976; 
50 FR 19921, May 13, 1985]



Sec. 452.150  Clarithromycin oral dosage forms.



Sec. 452.150a  Clarithromycin tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clarithromycin tablets are composed of 
clarithromycin and one or more suitable and harmless diluents, binders, 
lubricants, colorings, and flavorings. Each tablet contains 250 
milligrams or 500 milligrams of clarithromycin activity. Its 
clarithromycin content is satisfactory if it is not less than 90 percent 
and not more than 110 percent of the number of milligrams of 
clarithromycin that it is represented to contain. The loss on drying is 
not more than 6.0 percent. It passes the dissolution test. It passes the 
identity test. The clarithromycin used conforms to the standards 
prescribed by Sec. 452.50(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The clarithromycin used in making the batch for potency, 
moisture, pH, residue on ignition, heavy metals, specific rotation, 
identity, and crystallinity.
    (B) The batch for content, loss on drying, dissolution, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The clarithromycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch: A minimum of 100 tablets.
    (b) Tests and methods of assay--(1) Clarithromycin content. Proceed 
as directed in Sec. 452.50(b)(1), preparing the sample solution and 
calculating the clarithromycin content as follows:
    (i) Preparation of sample solution. Grind and composite five whole 
tablets in a glass mortar and pestle and quantitatively transfer the 
powder to a 500-milliliter volumetric flask with 50 milliliters of 
distilled water and shake mechanically until the tablets are well 
dispersed (approximately 5 to 10 minutes). Add 300 milliliters of 
methanol and shake mechanically for 30 minutes. Dilute with methanol to 
volume and mix. Allow the excipients to settle. Quantitatively transfer 
and dilute a convenient aliquot of the supernatant with mobile phase 
(described in Sec. 452.50(b)(1)(i)) to obtain a solution

[[Page 936]]

containing 125 micrograms of clarithromycin per milliliter (estimated). 
Filter through a suitable filter capable of removing particulate matter 
0.5 micron in diameter.
    (ii) Calculations. Calculate the clarithromycin content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.295
    
where:
AU = Area of the clarithromycin peak (at a retention time 
          equal to that observed for the clarithromycin standard) in the 
          chromatogram of the sample;
AS = Area of the clarithromycin peak in the chromatogram of 
          the clarithromycin working standard;
PS = Clarithromycin activity in the clarithromycin working 
          standard solution in micrograms per milliliter;
d = Dilution factor of the sample; and
n = Number of tablets in the sample.

    (2) Loss on drying. Proceed as directed in Sec. 436.200(c) of this 
chapter, using a sample weight of 1 to 2 grams.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q (the amount of clarithromycin dissolved) is 80 
percent at 30 minutes.
    (4) Identity. Using the high-performance liquid chromatographic 
procedure described in paragraph (b)(1) of this section, the retention 
time for the peak of the active ingredient must be within 2 percent of 
the retention time for the peak of the corresponding reference standard.
[58 FR 26654, May 4, 1993. Redesignated at 61 FR 34726, July 3, 1996]



Sec. 452.150b  Clarithromycin granules for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clarithromycin granules for oral 
suspension is a dry mixture containing clarithromycin-coated particles, 
suitable and harmless dispersing agents, diluents, preservatives, and 
flavorings. It contains the equivalent of 25 or 50 milligrams of 
clarithromycin activity per milliliter of the reconstituted suspension. 
Its potency is satisfactory if it is not less than 90 percent and not 
more than 115 percent of the number of milligrams of clarithromycin that 
it is represented to contain. Its loss on drying is not more than 2.0 
percent. When constituted as directed in the labeling, its pH is not 
less than 4.0 nor more than 5.4. The clarithromycin used conforms to the 
standards prescribed by Sec. 452.50(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The clarithromycin used in making the batch for potency, 
moisture, pH, residue on ignition, heavy metals, specific rotation, 
identity, and crystallinity.
    (B) The batch for content, loss on drying, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The clarithromycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Clarithromycin content. Proceed 
as directed in Sec. 452.50(b)(1), except use a known injection volume 
between 10 and 60 microliters. Also, prepare the mobile phase, working 
standard solution, and sample solution, and use system suitability 
requirements and calculation as follows:
    (i) Mobile phase. Add 600 milliliters of methanol and 400 
milliliters of 0.067M potassium phosphate, monobasic, to a suitable 
container, mix well, and adjust the pH to 3.5 with phosphoric acid. 
Filter through a suitable filter capable of removing particulate matter 
to 0.5 micron in diameter. Degas the mobile phase just before its 
introduction into the chromatographic system.
    (ii) Preparation of standard solution. Dissolve an accurately 
weighed portion of the clarithromycin working standard in sufficient 
methanol to obtain a solution having a known concentration of 
approximately 2.1 milligrams per milliliter of clarithromycin. 
Quantitatively transfer and dilute an aliquot of this solution with 
mobile phase and mix to obtain a solution of known

[[Page 937]]

concentration of approximately 415 micrograms of clarithromycin per 
milliliter.
    (iii) Preparation of sample solution. Constitute as directed in the 
labeling. Accurately measure a representative portion of the suspension 
that contains about 1 to 2 grams of clarithromycin activity and, using 
approximately 330 milliliters of 0.067M potassium phosphate, dibasic, 
quantitatively transfer into a 1,000 milliliter volumetric flask 
containing approximately 50 milliliters of 0.067M potassium phosphate, 
dibasic. Shake for 30 minutes. Dilute to volume with methanol. Mix well 
and place in an ultrasonic bath for 30 minutes. Cool to room temperature 
and adjust to volume with methanol. Add a magnetic stirring bar and stir 
for 60 minutes. Allow excipients to settle and dilute an appropriate 
aliquot of the solution with mobile phase to obtain a solution 
containing 500 micrograms of clarithromycin activity per milliliter and 
mix well. Filter through a suitable filter capable of removing 
particulate matter 0.5 micron in diameter.
    (iv) System suitability requirements--(A) Tailing factor. The 
tailing factor (T) is satisfactory if it is not less than 1.0 and not 
greater than 1.7 for the clarithromycin peak.
    (B) Efficiency of the column. The efficiency (n) is satisfactory if 
it is greater than 2,100 theoretical plates for the clarithromycin peak.
    (C) Capacity factor. The capacity factor (k') is satisfactory if it 
is between 2.5 and 6 for the clarithromycin peak.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent of three replicate 
injections) is satisfactory if it is not more than 2.0 percent.
    (v) Calculations. Calculate the clarithromycin content as follows:

                                                                        
                                                                        
       Milligrams of                             AU X PS X D            
    clarithromycin per         =    ------------------------------------
        milliliter                                  AS X V              
                                                                        

where:
AU = Area of the clarithromycin peak in the chromatogram of 
the sample;
AS = Area of the clarithromycin peak in the chromatogram of 
the clarithromycin working standard;
PS = Clarithromycin activity in the clarithromycin working 
standard solution in micrograms per milliliter;
D = Dilution factor of the sample test solution; and
V = Volume, in milliliters, of the portion of suspension taken.
    (2) Loss on drying. Proceed as directed in Sec. 436.200(a) of this 
chapter, using a sample weight of approximately 1 gram, weighing in a 
normal laboratory atmosphere.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the suspension prepared as directed in the labeling. Stir the suspension 
for 10 minutes with the electrode immersed and record the pH.
    (4) Identity. Using the high-performance liquid chromatographic 
procedure described in paragraph (b)(1) of this section, the retention 
times for the clarithromycin peak must be within 2 percent of the 
retention time for the peak of the reference standard.
[61 FR 34726, July 3, 1996]



Sec. 452.160  Azithromycin oral dosage forms.



Sec. 452.160a  Azithromycin capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Azithromycin capsules are composed of 
azithromycin and one or more suitable and harmless diluents, 
disintegrants, lubricants, and wetting agents enclosed in a gelatin 
capsule. Each capsule contains azithromycin equivalent to 250 milligrams 
of azithromycin. The azithromycin content is satisfactory if it is not 
less than 90 percent and not more than 110 percent of the number of 
milligrams of azithromycin that it is represented to contain. The 
moisture content of the capsules is not more than 5.0 percent. The 
capsules pass the dissolution test.

[[Page 938]]

The capsules pass the identity test. The azithromycin used conforms to 
the standards prescribed by Sec. 452.60(a)(1) of this part.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain;
    (i) Results of tests and assays on:
    (A) The azithromycin used in making the batch for potency, moisture, 
pH, residue on ignition, heavy metals, specific rotation, crystallinity, 
and identity.
    (B) The batch for content, moisture, dissolution, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The azithromycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch: A minimum of 100 capsules.
    (b) Tests and methods of assay--(1) Azithromycin content. Proceed as 
directed in Sec. 452.60(b)(1), preparing the sample solution and 
calculating the azithromycin content as follows:
    (i) Preparation of sample solution. Quantitatively transfer the 
contents of one capsule into a 250-milliliter volumetric flask. Add 
about 175 milliliters of acetonitrile and shake on a reciprocating 
shaker for 30 minutes. Dilute to volume with acetonitrile, stopper the 
flask and mix well. Place 40 milliliters of the resulting suspension 
into a suitably sized centrifuge tube. Stopper the tube and centrifuge 
the suspension (about 10 minutes at 1,000 rpm). Pipet 2.0 milliliters of 
the supernatant into a 50-milliliter volumetric flask and dilute to 
volume with the mobile phase. Pipet 2.0 milliliters of this solution 
into a 25-milliliter volumetric flask and dilute to volume with mobile 
phase. The final dilution of the sample and standard must be identical. 
The final concentration of azithromycin in the sample solution is 0.003 
milligram per milliliter (estimated).
    (ii) Calculations. Calculate the azithromycin content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.296
    
where:
AU = Area of the azithromycin peak (at a retention time equal 
          to that observed for the azithromycin standard) in the 
          chromatogram of the sample;
AS = Area of the azithromycin peak of the azithromycin 
          working standard;
PS = Azithromycin activity in the azithromycin working 
          standard solution in micrograms per milliliter; and
d = Dilution factor of the sample = 250 x 50 x 25/2 x 2.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Dissolution test. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q (the percentage of azithromycin activity 
dissolved) is 75 percent within 45 minutes.
    (4) Identity. Using the high-performance liquid chromatographic 
procedure described in paragraph (b)(1) of this section the retention 
time for the peak of the active ingredient must be with 2 percent of the 
retention time for the peak of the corresponding reference standard.
[58 FR 26658, May 4, 1993. Redesignated at 59 FR 52078, Oct. 14, 1994.]



Sec. 452.160b  Azithromycin for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Azithromycin for oral suspension is a dry 
mixture of azithromycin with a suitable and harmless buffer substance, 
sweetener, diluent, anticaking agent, and flavorings. The dry mixture is 
packaged in single dose packets each containing 1,000 milligrams of 
azithromycin. The azithromycin content is satisfactory if it is not less 
than 90 percent and not more than 110 percent of the number of 
milligrams of azithromycin that it is represented to contain. Its 
moisture content is not more than 1.5 percent. When constituted as 
directed in the labeling, the pH of the suspension is not less than 9 
and not more than 11. It gives a positive identity test for 
azithromycin. The azithromycin used conforms to the standards prescribed 
by Sec. 452.60(a)(1).

[[Page 939]]

    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain;
    (i) Results of tests and assays on:
    (A) The azithromycin used in making the batch for potency, moisture, 
pH, residue on ignition, heavy metals, specific rotation, crystallinity, 
and identity.
    (B) The batch for content, moisture, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The azithromycin used in making the batch: 10 packages, each 
containing approximately 1,000 milligrams.
    (B) The batch: A minimum of 30 packages.
    (b) Tests and methods of assay--(1) Azithromycin content. Proceed as 
directed in Sec. 452.60(b)(1), preparing the dissolving solvent and 
sample solution and calculating the azithromycin content as follows:
    (i) Dissolving solvent. Dissolve 2.2 grams of potassium phosphate 
monobasic in 1,590 milliliters of ultrapure deionized or high-
performance liquid chromatographic-grade water. Add 600 milliliters of 
2-propanol, 480 milliliters of ethanol, and 330 milliliters of 
acetonitrile, adjust to pH 8.4 with 10M potassium hydroxide and shake on 
a reciprocating shaker for 30 minutes. The dissolving solvent is 0.01M 
monobasic potassium phosphate:2-propanol:ethanol:acetonitrile 
(53:20:16:11, by volume).
    (ii) Preparation of sample solution. Quantitatively transfer the 
contents of one package into a 500-milliliter volumetric flask. Add 
about 350 milliliters of dissolving solvent and shake on a reciprocating 
shaker for 30 minutes. Dilute to volume with dissolving solvent, stopper 
the flask, and mix well. Place 40 milliliters of the resulting 
suspension into a suitably sized centrifuge tube. Stopper the tube and 
centrifuge the suspension (about 20 minutes at 1,000 revolutions per 
minute). Pipet 10.0 milliliters of the diluted solution into a 50-
milliliter volumetric flask and dilute to volume with mobile phase 
(described in Sec. 452.60(b)(1)(i)). Pipet 2.0 milliliters of the 
diluted solution into a 50-milliliter volumetric flask and dilute to 
volume with mobile phase. The final dilution of the sample and standard 
must be identical. The final concentration of azithromycin in the sample 
solution is 0.003 milligram per milliliter (estimated).
    (iii) Calculations. Calculate the azithromycin content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.297
    
where:
AU = Area of the azithromycin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          azithromycin standard);
AS = Area of the azithromycin peak in the chromatogram of the 
          azithromycin working standard;
PS = Azithromycin activity in the azithromycin working 
          standard solution in micrograms per milliliter; and
d = Dilution factor of the sample = 500 X 50/10 X 50/10 X 50/2.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug constituted as directed in the labeling. Allow the constituted 
suspension to sit for 10 minutes undisturbed before making the 
measurement.
    (4) Identity. Using the high-performance liquid chromatographic 
procedure described in paragraph (b)(1) of this section, the retention 
time for the peak of the active ingredient must be within 2 percent of 
the retention time for the peak of the corresponding reference standard.
[59 FR 52078, Oct. 14, 1994]



Sec. 452.175  Troleandomycin oral dosage forms.



Sec. 452.175a  Troleandomycin capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Troleandomycin capsules are capsules 
composed of troleandomycin and one or more suitable buffers, diluents, 
binders, lubricants, and colorings. Each capsule contains 125 milligrams 
or 250 milligrams

[[Page 940]]

of troleandomycin. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
troleandomycin that it is represented to contain. The loss on drying is 
not more than 5 percent. The troleandomycin used conforms to the 
standards prescribed by Sec. 452.75(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The troleandomycin used in making the batch for potency, loss on 
drying, pH, residue on ignition, identity, Rf value, acetyl 
value (only if more than one spot is present in the determination of 
Rf value), and crystallinity.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:
    (a) The troleandomycin used in making the batch: 10 packages, nine 
containing approximately equal portions of not less than 500 milligrams 
and one containing not less than 2 grams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing sufficient 80 percent isopropyl alcohol solution 
(solution 15) to obtain a stock solution containing 1,000 micrograms of 
troleandomycin per milliliter (estimated). Blend for 3 to 5 minutes. 
Further dilute an aliquot of the stock solution with distilled water to 
the reference concentration of 25 micrograms of troleandomycin per 
milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[39 FR 19149, May 30, 1974, as amended at 48 FR 3960, Jan. 28, 1983; 50 
FR 19921, May 13, 1985]



Sec. 452.175b  Troleandomycin oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Troleandomycin oral suspension is 
troleandomycin and one or more suitable buffers, dispersants, 
flavorings, colorings, and preservatives suspended in a suitable and 
harmless vehicle. Each milliliter contains 25 milligrams of 
troleandomycin. Its potency is satisfactory if it is not less than 90 
percent and not more than 125 percent of the number of milligrams of 
troleandomycin that it is represented to contain. Its pH is not less 
than 5.0 and not more than 8.0. The troleandomycin used conforms to the 
standards prescribed by Sec. 452.75(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The troleandomycin used in making the batch for potency, loss on 
drying, pH, residue on ignition, identity, Rf value, acetyl 
value (only if more than one spot is present in the determination of 
Rf value), and crystallinity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The troleandomycin used in making the batch: 10 packages, nine 
containing approximately equal portions of not less than 500 milligrams 
and one containing not less than 2 grams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dilute an accurately measured representative portion of the sample with 
80 percent isopropyl alcohol solution (solution 15) to obtain a stock 
solution containing 1,000 micrograms of troleandomycin per milliliter 
(estimated). Further dilute an aliquot of the stock solution with 
distilled water to the reference concentration of 25 micrograms of 
troleandomycin per milliliter (estimated).

[[Page 941]]

    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[39 FR 19149, May 30, 1974, as amended at 48 FR 3960, Jan. 28, 1983; 50 
FR 19921, May 13, 1985]



Sec. 452.175c  Troleandomycin for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Troleandomycin for oral suspension is 
troleandomycin with suitable buffers, dispersants, preservatives, 
colorings, and flavorings. When the suspension is prepared as directed 
in its labeling, each milliliter contains 25 milligrams of 
troleandomycin. However, if it is for pediatric use, each milliliter 
contains 100 milligrams of troleandomycin. Its potency is satisfactory 
if it is not less than 90 percent and not more than 120 percent of the 
number of milligrams of troleandomycin that it is represented to 
contain. Its loss on drying is not more than 2 percent. The pH of the 
suspension, when prepared as directed in its labeling, is not less than 
5.0 and not more than 7.0. The troleandomycin used conforms to the 
standards prescribed by Sec. 452.75(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The troleandomycin used in making the batch for potency, loss on 
drying, pH, residue on ignition, identity, Rf value, acetyl 
value (only if more than one spot is present in the determination of 
Rf value), and crystallinity.
    (b) The batch for potency, loss on drying, and pH.
    (ii) Samples required:
    (a) The troleandomycin used in making the batch: 10 packages, nine 
containing approximately equal portions of not less than 500 milligrams 
and one containing not less than 2 grams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute the drug as directed in the labeling. Dilute an accurately 
measured representative portion of the sample with sufficient 80 percent 
isopropyl alcohol solution (solution 15) to obtain a stock solution 
containing 1,000 micrograms of troleandomycin per milliliter 
(estimated). Further dilute an aliquot of the stock solution with 
distilled water to the reference concentration of 25 micrograms of 
troleandomycin per milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the suspension obtained after reconstituting the drug as directed in its 
labeling.
[39 FR 19149, May 30, 1974, as amended at 48 FR 3960, Jan. 28, 1983; 50 
FR 19921, May 13, 1985]



Sec. 452.175d  Troleandomycin chewable tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Each troleandomycin chewable tablet 
contains an amount equivalent to 125 milligrams of troleandomycin with 
suitable diluents, binders, buffers, colorings, and flavorings. Its 
potency is satisfactory if it is not less than 90 percent and not more 
than 125 percent of the number of milligrams of troleandomycin that it 
is represented to contain. The loss on drying is not more than 5 
percent. The troleandomycin used conforms to the standards prescribed by 
Sec. 452.75(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The troleandomycin used in making the batch for potency, loss on 
drying, pH, residue on ignition, identity, Rf value, acetyl 
value (only if more than one spot is present in the determination of 
Rf value), and crystallinity.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:

[[Page 942]]

    (a) The troleandomycin used in making the batch: 10 packages, nine 
containing approximately 500 milligrams each and one containing 
approximately 2 grams.
    (b) The batch: A minimum of 30 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of tablets into a high-speed glass blender 
jar with sufficient 80 percent isopropyl alcohol solution (solution 15) 
to obtain a stock solution containing 1,000 micrograms of troleandomycin 
per milliliter (estimated). Blend 3 to 5 minutes. Further dilute an 
aliquot of the stock solution with distilled water to the reference 
concentration of 25 micrograms of troleandomycin per milliliter 
(estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[39 FR 19149, May 30, 1974, as amended at 48 FR 3960, Jan. 28, 1983; 48 
FR 36571, Aug. 12, 1983; 50 FR 19921, May 13, 1985]



                   Subpart C--Injectable Dosage Forms



Sec. 452.225  Erythromycin ethylsuccinate injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin ethylsuccinate injection is 
erythromycin ethylsuccinate and butylaminobenzoate dissolved in 
polyethylene glycol 400. It contains a suitable and harmless 
preservative. Each milliliter contains 50 milligrams of erythromycin. 
Its potency is satisfactory if it is not less than 90 percent and not 
more than 115 percent of the number of milligrams of erythromycin that 
it is represented to contain. It contains 2 percent butylaminobenzoate. 
It is sterile. Its moisture content is not more than 1.5 percent. The 
erythromycin ethylsuccinate used conforms to the standards prescribed 
therefore by Sec. 452.25a(a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, each immediate container shall bear on its 
label and labeling the statement: ``Warning--For intramuscular use 
only''.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin ethylsuccinate used in making the batch for 
potency, moisture, pH, residue on ignition, identity, and crystallinity.
    (b) The batch for potency, sterility, and moisture.
    (ii) Samples required:
    (a) The erythromycin ethylsuccinate used in making the batch: 10 
packages, each containing 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation, except that if the 
product is sterilized after filling, a representative sample consisting 
of 10 immediate containers from each sterilizer load. If only one 
sterilizer load is involved, the sample shall consist of 20 immediate 
containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
By means of a suitable hypodermic needle and syringe, remove an 
accurately measured representative volume of the sample and dilute with 
sufficient methyl alcohol to give a solution containing 1.0 milligram of 
erythromycin base per milliliter (estimated). Further dilute with 0.1M 
potassium phosphate buffer, pH 8.0 (solution 3), to the reference 
concentration of 1.0 microgram of erythromycin base per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use a bacterial-retentive membrane resistant to the solvent polyethylene 
glycol 400 and add 1 milliliter from each immediate container directly 
to the membrane, thus eliminating the preliminary solubilization step.
    (3) [Reserved]

[[Page 943]]

    (4) Moisture. Proceed as directed in Sec. 436.201(e)(1) of this 
chapter.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19920, 19921, May 13, 
1985]



Sec. 452.230  Sterile erythromycin gluceptate.

    The requirements for certification and the tests and methods of 
assay for sterile erythromycin gluceptate packaged for dispensing are 
described in Sec. 452.30a.
[46 FR 16685, Mar. 13, 1981]



Sec. 452.232  Erythromycin lactobionate injectable dosage forms.



Sec. 452.232a  Erythromycin lactobionate for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin lactobionate for injection 
is a dry mixture of erythromycin lactobionate and a suitable 
preservative. It contains the equivalent of 300 milligrams, 500 
milligrams, or 1 gram of erythromycin per vial. Its potency is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of erythromycin that it is 
represented to contain. It is sterile. It is nonpyrogenic. Its moisture 
content is not more than 5 percent. Its pH is not less than 6.5 and not 
more than 7.5. The erythromycin used conforms to the standards 
prescribed by Sec. 452.10(a)(1) (i), (iii), (iv), (v), (vi), (vii), and 
(viii).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin used in making the batch for potency, pH, 
moisture, residue on ignition, heavy metals, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, moisture, pH, and 
identity.
    (ii) Samples required:
    (a) The erythromycin used in making the batch: 10 containers, each 
consisting of not less than 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 12 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Using a suitable hypodermic 
needle and syringe, remove the total withdrawable contents from each 
container represented as a single-dose container; or if the labeling 
specifies the amount of potency in a given volume of the preparation, 
withdraw an accurately measured volume from each container. Dilute with 
sterile distilled water to obtain a concentration of 10 milligrams of 
erythromycin base per milliliter (estimated). Further dilute with 0.1M 
potassium phosphate buffer, pH 8.0 (solution 3), to the reference 
concentration of 1.0 microgram of erythromycin base per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 30 milligrams of erythromycin per 
milliliter.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
concentration of 50 milligrams of erythromycin per milliliter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation method described in paragraph (b)(2) of 
that section.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19920, 19921, May 13, 
1985. Redesignated at 51 FR 35216, Oct. 2, 1986]



Sec. 452.232b  Sterile erythromycin lactobionate.

    The requirements for certification and the tests and methods of 
assay for sterile erythromycin lactobionate packaged for dispensing are 
described in Sec. 452.32a.
[51 FR 35216, Oct. 2, 1986]

[[Page 944]]



                   Subpart D--Ophthalmic Dosage Forms



Sec. 452.310  Erythromycin ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin ophthalmic ointment is 
erythromycin in a suitable and harmless ointment base. Each gram of 
ointment contains 5 milligrams of erythromycin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of erythromycin that it is 
represented to contain. It is sterile. The moisture content is not more 
than 1 percent. The erythromycin used conforms to the standards 
prescribed by Sec. 452.10(a)(1) (i), (iii), (iv), (v), (vii), and 
(viii).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin used in making the batch for potency, pH, 
moisture, residue on ignition, crystallinity, and identity.
    (b) The batch for potency, sterility, and moisture.
    (ii) Samples required:
    (a) The erythromycin used in making the batch: 10 packages, each 
containing 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of five immediate 
containers.
    (2) For sterility testing: Twenty immediate containers, collected at 
regular intervals throughout each filling operation.
    (b)(1) Potency. Proceed as directed in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Place an accurately weighed 
representative portion of the ointment in a separatory funnel containing 
50 milliliters of reagent-grade petroleum ether. Shake until dissolved. 
Wash with four separate washings of a 4:1 mixture of methyl alcohol and 
distilled water. Combine the washings and bring to volume with the 
methyl alcohol-water solution in a volumetric flask. Further dilute with 
0.1M potassium phosphate buffer, pH 8.0 (solution 3), to the reference 
concentration of 1.0 microgram of erythromycin base per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19149, May 30, 1974, as amended at 49 FR 5097, Feb. 10, 1984; 50 
FR 19921, May 13, 1985]



                          Subpart E  [Reserved]



                  Subpart F--Dermatologic Dosage Forms



Sec. 452.510  Erythromycin dermatologic dosage forms.



Sec. 452.510a  Erythromycin ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin ointment is erythromycin in 
a suitable and harmless ointment base. It may contain suitable 
preservatives. Each gram of ointment contains 20 milligrams of 
erythromycin. Its potency is satisfactory if it is not less than 90 
percent and not more than 125 percent of the number of milligrams of 
erythromycin that it is represented to contain. The moisture content is 
not more than 1.0 percent. The erythromycin used conforms to the 
standards prescribed by Sec. 452.10(a)(1) (i), (iii), (iv), (v), (vii), 
and (viii).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin used in making the batch for potency, pH, 
moisture, residue on ignition, crystallinity, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The erythromycin used in making the batch: 10 packages, each 
containing not less than 500 milligrams.

[[Page 945]]

    (b) The batch: A minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the ointment in a 
separatory funnel containing 50 milliliters of reagent-grade petroleum 
ether. Shake until dissolved. Wash with four separate washings of a 4:1 
mixture of methyl alcohol and distilled water. Combine the washings and 
bring to volume with the methyl alcohol-water solution in a volumetric 
flask. Further dilute with 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to the reference concentration of 1.0 microgram of 
erythromycin base per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19149, May 30, 1974, as amended at 49 FR 5097, Feb. 10, 1984; 49 
FR 47829, Dec. 7, 1984; 50 FR 47214, Nov. 15, 1985]



Sec. 452.510b  Erythromycin topical solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin topical solution contains in 
each milliliter 15.0 or 20.0 milligrams of erythromycin. It may also 
contain one or more suitable and harmless solvents, surfactants, buffer 
substances, diluents, and perfumes. Its potency is satisfactory if it is 
not less than 90 percent and not more than 125 percent of the number of 
milligrams of erythromycin that it is represented to contain. If it 
contains 15.0 milligrams of erythromycin per milliliter, its moisture 
content is not more than 5.0 percent. If it contains 20.0 milligrams of 
erythromycin per milliliter, its moisture content is not more than 8.0 
percent, except if it contains acetone, its moisture content is not more 
than 2.0 percent. The erythromycin used conforms to the standards 
prescribed by Sec. 452.10(a)(1), except heavy metals.
    (2) Packaging. In addition to the requirements of Sec. 432.1 of this 
chapter, it may either be dispensed on individually packaged pledgets, 
each individual pledget containing 0.8 milliliter of erythromycin 
topical solution, or in a jar containing 60 pledgets. The jar contains 
0.8 milliliter of erythromycin topical solution per pledget. Although 
the pledgets in the jar are not individually packaged, the drug is 
uniformly distributed throughout the pledgets. The erythromycin topical 
solution used on the pledgets contains 20 milligrams of erythromycin per 
milliliter.
    (3) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The erythromycin used in making the batch for potency, moisture, 
pH, residue on ignition, identity, and crystallinity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The erythromycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch: A minimum of 6 immediate containers.
    (b) Tests and methods of assay. If the erythromycin topical solution 
is dispensed on a pledget, express the contents of a representative 
number of pledgets into a suitable container to obtain a volume of 
sample adequate to perform each assay described in paragraph (b)(1) and 
(2) of this section.
    (1) Potency. Proceed as directed in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Using a suitable hypodermic 
needle and syringe, remove an accurately measured representative portion 
of the sample and dilute with 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to obtain a stock solution of convenient concentration. 
Further dilute an aliquot of the stock solution with solution 3 to the 
reference concentration of 1.0 microgram of erythromycin base per 
milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
except if the

[[Page 946]]

sample contains acetone, in lieu of Solvent A, use a mixture of pyridine 
and methanol (1:1).
[46 FR 2995, Jan. 13, 1981, as amended at 48 FR 51293, Nov. 8, 1983; 49 
FR 374, Jan. 4, 1984; 50 FR 1504, Jan. 11, 1985; 50 FR 19921, May 13, 
1985; 50 FR 20204, May 15, 1985; 54 FR 47352, Nov. 14, 1989; 54 FR 
50472, Dec. 6, 1989]



Sec. 452.510d  Erythromycin-benzoyl peroxide topical gel.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin-benzoyl peroxide topical gel 
is erythromycin packaged in combination with a suitable and harmless gel 
containing benzoyl peroxide and one or more suitable dispersants, 
stabilizing agents, perfumes, and wetting agents. When reconstituted as 
directed in the labeling, each gram contains 30 milligrams of 
erythromycin and 50 milligrams of benzoyl peroxide. The erythromycin 
content is satisfactory if it contains not less than 90 percent and not 
more than 125 percent of the number of milligrams of erythromycin that 
it is represented to contain. The benzoyl peroxide content is 
satisfactory if it contains not less than 90 percent and not more than 
115 percent of the milligrams of benzoyl peroxide that it is represented 
to contain. The erythromycin used conforms to the standards prescribed 
by Sec. 452.10(a)(1), except with respect to heavy metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The erythromycin used in making the batch for potency, moisture, 
pH, residue on ignition, identity, and crystallinity.
    (b) The batch for erythromycin content and benzoyl peroxide content.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The erythromycin used in making the batch: 5 packages, each 
containing approximately 100 milligrams.
    (b) The batch: A minimum of 8 containers.
    (b) Tests and methods of assay--(1) Erythromycin content. Proceed as 
directed in Sec. 436.105 of this chapter, preparing the sample for assay 
as follows: Reconstitute the sample as directed in the labeling. Place 
an accurately weighed representative portion of the constituted product 
into a high-speed glass blender jar containing 0.5 milliliter of 
polysorbate 80 and sufficient 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to obtain a stock solution of convenient concentration. 
Blend for 3 to 5 minutes. Further dilute an aliquot of the stock 
solution with solution 3 to the reference concentration of 1.0 microgram 
of erythromycin base per milliliter (estimated).
    (2) Benzoyl peroxide content. Reconstitute the sample as directed in 
the labeling. Place an accurately weighted representative portion (about 
2.5 grams) of the constituted product into a tared 250-milliliter glass-
stoppered flask. Add 50 milliliters of glacial acetic acid and 20 
milliliters of methylene chloride. Stopper and shake vigorously to 
disperse the gel. Add 1.0 milliliter phenylsulfide, swirl, stopper, and 
allow to stand at room temperature for 2 minutes. Purge the flask with 
nitrogen for 3 seconds. Add 5 milliliters for freshly prepared saturated 
sodium iodide solution, stopper, and swirl to mix. Let stand in the dark 
for 30 minutes. Add 50 milliliters of previously boiled and cooled 
distilled water and titrate the liberated iodine with 0.1N sodium 
thiosulfate, adding starch T.S. near the endpoint. Perform a blank 
determination and correct the sample titer. Each milliliter of 0.1N 
sodium thiosulfate is equivalent to 12.11 milligrams of benzoyl 
peroxide. Calculate the benzoyl peroxide content as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.298

where:

Vu=Milliliters of sodium thiosulfate used in the titration of 
          the sample minus the milliliters of sodium thiosulfate used in 
          the titration of the sample blank.
[49 FR 47485, Dec. 5, 1984; 49 FR 49090, Dec. 18, 1984; 49 FR 49449, 
Dec. 20, 1984, as amended at 55 FR 11584, Mar. 29, 1990]

[[Page 947]]



Sec. 452.510e  Erythromycin topical gel.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin topical gel is erythromycin 
in a suitable and harmless gel. Each gram contains 20 milligrams of 
erythromycin. The erythromycin content is satisfactory if it contains 
not less than 90 percent and not more than 125 percent of the number of 
milligrams of erythromycin that it is represented to contain. The 
erythromycin used conforms to the standards prescribed by 
Sec. 452.10(a)(1), except with respect to heavy metals.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each request shall 
contain:
    (i) Results of tests and assays on:
    (A) The erythromycin used in making the batch for potency, moisture, 
pH, residue on ignition, identity, and crystallinity.
    (B) The batch for erythromycin content.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The erythromycin used in making the batch: 5 packages, each 
containing approximately 100 milligrams.
    (B) The batch: A minimum of 8 containers.
    (b) Tests and methods of assay; erythromycin content. Proceed as 
directed in Sec. 436.105 of this chapter, preparing the sample for assay 
as follows: Place approximately 1 gram, accurately weighed, of the 
product into a high-speed glass blender jar containing 200 milliliters 
of 0.5 percent (volume by volume) polysorbate 80 in 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), to obtain a stock solution of 
convenient concentration. Blend for 3 to 5 minutes. Further dilute an 
aliquot of the stock solution with solution 3 to the reference 
concentration of 1.0 microgram of erythromycin base per milliliter 
(estimated).
[53 FR 12415, Apr. 14, 1988; 53 FR 16837, May 11, 1988]



                          Subpart G  [Reserved]



                     Subpart H--Rectal Dosage Forms



Sec. 452.710  Erythromycin suppositories.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin suppositories contain in 
each suppository 125 milligrams of erythromycin in a suitable and 
harmless base. The erythromycin content is satisfactory if it is not 
less than 90 percent nor more than 120 percent of the number of 
milligrams of erythromycin that it is represented to contain. The 
moisture content is not more than 1.0 percent. The erythromycin used 
conforms to the standards prescribed by Sec. 452.10(a)(1), (i), (iii), 
(iv), (v), (vii), and (viii), except its moisture content is not more 
than 5.0 percent.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The erythromycin used in making the batch for potency, moisture, 
pH, residue on ignition, identity, and crystallinity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The erythromycin used in making the batch: 10 packages, each 
containing not less than 500 milligrams.
    (b) The batch: A minimum of 30 suppositories.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Blend a representative number of suppositories for 3 to 5 minutes in a 
high-speed glass blender with 200 milliliters of methyl alcohol. Add 300 
milliliters of 0.1M potassium phosphate buffer, pH 8.0 (solution 3), and 
blend again for 3 to 5 minutes. Remove an aliquot and dilute with 
solution 3 to the reference concentration of 1.0 microgram of 
erythromycin base per milliliter (estimated).

[[Page 948]]

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19149, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



                          Subpart I  [Reserved]



                  Subpart J--Certain Other Dosage Forms



Sec. 452.910  Erythromycin for prescription compounding.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin for prescription compounding 
is the odorless, white to grayish-white or slightly yellow compound of a 
kind of erythromycin or a mixture of two or more such compounds. It is 
so purified and dried that:
    (i) It contains not less than 850 micrograms of erythromycin per 
milligram calculated on an anhydrous basis.
    (ii) Its moisture content is not more than 10 percent.
    (iii) Its pH is not less than 8.0 nor more than 10.5.
    (iv) Its residue on ignition is not more than 2.0 percent.
    (v) It gives a positive identity test for erythromycin.
    (vi) It is crystalline.
    (2) Packaging. The immediate container shall be a tight container as 
defined by the United States Pharmacopeia XXI. It shall be so sealed 
that the contents cannot be used without destroying such seal. Each such 
container shall contain 10 grams, 25 grams, or 100 grams of 
erythromycin.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its outside wrapper or 
container and on the immediate container the following:
    (i) The statement ``Caution: Federal law prohibits dispensing 
without prescription.''
    (ii) The statement ``Not sterile.''
    (iii) The batch mark.
    (iv) The number of micrograms of erythromycin activity in each 
milligram of erythromycin and the number of grams of erythromycin in the 
immediate container.
    (v) The statement ``The potency of this drug cannot be assured for 
longer than 90 days after the container is first opened for compounding 
a prescription.''
    (vi) The statements ``For use only in extemporaneous prescription 
compounding. Not for manufacturing use.''
    (4) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, residue on ignition, identity, and crystallinity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages, each containing not less than 500 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient methyl alcohol to 
obtain a concentration of 10 milligrams of erythromycin base per 
milliliter (estimated). Dilute this solution further with sufficient 
0.1M potassium phosphate buffer, pH 8.0 (solution 3), to obtain a stock 
solution containing 1.0 milligram of erythromycin base per milliliter 
(estimated). Further dilute an aliquot of the stock solution with 
solution 3 to the reference concentration of 1.0 microgram of 
erythromycin base per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, except 
standardize the pH meter with pH 7.0 and pH 10.0 buffers and prepare the 
sample as follows: Dissolve 200 milligrams of sample in 5 milliliters of 
reagent grade methyl alcohol. Add 95 milliliters of water and mix. 
Record the pH when an equilibrium value has been reached.
    (4) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (5) Identity test. Proceed as directed in Sec. 436.211 of this 
chapter, using the sample preparation method described in paragraph 
(b)(3) of that section.
    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[51 FR 35213, Oct. 2, 1986, as amended at 55 FR 11584, Mar. 29, 1990; 55 
FR 25392, June 21, 1990]

[[Page 949]]



PART 453--LINCOMYCIN ANTIBIOTIC DRUGS--Table of Contents




                          Subpart A--Bulk Drugs

Sec.
453.20  Clindamycin hydrochloride hydrate.
453.21  Clindamycin palmitate hydrochloride.
453.22  Clindamycin phosphate.
453.22a  Sterile clindamycin phosphate.
453.30  Lincomycin hydrochloride monohydrate.
453.30a  Sterile lincomycin hydrochloride monohydrate.

                      Subpart B--Oral Dosage Forms

453.120  Clindamycin hydrochloride hydrate capsules.
453.121  Clindamycin palmitate hydrochloride oral dosage forms.
453.121a  Clindamycin palmitate hydrochloride for oral suspension.
453.121b  Clindamycin palmitate hydrochloride for oral solution.
453.130  Lincomycin hydrochloride oral dosage forms.
453.130a  Lincomycin hydrochloride monohydrate capsules.
453.130b  Lincomycin hydrochloride syrup.

                   Subpart C--Injectable Dosage Forms

453.222  Clindamycin phosphate injection.
453.230  Lincomycin hydrochloride injection.

                        Subparts D-E  [Reserved]

                  Subpart F--Dermatologic Dosage Forms

453.522  Clindamycin phosphate dermatologic dosage forms.
453.522a  Clindamycin phosphate topical solution.
453.522b  Clindamycin phosphate gel.
453.522c  Clindamycin phosphate lotion.
453.522d  Clindamycin phosphate vaginal cream.

    Authority:  21 U.S.C. 357.



                          Subpart A--Bulk Drugs



Sec. 453.20  Clindamycin hydrochloride hydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clindamycin hydrochloride hydrate is the 
hydrated hydrochloride salt of clindamycin. It is so purified and dried 
that:
    (i) Its clindamycin content is not less than 800 micrograms of 
clindamycin per milligram.
    (ii) Its microbiological activity is not less than 800 micrograms of 
clindamycin per milligram.
    (iii) [Reserved]
    (iv) Its moisture content is not less than 3.0 percent and not more 
than 6.0 percent.
    (v) Its pH in an aqueous solution containing 100 milligrams per 
milliliter is not less than 3.0 and not more than 5.5.
    (vi) It is crystalline.
    (vii) It passes the identity test for clindamycin hydrochloride 
hydrate.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for clindamycin 
content, microbiological activity, moisture, pH, crystallinity, and 
identity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Clindamycin content (vapor phase 
chromatography). Proceed as directed in Sec. 436.302 of this chapter.
    (2) Microbiological activity (microbiological agar diffusion assay.) 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Dissolve an accurately weighed sample in 
sufficient sterile distilled water to give a stock solution of 
convenient concentration. Further dilute the stock solution with 0.1M 
potassium phosphate buffer, pH 8.0 (solution 3), to the reference 
concentration of 1.0 microgram of clindamycin per milliliter 
(estimated).
    (3) [Reserved]
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (6) Crystallinity. Proceed as directed in Sec. 436.203 of this 
chapter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation method described in paragraph (b)(2) of 
that section.
[39 FR 19161, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]

[[Page 950]]



Sec. 453.21  Clindamycin palmitate hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clindamycin palmitate hydrochloride is 
the white to off-white amorphous powder of the hydrochloride salt of the 
palmitic acid ester of clindamycin. It is freely soluble in water, 
ethanol, chloroform, and ether. It is so purified and dried that:
    (i) It contains not less than 540 micrograms of clindamycin per 
milligram.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 3.0 percent.
    (iv) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 2.8 and not more than 3.8.
    (v) It passes the identity test for clindamycin palmitate 
hydrochloride.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for clindamycin 
content, moisture, pH, and identity.
    (ii) Samples required: 10 packages, nine containing not less than 
300 milligrams and one package containing not less than 2 grams.
    (b) Tests and methods of assay--(1) Clindamycin content. Proceed as 
directed in Sec. 436.303 of this chapter.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (5) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation method described in paragraph (b)(2) of 
that section.
[39 FR 19161, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 453.22  Clindamycin phosphate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clindamycin phosphate is a water-soluble 
ester of clindamycin and phosphoric acid. It occurs as a white to off-
white powder. It is so purified and dried that:
    (i) Its clindamycin content is not less than 758 micrograms of 
clindamycin per milligram calculated on an anhydrous basis.
    (ii) Its microbiological activity is not less than 758 micrograms of 
clindamycin per milligram calculated on an anhydrous basis.
    (iii) [Reserved]
    (iv) Its moisture content is not more than 6.0 percent.
    (v) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 3.5 and not more than 4.5.
    (vi) It is crystalline.
    (vii) It passes the identity test for clindamycin phosphate.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for clindamycin 
content, microbiological activity, moisture, pH, crystallinity, and 
identity.
    (ii) Samples required: 10 packages, nine containing approximately 
300 milligrams and one containing 1.5 grams.
    (b) Tests and methods of assay--(1) Clindamycin content (vapor phase 
chromatography). Proceed as directed in Sec. 436.304 of this chapter.
    (2) Microbiological activity (microbiological agar diffusion assay). 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Accurately weigh approximately 12 
milligrams of the clindamycin phosphate sample into a 50-milliliter 
glass-stoppered centrifuge tube. Pipet 25 milliliters of the pH 9.0 
borate buffer into the centrifuge tube. Add 10 milliliters of chloroform 
and shake vigorously for 15 minutes. Centrifuge the resulting mixture 
and pipet a 20-milliliter aliquot of the aqueous phase into a 35-
milliliter centrifuge tube. Add a weighed amount of intestinal alkaline 
phosphatase equivalent to 50 units of

[[Page 951]]

activity\1\ and allow the solution to stand until the enzyme has 
completely dissolved. Place the tube into a water bath at 37 deg. C 
plus-minus2 deg. C for 2.5 hours. After the 2.5-hours 
hydrolysis, allow the solution to cool. Further dilute an aliquot of the 
solution with 0.1M potassium phosphate buffer, pH 8.0 (solution 3), to 
the reference concentration of 1.0 microgram of clindamycin per 
milliliter (estimated).
---------------------------------------------------------------------------

    \1\ Defined such that 50 units hydrolyzes at least 20 micromoles of 
a clindamycin phosphate authentic sample under the assay conditions 
described in this section.
---------------------------------------------------------------------------

    (3) [Reserved]
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (6) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation method described in paragraph (b)(2) of 
that section, except dry the sample for 2 hours at 100 deg. C and allow 
to equilibrate with the atmosphere for 1 hour.
[46 FR 2996, Jan. 13, 1981, as amended at 50 FR 19921, May 13, 1985]



Sec. 453.22a  Sterile clindamycin phosphate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile clindamycin phosphate is a water-
soluble ester of clindamycin and phosphoric acid. It occurs as a white 
to off-white powder. It is so purified and dried that:
    (i) Its clindamycin content is not less than 758 micrograms of 
clindamycin per milligram calculated on an anhydrous basis.
    (ii) Its microbiological activity is not less than 758 micrograms of 
clindamycin per milligram calculated on an anhydrous basis.
    (iii) It is sterile.
    (iv) It is nonpyrogenic.
    (v) [Reserved]
    (vi) It contains no depressor substances.
    (vii) Its moisture content is not more than 6 percent.
    (viii) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 3.5 and not more than 4.5.
    (ix) It is crystalline.
    (x) It passes the identity test for clindamycin phosphate.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for clindamycin 
content, microbiological activity, sterility, pyrogens, depressor 
substances, moisture, pH, crystallinity, and identity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, nine containing 
approximately 300 milligrams and one containing 1.5 grams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Clindamycin content (vapor phase 
chromatography). Proceed as directed in Sec. 436.304 of this chapter.
    (2) Microbiological activity (microbiological agar diffusion assay). 
Proceed as directed in Sec. 436.105 of this chapter, preparing the 
sample for assay as follows: Accurately weigh approximately 12 
milligrams of the clindamycin phosphate sample into a 50-milliliter 
glass-stoppered centrifuge tube. Pipet 25 milliliters of the pH 9.0 
borate buffer into the centrifuge tube. Add 10 milliliters of chloroform 
and shake vigorously for 15 minutes. Centrifuge the resulting mixture 
and pipet a 20-milliliter aliquot of the aqueous phase into a 35-
milliliter centrifuge tube. Add a weighed amount of intestinal alkaline 
phosphatase equivalent to 50 units of activity 1 and allow 
the solution to stand until the enzyme has completely dissolved. Place 
the tube into a water bath at 37 deg. C.plus-minus2 deg. C. 
for 2.5 hours. After the 2.5-hour hydrolysis, allow the solution to 
cool. Further dilute an aliquot of the solution with

[[Page 952]]

0.1M potassium phosphate buffer, pH 8.0 (solution 3), to the reference 
concentration of 1.0 microgram of clindamycin per milliliter 
(estimated).
---------------------------------------------------------------------------

    1 Defined such that 50 units hydrolyzes at least 20 
micromoles of a clindamycin phosphate authentic sample under the assay 
conditions described in this section.
---------------------------------------------------------------------------

    (3) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (4) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 24 milligrams of clindamycin per milliliter.
    (5) [Reserved]
    (6) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (7) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (8) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (9) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (10) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation method described in paragraph (b)(2) of 
that section, except dry the sample for 2 hours at 100 deg. C. and allow 
to equilibrate with the atmosphere for 1 hour.
[39 FR 19161, May 30, 1974, as amended at 46 FR 60568, Dec. 11, 1981; 50 
FR 19921, May 13, 1985]



Sec. 453.30  Lincomycin hydrochloride monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Lincomycin hydrochloride monohydrate is 
the monohydrated hydrochloride salt of lincomycin. It is freely soluble 
in water and soluble in acetone and dimethylformamide. It is so purified 
and dried that:
    (i) Its potency is not less than 790 micrograms of lincomycin per 
milligram.
    (ii) [Reserved]
    (iii) Its moisture content is not less than 3.0 percent and is not 
more than 6.0 percent.
    (iv) Its pH in an aqueous solution containing 100 milligrams per 
milliliter is not less than 3.0 and not more than 5.5.
    (v) Its specific rotation in an aqueous solution at 25 deg. C. is 
not less than +135 deg. and not more than +150 deg..
    (vi) It passes the infrared identity test.
    (vii) Its content of lincomycin B is not more than 5 percent.
    (viii) It passes the identity test if the elution pattern of the 
lincomycin sample compares quantitatively to that of the lincomycin 
working standard under identical conditions of gas liquid 
chromatography.
    (ix) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, specific rotation, infrared absorption spectrum, lincomycin B 
content, crystallinity, and identity.
    (ii) Samples of the batch: 10 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the gas liquid 
chromatography assay shall be conclusive.
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient sterile distilled 
water to obtain a stock solution of convenient concentration. Further 
dilute an aliquot of the stock solution with sterile distilled water to 
the reference concentration of 0.5 microgram of lincomycin per 
milliliter (estimated).
    (ii) Gas liquid chromatography assay. Proceed as directed in 
Sec. 436.306 of this chapter.
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (5) Specific rotation. Accurately weigh 500 milligrams of lincomycin 
hydrochloride monohydrate in a 25-milliliter, glass stoppered volumetric 
flask and fill to volume with distilled water. Proceed as directed in 
Sec. 436.210 of this

[[Page 953]]

chapter, using a 2.0-decimeter polarimeter tube and calculate the 
specific rotation on an anhydrous basis.
    (6) Infrared absorption spectrum. Proceed as directed in 
Sec. 436.211 of this chapter, using the sample preparation method 
described in paragraph (b)(2) of that section.
    (7) Lincomycin B content. Proceed as directed in Sec. 436.306 of 
this chapter.
    (8) Identity. Proceed as described in Sec. 436.306 of this chapter.
    (9) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19161, May 30, 1974, as amended at 46 FR 3839, Jan. 16, 1981; 50 
FR 19921, May 13, 1985]



Sec. 453.30a  Sterile lincomycin hydrochloride monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Lincomycin hydrochloride monohydrate is 
the monohydrated hydrochloride salt of lincomycin. It is freely soluble 
in water and soluble in acetone and dimethylformamide. It is so purified 
and dried that:
    (i) Its potency is not less than 790 micrograms of lincomycin per 
milligram.
    (ii) It is sterile.
    (iii) [Reserved]
    (iv) It is nonpyrogenic.
    (v) It contains no depressor substances.
    (vi) Its moisture content is not less than 3.0 percent and not more 
than 6.0 percent.
    (vii) Its pH in an aqueous solution containing 100 milligrams per 
milliliter is not less than 3.0 and not more than 5.5.
    (viii) Its specific rotation in an aqueous solution at 25 deg. C. is 
not less than +135 deg. and not more than +150 deg..
    (ix) It passes the infrared identity test.
    (x) Its content of lincomycin B is not more than 5 percent.
    (xi) It passes the identity test if the elution pattern of the 
lincomycin sample compares quantitatively to that of the lincomycin 
working standard under identical conditions of gas liquid 
chromatography.
    (xii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain.
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, depressor substances, moisture, pH, specific rotation, 
infrared absorption spectrum, lincomycin B content, identity, and 
crystallinity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 300 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the gas liquid 
chromatography assay shall be conclusive.
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient sterile distilled 
water to obtain a stock solution of convenient concentration. Further 
dilute an aliquot of the stock solution with sterile distilled water to 
the reference concentration of 0.5 microgram of lincomycin per 
milliliter (estimated).
    (ii) Gas liquid chromatography assay. Proceed as directed in 
Sec. 436.306 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) [Reserved]
    (4) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 0.5 milligram of lincomycin per milliliter.
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.

[[Page 954]]

    (8) Specific rotation. Accurately weigh 500 milligrams of lincomycin 
hydrochloride monohydrate in a 25 milliliter, glass-stoppered volumetric 
flask and fill to lincomycin B content, crystallinity, and volume with 
distilled water. Proceed as directed in Sec. 436.210, using a 2.0-
decimeter polarimeter tube and calculate the specific rotation on an 
anhydrous basis.
    (9) Infrared absorption spectrum. Proceed as directed in 
Sec. 436.211 of this chapter, using the sample preparation method 
described in paragraph (b)(2) of that section.
    (10) Lincomycin B content. Proceed as directed in Sec. 436.306 of 
this chapter.
    (11) Identity. Proceed as directed in Sec. 436.306 of this chapter.
    (12) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19161, May 30, 1974, as amended at 46 FR 3839, Jan. 16, 1981; 46 
FR 60568, Dec. 11, 1981; 50 FR 19921, May 13, 1985]



                      Subpart B--Oral Dosage Forms



Sec. 453.120  Clindamycin hydrochloride hydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clindamycin hydrochloride hydrate 
capsules are composed of clindamycin hydrochloride hydrate and one or 
more suitable and harmless diluents and lubricants. Each capsule 
contains clindamycin hydrochloride hydrate equivalent to 75, 150, or 300 
milligrams of clindamycin. Its content of clindamycin is satisfactory if 
it is not less than 90 percent and not more than 120 percent of the 
amount of clindamycin that it is represented to contain. The moisture 
content is not more than 7.0 percent. The clindamycin hydrochloride 
hydrate used conforms to the standards prescribed by Sec. 453.20(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The clindamycin hydrochloride hydrate used in making the batch 
for clindamycin content, microbiological activity, moisture, pH, 
crystallinity, and identity.
    (b) The batch for clindamycin content and moisture.
    (ii) Samples required:
    (a) The clindamycin hydrochloride hydrate used in making the batch: 
10 packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Clindamycin content (vapor phase 
chromatography). Proceed as directed in Sec. 436.302 of this chapter, 
except:
    (i) Preparation of clindamycin sample and working standard 
solutions. Accurately weigh a portion of the clindamycin working 
standard equivalent to about 45 milligrams of clindamycin and transfer 
to a 15-milliliter glass-stoppered centrifuge tube. Empty 20 capsules, 
collecting the contents quantitatively. Weigh the powder and determine 
the average capsule fill weight. Mix the powder and accurately weigh a 
portion containing the equivalent of about 45 milligrams of clindamycin 
into a second 15-milliliter glass-stoppered centrifuge tube. Add 3 
milliliters of 1 percent sodium carbonate solution and 3 milliliters of 
chloroform to each tube. Shake the solution vigorously and then 
centrifuge. Remove the top aqueous layer and add approximately 1 gram of 
anhydrous sodium sulfate to dry the chloroform layer. Place a 1-
milliliter aliquot of the chloroform solution into a 15-milliliter 
centrifuge tube, add 1 milliliter of internal standard and 0.6 
milliliter of acetic anhydride. Agitate the vials to insure complete 
mixing of the liquids.
    (ii) Calculations. Calculate the clindamycin content of the capsules 
as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.299

where:
Ru=Area of the clindamycin sample peak (at a retention time 
equal to that observed for the clindamycin standard)/Area of internal 
standard peak;
Rs=Area of the clindamycin standard peak/Area of internal 
standard peak;
Ws=Weight of clindamycin working standard in milligrams;
Wu=Sample weight in milligrams;

[[Page 955]]

f=Potency of clindamycin working standard in milligrams of clindamycin 
per milligram;
Wa=Average capsule fill weight in milligrams,

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19161, May 30, 1974, as amended at 50 FR 19921, May 13, 1985; 54 
FR 41824, Oct. 12, 1989; 54 FR 43384, Oct. 24, 1989]



Sec. 453.121  Clindamycin palmitate hydrochloride oral dosage forms.



Sec. 453.121a  Clindamycin palmitate hydrochloride for oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clindamycin palmitate hydrochloride for 
oral suspension is composed of clindamycin palmitate hydrochloride with 
one or more suitable and harmless diluents, buffer substances, 
colorings, and flavorings. When reconstituted as directed in the 
labeling, using the accompanying diluent when provided, each milliliter 
contains clindamycin palmitate hydrochloride equivalent to 15 milligrams 
of clindamycin. Its clindamycin content is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the amount of 
clindamycin that it is represented to contain. The moisture content is 
not more than 3.0 percent. When reconstituted as directed in the 
labeling, its pH is not less than 3.0 and not more than 5.0. The 
clindamycin palmitate hydrochloride used conforms to the standards 
prescribed by Sec. 453.21(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The clindamycin palmitate hydrochloride used in making the batch 
for clindamycin content, moisture, pH, and identity.
    (b) The batch for clindamycin content, moisture, and pH.
    (ii) Samples required:
    (a) The clindamycin palmitate hydrochloride used in making the 
batch: 10 packages, nine containing not less than 300 milligrams, and 
one containing not less than 2 grams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Clindamycin content. Proceed as 
directed in Sec. 436.303 of this chapter, except:
    (i) Preparation of clindamycin palmitate hydrochloride sample and 
working standard solutions. Accurately weigh about 130 milligrams of the 
clindamycin palmitate hydrochloride working standard and transfer to a 
25-milliliter volumetric flask. Add 5 milliliters of distilled water. 
Reconstitute the clindamycin palmitate hydrochloride for oral suspension 
as directed in the labeling, using the accompanying diluent when 
provided, and transfer exactly 5.0 milliliters to a 25-milliliter 
volumetric flask. Add exactly 5.0 milliliters of internal standard and 1 
milliliter of 30 percent sodium carbonate to each flask. Shake both 
flasks mechanically for 5 minutes. Transfer the contents of each flask 
to separate 15-milliliter glass-stoppered centrifuge tubes and 
centrifuge. Remove the top aqueous layer by suction and transfer exactly 
1.0 milliliter of the chloroform layer to separate glass-stoppered, 
conical, 15-milliliter centrifuge tubes. Add 1 milliliter of pyridine 
and 0.5 milliliter of acetic anhydride. Agitate the tubes to insure 
complete mixing of the liquids. Proceed as directed in Sec. 436.303(e) 
of this chapter.
    (ii) Calculations: Calculate the clindamycin content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.300
    
where:
Ru=Area of the sample peak (at a retention time equal to that 
observed for the clindamycin palmitate hydrochloride standard)/Area of 
internal standard peak;
Rs=Area of the clindamycin palmitate hydrochloride standard 
peak/Area of internal standard peak;
Ws=Weight of the clindamycin palmitate hydrochloride working 
standard in milligrams;
V=Volume of reconstituted sample in milliliters;
f=Milligrams of clindamycin activity per milligram of clindamycin 
palmitate hydrochloride working standard.


[[Page 956]]


    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
[39 FR 19161, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 453.121b  Clindamycin palmitate hydrochloride for oral solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clindamycin palmitate hydrochloride for 
oral solution is composed of clindamycin palmitate hydrochloride with 
one or more suitable and harmless diluents, buffer substances, 
colorings, flavorings, and preservatives. When reconstituted as directed 
in the labeling, each milliliter contains clindamycin palmitate 
hydrochloride equivalent to 15 milligrams of clindamycin. Its 
clindamycin content is satisfactory if it is not less than 90 percent 
and not more than 120 percent of the number of milligrams of clindamycin 
that it is represented to contain. The moisture content is not more than 
3.0 percent. When reconstituted as directed in the labeling, its pH is 
not less than 2.5 and not more than 5.0. The clindamycin palmitate 
hydrochloride used conforms to the standards prescribed by 
Sec. 453.21(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this subchapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The clindamycin palmitate hydrochloride used in making the batch 
for clindamycin content, moisture, pH, and identity.
    (b) The batch for clindamycin content, moisture, and pH.
    (ii) Samples required:
    (a) The clindamycin palmitate hydrochloride used in making the 
batch: 10 packages, nine containing not less than 300 milligrams, and 
one containing not less than 2 grams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Clindamycin content. Proceed as 
directed in Sec. 436.303 of this chapter, except:
    (i) Preparation of clindamycin palmitate hydrochloride sample and 
working standard solutions. Accurately weigh about 130 milligrams of the 
clindamycin palmitate hydrochloride working standard and transfer to a 
25-milliliter volumetric flask. Add 5 milliliters of distilled water. 
Reconstitute the clindamycin palmitate hydrochloride for oral solution 
as directed in the labeling and transfer exactly 5.0 milliliters to a 
25-milliliter volumetric flask. Add exactly 5.0 milliliters of internal 
standard and 1 milliliter of 30-percent sodium carbonate to each flask. 
Shake both flasks mechanically for 5 minutes. Transfer the contents of 
each flask to separate 15-milliliter glass-stoppered centrifuge tubes 
and centrifuge. Remove the top aqueous layer by suction and transfer 
exactly 1.0 milliliter of the chloroform layer to separate glass-
stoppered, conical, 15-milliliter centrifuge tubes. Add 1 milliliter of 
pyridine and 0.5 milliliter of acetic anhydride. Agitate the tubes to 
insure complete mixing of the liquids. Proceed as directed in 
Sec. 436.303(e) of this subchapter.
    (ii) Calculations. Calculate the clindamycin content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.301
    
where:
Ru=Area of the sample peak (at a retention time equal to that 
observed for the clindamycin palmitate hydrochloride standard);/Area of 
internal standard peak;
Rs=Area of the clindamycin palmitate hydrochloride standard 
peak;/Area of internal standard peak;
Ws=Weight of the clindamycin palmitate hydrochloride working 
standard in milligrams;
V=Volume of reconstituted sample in milliliters;
f=Milligrams of clindamycin activity per milligram of clindamycin 
palmitate hydrochloride working standard.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this 
subchapter.

[[Page 957]]

    (3) pH. Proceed as directed in Sec. 436.202 of this subchapter, 
using the drug reconstituted as directed in the labeling.
[39 FR 19161, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 453.130  Lincomycin hydrochloride oral dosage forms.



Sec. 453.130a  Lincomycin hydrochloride monohydrate capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Lincomycin hydrochloride monohydrate 
capsules are composed of lincomycin hydrochloride monohydrate and 
suitable diluents, enclosed in a gelatin capsule. Each capsule contains 
250 milligrams of lincomycin or 500 milligrams of lincomycin. The 
lincomycin content is satisfactory if it is not less than 90 percent and 
not more than 120 percent of the number of milligrams of lincomycin that 
it is represented to contain. Its moisture content is not more than 7.0 
percent. The lincomycin hydrochloride monohydrate used conforms to the 
standards prescribed by Sec. 453.30(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The lincomycin hydrochloride monohydrate used in making the 
batch for potency, moisture, pH, specific rotation, infrared absorption 
spectrum, and identity.
    (b) The batch for potency and moisture.
    (ii) Samples required:
    (a) The lincomycin hydrochloride monohydrate used in making the 
batch: 10 packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the gas liquid 
chromatography assay shall be conclusive.
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar with sufficient sterile distilled water to obtain a stock 
solution of convenient concentration. Blend for 3 to 5 minutes. Remove 
an aliquot of the stock solution and further dilute with sterile 
distilled water to the reference concentration of 0.5 microgram of 
lincomycin per milliliter (estimated).
    (ii) Gas liquid chromatography assay. Proceed as directed in 
Sec. 436.306 of this chapter, except prepare the sample for assay as 
follows: Place the contents of 5 capsules in a 100-milliliter volumetric 
flask and add about 60 milliliters of methanol. Place on a steam bath 
and allow to boil gently for 5 minutes. Remove from the steam bath, add 
more methanol, and adjust to mark after cooling to ambient temperature. 
Dilute an aliquot equivalent to 50 milligrams of lincomycin to 25 
milliliters with methanol. Transfer 2 milliliters to a centrifuge tube 
and evaporate to dryness on a steam bath with a stream of dry air. 
Dissolve the residue in 1 milliliter of dry pyridine. Calculate the 
lincomycin content of the capsules as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.302

where:
Ru=Area of lincomycin sample peak/Area of internal standard;
Rs=Area of lincomycin standard peak/Area of internal 
standard;
Ws=Weight of lincomycin working standard in milligrams;
d=Dilution factor;
f=Potency of lincomycin working standard in milligrams of lincomycin per 
milligram;
N=Number of capsules used.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
[39 FR 19161, May 30, 1974, as amended at 46 FR 3839, Jan. 16, 1981; 50 
FR 19921, May 13, 1985]



Sec. 453.130b  Lincomycin hydrochloride syrup.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Lincomycin hydrochloride syrup is a syrup 
containing lincomycin hydrochloride monohydrate, one or

[[Page 958]]

more suitable preservatives, flavorings, sweetening agents, colorings, 
and purified water. Each milliliter contains lincomycin hydrochloride 
equivalent to either 25 milligrams or 50 milligrams of lincomycin. Its 
lincomycin content is satisfactory if it is not less than 90 percent and 
not more than 120 percent of the number of milligrams of lincomycin that 
it is represented to contain. The pH is not less than 3 and not more 
than 5.5. The lincomycin hydrochloride monohydrate used conforms to the 
standards prescribed by Sec. 453.30(a)(1) (i), (iv), (v), (vi), (vii), 
(viii), and (ix).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The lincomycin hydrochloride monohydrate used in making the 
batch for potency, pH, specific rotation, infrared absorption spectrum, 
lincomycin B content, crystallinity, and identity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The lincomycin hydrochloride monohydrate used in making the 
batch: 10 packages, each containing approximately 300 milligrams.
    (b) The batch: A minimum of five immediate containers.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the gas liquid 
chromatography assay shall be conclusive.
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Remove an accurately measured representative sample with a suitable 
hypodermic needle and syringe. Place into a high-speed glass blender jar 
with sufficient sterile distilled water to give a total volume of 500 
milliliters. Blend for 3 to 5 minutes. Further dilute an aliquot with 
sterile distilled water to the reference concentration of 0.5 microgram 
of lincomycin per milliliter (estimated).
    (ii) Gas liquid chromatography assay. Proceed as directed in 
Sec. 436.306 of this chapter, except prepare the sample for assay by 
either of the following methods:
    (a) Place an aliquot of syrup, containing the equivalent of 250 
milligrams of lincomycin into a 50-milliliter volumetric flask and add 
30 milliliters of absolute ethanol. Place on a steam bath and boil 
gently for 5 minutes. Remove from the steam bath and cool. Add ethanol 
to prior volume level and let stand overnight. Adjust to mark, shake 
well, and transfer a 5-milliliter aliquot into a 25-milliliter 
volumetric flask and make to mark with methanol. Place 4 milliliters of 
this solution in a 15-milliliter centrifuge tube and evaporate to 
dryness on a steam bath with a stream of dry air. Dissolve the residue 
in 1 milliliter of dry pyridine. Calculate the lincomycin content as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.303

where:
Ru=Area of lincomycin sample peak/Area of internal standard;
Rs=Area of lincomycin standard peak/Area of internal 
standard;
Ws=Weight of lincomycin working standard in milligrams;
d=Dilution factor;
f=Potency of lincomycin working standard in milligrams of lincomycin per 
milligram;
M=Milliliters of syrup used.

    (b) Treat the lincomycin working standard and sample in a similar 
manner, except lyophilize an aliquot of the sample containing the 
equivalent of 50 milligrams of lincomycin. To approximately 50 
milligrams of the standard, accurately weighed, and to the dried residue 
of the sample, add 5 milliliters of dry pyridine which contains 10 
milligrams of tetraphenylcyclopentadienone per milliliter. Warm on a hot 
plate for 5 minutes to attain complete solution. Remove from the hot 
plate and add 5 milliliters of hexamethyldisilazane and 2 milliliters of 
trimethylchlorosilane. Shake mechanically for 60 minutes, then 
centrifuge for 15 minutes. Inject 2 microliters of the supernate into 
the chromatograph. Calculate the lincomycin content as follows:

[[Page 959]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.304


where:
Ru=Area of lincomycin sample peak/Area of internal standard;
Rs=Area of lincomycin standard peak/Area of internal 
standard;
Ws=Weight of lincomycin working standard in milligrams;
f=Potency of lincomycin working standard in milligrams of lincomycin per 
milligram;
M=Milliliters of syrup used.

    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
[39 FR 19161, May 30, 1974, as amended at 46 FR 3840, Jan. 16, 1981; 50 
FR 19921, May 13, 1985]



                   Subpart C--Injectable Dosage Forms



Sec. 453.222  Clindamycin phosphate injection.

    (a)(1) Standards of identity, strength, quality, and purity. 
Clindamycin phosphate injection is an aqueous solution of clindamycin 
phosphate with one or more suitable and harmless preservatives, 
sequestering agents, or tonicity agents. It may be frozen. Its 
clindamycin phosphate content is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
clindamycin that it is represented to contain. It is sterile. It is 
nonpyrogenic. It contains no depressor substances. Its pH is not less 
than 5.5 and not more than 7. The clindamycin phosphate used conforms to 
the standards prescribed by Sec. 453.22a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The clindamycin phosphate used in making the batch for 
clindamycin content, microbiological activity, moisture, pH, 
crystallinity, and identity.
    (b) The batch for clindamycin content, sterility, pyrogens, 
depressor substances, and pH.
    (ii) Samples required:
    (a) The clindamycin phosphate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Clindamycin content. Use any of 
the following methods. However, the results obtained from the high 
performance liquid chromatographic assay shall be conclusive.
    (i) Vapor phase chromatography. Proceed as directed in Sec. 436.304 
of this chapter, except prepare the sample for assay as follows: Shake 
the sample and dilute a portion with pH 9.0 borate buffer to obtain a 
solution containing the equivalent of approximately 0.4 milligrams of 
clindamycin per milliliter. Place 25 milliliters of this solution into a 
50-milliliter stoppered centrifuge tube. Add 10 milliliters of 
chloroform. Shake vigorously for 15 minutes and centrifuge. There should 
be no emulsion present after centrifugation. Transfer 20 milliliters of 
the aqueous phase from the tube into a 35-milliliter stoppered 
centrifuge tube. Add to the tube a weighed amount of intestinal alkaline 
phosphatase equivalent to 50 units of activity \1\ and allow to stand 
until the phosphatase has dissolved completely. Place the centrifuge 
tube into a water bath at 37  deg.Cplus-minus2  deg.C for 2.5 
hours. After the 2.5-hours hydrolysis, allow the solution to cool.
---------------------------------------------------------------------------

    \1\ Defined such that 50 units hydrolyzes at least 20 micromoles of 
a clindamycin phosphate authentic sample under the assay conditions 
described in Sec. 436.304 of this chapter.
---------------------------------------------------------------------------

    (ii) High performance liquid chromatographic assay. Proceed as 
directed in Sec. 436.216 of this chapter, using ambient temperature, an 
ultraviolet detection system operating at a wavelength of 210 
nonometers, a 25-centimeter long  x  4.6 millimeter ID column packed 
with microparticulate (5 to 10 micrometers in diameter) reversed phase 
octysilane hydrocarbon bonded

[[Page 960]]

silica packing material, a flow rate of about 1.0 milliliter per minute, 
and a known injection volume of between 10 and 20 microliters. The 
retention time of clindamycin phosphate, and clindamycin are 
approximately 6 and 9 minutes, respectively. Reagents, working standard 
and sample solutions, resolution test solution, system suitability 
requirements, and calculations are as follows:
    (a) Reagents--(1) 0.1M Potassium phosphate monobasic buffer. 
Dissolve 13.61 grams of potassium phosphate monobasic in 775 milliliters 
of water. Adjust the pH to 2.5 with phosphoric acid. Further dilute with 
water to a volume of 1,000 milliliters.
    (2) Mobile phase. Mix 225 milliliters of acetonitrile and 775 
milliliters of 0.1M potassium phosphate, pH 2.5 buffer (225:775). Filter 
through a suitable filter capable of removing particulate matter greater 
than 0.5 micron in diameter. Degas the mobile phase just prior to its 
introduction into the chromatograph.
    (b) Preparation of working standard, sample, and resolution test 
solutions--(1) Working standard solution. Dissolve an accurately weighed 
portion of the clindamycin phosphate working standard with sufficient 
mobile phase (prepared as directed in paragraph (b)(1)(ii)(a)(2) of this 
section) to obtain a solution containing 200 micrograms of clindamycin 
activity per milliliter.
    (2) Sample solution. Using a suitable hypodermic needle and syringe, 
remove an accurately measured representative portion from each container 
and dilute with sufficient mobile phase (prepared as directed in 
paragraph (b)(1)(ii)(a)(2) of this section) to obtain a solution 
containing 200 micrograms of clindamycin per milliliter (estimated).
    (3) Resolution test solution. Place 15 milligrams each of 
clindamycin phosphate, and clindamycin hydrochloride in a 25-milliliter 
volumetric flask and dissolve and dilute with mobile phase and mix well. 
Use this solution to determine the resolution factor.
    (c) System suitability requirements--(1) Asymmetry factor. Calculate 
the asymmetry factor (As), measured at a point 5 percent of 
the peak height from the baseline as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.305

where:
a = Horizontal distance from point of ascent to point of maximum peak 
          height; and
b = Horizontal distance from the point of maximum peak height to point 
          of descent.

    The asymmetry factor (As) is satisfactory if it is not 
more than 1.3.

    (2) Efficiency of the column. From the number of theoretical plates 
(n) calculated as described in Sec. 436.216(c)(2) of this chapter 
calculate the reduced plate height (hr) as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.306

where:
L = Length of the column in centimeters;
n = Number of theoretical plates; and
dp = Average diameter of the particles in the analytical 
          column packing in micrometers.

    The absolute efficiency (hr) is satisfactory if it is not 
more than 15.

    (3) Resolution factor. The resolution factor (R) between the peak 
for clindamycin phosphate and the peak for clindamycin hydrochloride in 
the chromatogram of the resolution test solution is satisfactory if it 
is not less than 6.0.
    (4) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent) of 5 replicate 
injections of the working standard solution (prepared as directed in 
paragraph (b)(1)(ii)(b)(1) of this section is satisfactory if it is not 
more than 2.5 percent.
    If the system suitability parameters have been met, then proceed as 
described in Sec. 436.216(b) of this chapter.
    (d) Calculations. Calculate the clindamycin content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.307
    
where:
Au = Area of the clindamycin phosphate peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
As = Area of the clindamycin phosphate peak in the 
          chromatogram of the clindamycin

[[Page 961]]

          phosphate working standard;
Ps = Clindamycin activity in the clindamycin phosphate 
          working standard solution in micrograms per milliliter; and
d = Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing the equivalent of 24 milligrams of 
clindamycin per milliliter.
    (4) [Reserved]
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted drug.
[39 FR 19161, May 30, 1974, as amended at 46 FR 60568, Dec. 11, 1981; 50 
FR 19921, May 13, 1985; 54 FR 43289, Oct. 24, 1989; 55 FR 5842, Feb. 20, 
1990]



Sec. 453.230  Lincomycin hydrochloride injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Lincomycin hydrochloride injection is an 
aqueous solution of lincomycin hydrochloride monohydrate containing 
benzyl alcohol as a preservative. Each immediate container contains 
either 1, 2, or 10 milliliters of a solution containing, in each 
milliliter, 300 milligrams of lincomycin, and 9 milligrams of benzyl 
alcohol. The lincomycin content is satisfactory if it is not less than 
90 percent and not more than 120 percent of the number of milligrams of 
lincomycin that it is represented to contain. It is sterile. It is 
nonpyrogenic. It contains no depressor substances. Its pH is not less 
than 3.0 and not more than 5.5. The lincomycin hydrochloride monohydrate 
used conforms to the standards prescribed by Sec. 453.30a(a)(1) (i), 
(vi), (vii), (viii), (ix), (x), and (xi).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. If each immediate container 
contains only 1 milliliter of the drug, the labeling shall include the 
statement ``For pediatric use''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The lincomycin hydrochloride monohydrate used in making the 
batch for potency, moisture, pH, specific rotation, infrared absorption 
spectrum, lincomycin B content, identity, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, depressor 
substances, and pH.
    (ii) Samples required:
    (a) The lincomycin hydrochloride monohydrate used in making the 
batch: 10 packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the gas liquid 
chromatography assay shall be conclusive.
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Using a suitable hypodermic needle and syringe, remove all of the 
withdrawable contents if it is represented as a single-dose container; 
or, if the labeling specifies the amount of potency in a given volume of 
the resultant preparation, remove an accurately measured representative 
portion from each container. Place the portion, thus obtained, into a 
suitably-sized volumetric flask and dilute to volume with sterile 
distilled water. Remove an aliquot and further dilute with sterile 
distilled water to the reference concentration of 0.5 microgram of 
lincomycin per milliliter (estimated).
    (ii) Gas liquid chromatography assay. Proceed as directed in 
Sec. 436.306 of this chapter, except prepare the sample for assay as 
follows: Dilute the equivalent of 300 milligrams of lincomycin to 50 
milliliters with methanol and shake. Transfer a 3-milliliter aliquot to 
a 10-milliliter volumetric flask and make to

[[Page 962]]

mark with methanol. Place a 2-milliliter aliquot into a 15-milliliter 
centrifuge tube and evaporate to dryness on a steam bath with a stream 
of dry air. Dissolve the residue in 1 milliliter of dry pyridine. 
Calculate the lincomycin content as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.308

where:
[GRAPHIC] [TIFF OMITTED] TR01JA93.309

Ws=Weight of lincomycin working standard in milligrams;
d=Dilution factor;
f=Potency of lincomycin working standard in milligrams of lincomycin per 
milligram

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) [Reserved]
    (4) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 0.5 milligram of lincomycin per milliliter.
    (5) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[39 FR 19161, May 30, 1974, as amended at 46 FR 3841, Jan. 16, 1981; 46 
FR 60568, Dec. 11, 1981; 50 FR 19921, May 13, 1985]



                        Subparts D-E  [Reserved]



                  Subpart F--Dermatologic Dosage Forms



Sec. 453.522  Clindamycin phosphate dermatologic dosage forms.



Sec. 453.522a  Clindamycin phosphate topical solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clindamycin phosphate is a solution of 
clindamycin phosphate in a suitable and harmless vehicle. Each 
milliliter contains 10 milligrams of clindamycin activity. Its 
clindamycin content is satisfactory if it is not less than 90 percent 
and not more than 110 percent of the number of milligrams of clindamycin 
that it is represented to contain. Its pH is not less than 4.0 and not 
more than 7.0. The clindamycin phosphate used conforms to the standards 
prescribed by Sec. 453.22(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The clindamycin phosphate used in making the batch for 
clindamycin content, microbiological activity, moisture, pH, 
crystallinity, and identity.
    (b) The batch for clindamycin content and pH.
    (ii) Samples required:
    (a) The clindamycin phosphate used in making the batch: 6 packages, 
each containing approximately 300 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Clindamycin content (vapor phase 
chromatography). Proceed as directed in Sec. 436.304 of this chapter, 
except prepare the sample for assay and calculate the clindamycin 
content as follows:
    (i) Preparation of the sample. Accurately transfer a volume of 
sample equivalent to approximately 20 milligrams of clindamycin activity 
to a 50-milliliter volumetric flask. Evaporate the sample to near 
dryness under a stream of nitrogen. Dilute to 50 milliliters with pH 9.0 
borate buffer and mix well. Place 25.0 milliliters of this solution into 
a 50-milliliter stoppered centrifuge tube. Add 10 milliliters of 
chloroform. Shake vigorously for 15 minutes and centrifuge to obtain 
adequate phase separation of the chloroform and aqueous phase. Transfer 
20 milliliters of the aqueous phase from the tube into a 35-milliliter 
stoppered centrifuge tube. Add to the tube a weighed

[[Page 963]]

amount of intestinal alkaline phosphatase equivalent to 50 units of 
activity \1\ and allow to stand until the phosphatase has dissolved 
completely. Place the centrifuge tube into a water bath at 37 deg. C 
plus-minus 2 deg. C for 2.5 hours. After the 2.5-hours 
hydrolysis, allow the solution to cool.
---------------------------------------------------------------------------

    \1\ Defined such that 50 units hydrolyzes at least 20 micromoles of 
a clindamycin phosphate authentic sample under the assay conditions 
described in Sec. 436.304 of this chapter.
---------------------------------------------------------------------------

    (ii) Calculations. Calculate the clindamycin content as follows:

Clindamycin content per milliliter=(Ru  x W  x s  
          x d  x f)/(Rs  x V)

where:

    Ru=Area of clindamycin sample peak/Area of internal 
standard;
    Rs=Area of clindamycin standard peak/Area of internal 
standard;
    Ws=Weight of clindamycin working standard in milligrams;
    d=Dilution factor;
    f=Potency of clindamycin working standard in milligrams of 
clindamycin per milligram;
    V=Volume of sample in milliliters.

    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted drug.
[46 FR 2997, Jan. 13, 1981. Redesignated at 54 FR 38224, Sept. 15, 1989]



Sec. 453.522b  Clindamycin phosphate gel.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clindamycin phosphate gel contains 
clindamycin phosphate in a suitable and harmless vehicle. Each gram 
contains clindamycin phosphate equivalent to 10 milligrams of 
clindamycin activity. Its clindamycin content is satisfactory if it is 
not less than 90 percent and not more than 110 percent of the number of 
milligrams of clindamycin that it is represented to contain. Its pH is 
not less than 4.5 and not more than 6.5. It passes the identity test. 
The clindamycin phosphate used conforms to the standards prescribed by 
Sec. 453.22(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification: samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The clindamycin phosphate used in making the batch for 
clindamycin content, microbiological activity, moisture, pH, 
crystallinity, and identity.
    (B) The batch for clindamycin content, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The clindamycin phosphate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (B) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Clindamycin content (High 
performance liquid chromatographic assay). Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 210 nanometers, a 25-
centimeter long x 4.6-millimeter ID column packed with microparticulate 
(5 to 10 micrometers in diameter) reversed phase octysilane hydrocarbon 
bonded silica packing material, a flow rate of about 1.0 milliliter per 
miunute, and a known injection volume of between 10 and 20 microliters. 
The retention time of clindamycin phosphate, and clindamycin are 
approximately 6 and 9 minutes, respectively. Reagents, working standards 
and sample solutions, resolution test solution, system suitability 
requirements, and calculations are as follows:
    (i) Reagents--(A) 0.1M Potasium phosphate monobasic buffer. Dissolve 
13.61 grams of potassium phosphate monobasic in 775 milliliters of 
water. Adjust the pH to 2.5 with phosphoric acid. Further dilute with 
water to a volume of 1,000 milliliters.
    (B) Mobile phase. Mix 225 milliliters of acetonitrile and 775 
milliliters of 0.1M potassium phosphate, pH 2.5 buffer (225:775). Filter 
through a suitable filter capable of removing particulate matter greater 
than 0.5 micron in diameter. Degas the mobile phase just prior to its 
introduction into the chromatograph.
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(A)

[[Page 964]]

Working standard solution. Dissolve an accurately weighed portion of the 
clindamycin phosphate working standard with sufficient mobile phase 
(prepared as directed in paragraph (b)(1)(i)(B) of this section) to 
obtain a solution containing 200 micrograms of clindamycin activity per 
milliliter.
    (B) Sample solution. Accurately weigh and transfer approximately 2.0 
grams of the sample into a 100-milliliter volumetric flask. Dilute to 
volume with sufficient mobile phase (prepared as directed in paragraph 
(b)(1)(i)(B) of this section) and shake vigorously for 30 minutes. 
Centrifuge a portion of the solution and if necessary filter a few 
milliliters of the centrifuged solution through a 2-micron millipore 
filter, type BS.
    (C) Resolution test solution. Place 15 milligrams each of 
clindamycin phosphate and clindamycin hydrochloride in a 25-milliliter 
volumetric flask and dissolve and dilute to volume with mobile phase and 
mix well. Use this solution to determine the resolution factor.
    (iii) System suitability requirements--(A) Asymmetry factor. 
Calculate the asymmetry factor (As), measured at a point 5 
percent of the peak height from the baseline as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.310

where:
a = Horizontal distance from point of ascent to point of maximum peak 
          height; and
b = Horizontal distance from the point of maximum peak height to point 
          of descent.

The asymmetry factor (As) is satisfactory if it is not more 
          than 1.3.

    (B) Efficiency of the column. From the number of theoretical plates 
(n) calculated as described in Sec. 436.216(c)(2) of this chapter 
calculate the reduced plate height (hr) as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.311

where:
L = Length of the column in centimeters;
n = Number of theoretical plates; and
dp = Average diameter of the particles in the analytical 
          column packing in micrometers.
    The absolute efficiency (hr) is satisfactory if it is not 
more than 15.

    (C) Resolution factor. The resolution factor (R) between the peak 
for clindamycin phosphate and the peak for clindamycin (hydrochloride) 
in the chromatogram of the resolution test solution is satisfactory if 
it is not less than 6.0.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent) of 5 replicate 
injections of the working standard solution (prepared as directed in 
paragraph (b)(1)(ii)(A) of this section is satisfactory if it is not 
more than 2.5 percent. If the system suitability parameters have been 
met, then proceed as described in Sec. 436.216(b) of this chapter.
    (iv) Calculations. Calculate the clindamycin content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.312
    
where:
Au = Area of the clindamycin phosphate peak in the 
          chromatogram of the sample (at a retention time equal to that 
          observed for the standard);
As = Area of the clindamycin phosphate peak in the 
          chromatogram of the clindamycin phosphate working standard;
Ps = Clindamycin activity in the clindamycin phosphate 
          working standard solution in micrograms per milliliter; and
d = Dilution factor of the sample.

    (A) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted gel.
    (B) Identity. The high-performance liquid chromatogram of the sample 
determined in paragraph (b)(1) of this section compares qualitatively to 
that of the clindamycin phosphate working standard.
[54 FR 38224, Sept. 15, 1989]



Sec. 453.522c  Clindamycin phosphate lotion.

    (a) Requirements for certification--(1) Standards for identity, 
strength, quality, and purity. Clindamycin phosphate lotion contains 
clindamycin phosphate in a suitable and harmless lotion vehicle, with 
one or more suitable and harmless emollients, buffers, and dispersants. 
Each milliliter contains clindamycin phosphate equivalent to 10 
milligrams

[[Page 965]]

of clindamycin. Its clindamycin content is satisfactory if it is not 
less than 90 percent and not more than 110 percent of the number of 
milligrams of clindamycin that it is represented to contain. Its pH is 
not less than 4.5 and not more than 6.5. It passes the identity test. 
The clindamycin phosphate used conforms to the standards prescribed by 
Sec. 453.22(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The clindamycin phosphate used in making the batch for 
clindamycin content, microbiological activity, moisture, pH, 
crystallinity, and identity.
    (B) The batch for clindamycin content, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The clindamycin phosphate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (B) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Clindamycin content (high 
performance liquid chromatographic assay). Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an untraviolet 
detection system operating at a wavelength of 210 nanometers, a 25-
centimeter long x 4.6 millimeter ID column packed with microparticulate 
(5 to 10 micrometers in diameter) reversed phase octysilane hydrocarbon 
bonded silica packing material, a flow rate of about 1.08 milliliter per 
minute, and a known injection volume of between 10 and 20 microliters. 
The retention time of clindamycin phosphate and clindamycin are 
approximately 6 and 9 minutes, respectively. Reagents, working standard 
and sample solutions, resolution test solution, system suitability 
requirements, and calculations are as follows:
    (i) Reagents--(A) O.1M Potassium phosphate monobasic buffer. 
Dissolve 13.61 grams of potassium phosphate monobasic in 775 milliliters 
of water. Adjust the pH to 2.5 with phosphoric acid. Further dilute with 
water to a volume of 1,000 milliliters.
    (B) Mobile phase. Mix 225 milliliters of acetonitrile and 775 
milliliters of 0.1M potassium phosphate, pH 2.5 buffer (225:775). Filter 
through a suitable filter capable of removing particulate matter greater 
than 0.5 micron in diameter. Degas the mobile phase just prior to its 
introduction into the chromatograph.
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(A) Working standard solution. Dissolve an accurately 
weighted portion of the clindamycin phosphate working standard with 
sufficient mobile phase (prepared as directed in paragraph (b)(1)(i)(B) 
of this section) to obtain a solution containing 200 micrograms of 
clindamycin activity per milliliter.
    (B) Sample solution. Using a suitable hypodermic needle and syringe, 
remove an accurately measured representative portion from each container 
and dilute with sufficient mobile phase (prepared as directed in 
paragraph (b)(1)(i)(B) of this section) to obtain a solution containing 
200 micrograms of clindamycin per milliliter (estimated).
    (C) Resolution test solution. Dissolve 30 milligrams of clindamycin 
phosphate in 25 milliliters of mobile phase. Dissolve 30 milligrams of 
clindamycin hydrochloride in 25 milliliters of mobile phase. Combine 
both solutions in a 50-milliliter volumetric flask and shake or use a 
vortex shaker to assure mixture of both solutions. Use this solution to 
determine the resolution factor.
    (iii) System suitability requirements--(A) Asymmetry factor. 
Calculate the asymmetry factor (As), measured at a point 5 
percent of the peak height from the base line as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.313

where:

a=Horizontal distance from point of ascent to point of maximum peak 
          height; and
b=Horizontal distance from the point of maximum peak height to point of 
          descent.
The asymmetry factor (As) is satisfactory if it is not more 
          than 1.3.


[[Page 966]]


    (B) Efficiency of the column. From the number of theoretical plates 
(n) calculated as described in Sec. 436.216(c)(2) of this chapter, 
calculate the reduced plate height (hr) as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.314

where:

L=Length of the column in centimeters;
n=Number of theoretical plates; and
dp=Average diameter of the particles in the analytical column 
          packing in micrometers.

    The absolute efficiency (hr) is satisfactory if it is not 
more than 15.

    (C) Resolution factor. The resolution factor (R) between the peak 
for clindamycin phosphate and the peak for clindamycin (hydrochloride) 
in the chromatogram of the resolution test solution is satisfactory if 
it is not less than 6.0.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SRin percent) of 5 replicate 
injections of the working standard solution (prepared as directed in 
paragraph (b)(1)(ii)(A) of this section is satisfactory if it is not 
more than 2.5 percent.
    If the system suitability parameters have been met, then proceed as 
described in Sec. 436.216(b) of this chapter.
    (iv) Calculations. Calculate the clindamycin content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.315
    
where:

Au=Area of the clindamycin phosphate peak in the chromatogram 
          of the sample (at a retention time equal to that observed for 
          the standard);
As=Area of the clindamycin phosphate peak in the chromatogram 
          of the clindamycin phosphate working standard;
Ps=Clindamycin activity in the clindamycin phosphate working 
          standard solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted lotion.
    (3) Identity. The high-performance liquid chromatogram of the sample 
determined in paragraph (b)(1) of this section compares qualitatively to 
that of the clindamycin phosphate working standard.
[54 FR 40655, Oct. 3, 1989]



Sec. 453.522d  Clindamycin phosphate vaginal cream.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clindamycin phosphate vaginal cream 
contains clindamycin phosphate in a suitable and harmless cream vehicle. 
Each gram contains clindamycin phosphate equivalent to 20 milligrams of 
clindamycin activity. Its clindamycin content is satisfactory if it is 
not less than 90 percent and not more than 110 percent of the number of 
milligrams of clindamycin that it is represented to contain. Its pH is 
not less than 3.0 and not more than 6.0. It passes the identity test. 
The clindamycin phosphate used conforms to the standards prescribed by 
Sec. 453.22(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The clindamycin phosphate used in making the batch for 
clindamycin content, microbiological activity, moisture, pH, 
crystallinity, and identity.
    (B) The batch for clindamycin content, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The clindamycin phosphate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (B) The batch: a minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Clindamycin content (high 
performance liquid chromatography assay). Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 210 nanometers, a 25-
centimeter long x 4.6 millimeter ID column packed with microparticulate 
(5 to 10 micrometers in diameter) reverse

[[Page 967]]

phase octylsilane hydrocarbon bonded silica packing material, a flow 
rate of 1.0 milliliter per minute, and a known injection volume of 20 
microliters. The retention time of clindamycin phosphate, and 
clindamycin are approximately 6 and 9 minutes, respectively. Reagents, 
working standards and sample solutions, resolution test solution, system 
suitability requirements, and calculations are as follows:
    (i) Reagents--(A) 0.1M Potassium phosphate monobasic buffer. 
Dissolve 13.61 grams of potassium phosphate monobasic in 775 milliliters 
of water. Adjust the pH to 2.5 with phosphoric acid. Further dilute with 
water to a volume of 1,000 milliliters.
    (B) Mobile phase. Mix 225 milliliters of acetonitrile and 775 
milliliters of 0.1M potassium phosphate, pH 2.5 buffer (225:775). Filter 
through a suitable filter capable of removing particulate matter greater 
than 0.5 micron in diameter. Degas the mobile phase just prior to its 
introduction into the chromatograph.
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(A) Working standard solution. Dissolve an accurately weighed 
portion of the clindamycin phosphate working standard in sufficient 
mobile phase (prepared as directed in paragraph (b)(1)(i)(B) of this 
section) to obtain a solution containing 200 micrograms of clindamycin 
activity per milliliter.
    (B) Sample solutions. Accurately weigh and transfer approximately 
1.0 gram of the sample into a 125-milliliter Erlenmeyer flask. Add 100.0 
milliliters of mobile phase (prepared as directed in paragraph 
(b)(1)(i)(B) of this section), accurately measured, and 8 to 10 glass 
beads (4 to 5 millimeters). Close the flask securely using a plastic 
stopper and shake vigorously by mechanical means for 1 hour at 50 
deg.C. Cool in an ice bath for approximately 20 minutes. Centrifuge a 
portion of the mixture. Use the lower cloudy solution for 
chromatographic analysis. Filter a few milliliters of the centrifuged 
solution through an appropriate 2 micron filter.
    (C) Resolution test solution. Place 15 milligrams each of 
clindamycin phosphate and clindamycin hydrochloride in a 25-milliliter 
volumetric flask and dissolve and dilute to volume with mobile phase and 
mix well. Use this solution to determine the resolution factor.
    (iii) System suitability requirements--(A) Asymmetry factor. 
Calculate the asymmetry factor (AS), measured at a point 5 
percent of the peak height from the baseline as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.410

where:

a = Horizontal distance from point of ascent to point of maximum peak 
          height; and
b = Horizontal distance from point of maximum peak height to point of 
          descent.
The asymmetry factor (As) is satisfactory if it is not less 
          than 1.0 and not more than 1.3.

    (B) Efficiency of the column. From the number of theoretical plates 
(n) calculated as described in Sec. 436.216(c)(2) of this chapter, 
calculate the reduced plate height (hr) as follows;
[GRAPHIC] [TIFF OMITTED] TR01JA93.314

where:

L = Length of the column in centimeters;
n = Number of theoretical plates; and
dp = Average diameter of the particles in the analytical 
          column packing in micrometers.
    The absolute efficiency (hr) is satisfactory if it is not 
more than 15.

    (C) Resolution factor. The resolution factor (R) between the peak 
for clindamycin phosphate and the peak for clindamycin (hydrochloride) 
in the chromatogram of the resolution test solution is satisfactory if 
it is not less than 6.0.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SRin percent) of 5 replicate 
injections of the working standard solution is satisfactory if it is not 
more than 2.5 percent. If the system suitability parameters have been 
met, then proceed as described in Sec. 436.216(b) of this chapter.
    (iv) Calculation. Calculate the clindamycin content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.312
    
where:

[[Page 968]]

    Au = Area of the clindamycin phosphate peak in the 
chromatogram of the sample (at a retention time equal to that observed 
for the standard);
    As = Area of the clindamycin phosphate peak in the 
chromatogram of the clindamycin phosphate working standard;
    Ps = Clindamycin activity in the clindamycin phosphate 
working standard solution in micrograms per milliliter; and
    d = Dilution factor of the sample.

    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted cream.
    (3) Identity. The high-pressure liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the clindamycin phosphate working standard.
[60 FR 49508, Sept. 26, 1995]



PART 455--CERTAIN OTHER ANTIBIOTIC DRUGS--Table of Contents




                          Subpart A--Bulk Drugs

Sec.
455.4  Aztreonam.
455.4a  Sterile aztreonam.
455.10  Chloramphenicol.
455.10a  Sterile chloramphenicol.
455.11  Chloramphenicol palmitate.
455.12a  Sterile chloramphenicol sodium succinate.
455.15  Clavulanate potassium.
455.15a  Sterile clavulanate potassium.
455.20  Cycloserine.
455.40  Mupirocin.
455.50  Calcium novobiocin.
455.51  Sodium novobiocin.
455.51a  Sterile sodium novobiocin.
455.70  Rifampin.
455.80a  Sterile spectinomycin hydrochloride.
455.82a  Sterile sulbactam sodium.
455.85  Vancomycin hydrochloride.
455.85a  Sterile vancomycin hydrochloride.
455.86  Vancomycin.
455.88  Rifabutin.
455.90a  Sterile vidarabine monohydrate.

                      Subpart B--Oral Dosage Forms

455.110  Chloramphenicol capsules.
455.111  Chloramphenicol palmitate oral suspension.
455.120  Cycloserine capsules.
455.150  Calcium novobiocin oral suspension.
455.151  Sodium novobiocin oral dosage forms.
455.151a  Sodium novobiocin tablets.
455.151b  Sodium novobiocin capsules.
455.170  Rifampin oral dosage forms.
455.170a  Rifampin capsules.
455.170b  Rifampin-isoniazid capsules.
455.185  Vancomycin hydrochloride oral dosage forms.
455.185a  Vancomycin hydrochloride for oral solution.
455.185b  Vancomycin hydrochloride capsules.
455.188  Rifabutin capsules.

                   Subpart C--Injectable Dosage Forms

455.204  Aztreonam injectable dosage forms.
455.204a  Aztreonam for injection.
455.204b  Aztreonam injection.
455.210  Chloramphenicol injection.
455.212  Sterile chloramphenicol sodium succinate.
455.230  Moxalactam disodium for injection.
455.251  Sodium novobiocin for injection.
455.270  Rifampin for injection.
455.280a  Sterile spectinomycin hydrochloride.
455.285  Vancomycin hydrochloride injectable dosage forms.
455.285a  Sterile vancomycin hydrochloride.
455.285b  Vancomycin hydrochloride for injection.
455.285c  Vancomycin hydrochloride injection.
455.290  Vidarabine monohydrate for infusion.

                   Subpart D--Ophthalmic Dosage Forms

455.310  Chloramphenicol ophthalmic dosage forms.
455.310a  Chloramphenicol ophthalmic solution.
455.310b  Chloramphenicol for ophthalmic solution.
455.310c  Chloramphenicol ointment (chloramphenicol cream).
455.310d  Chloramphenicol-polymyxin ointment.
455.310e  Chloramphenicol-hydrocortisone acetate for ophthalmic 
          suspension.
455.390  Vidarabine monohydrate ophthalmic ointment.

                      Subpart E--Otic Dosage Forms

455.410  Chloramphenicol otic.

                  Subpart F--Dermatologic Dosage Forms

455.510  Chloramphenicol dermatologic dosage forms.
455.510a  Chloramphenicol ointment (chloramphenicol cream).
455.510b  [Reserved]
455.510c  Chloramphenicol-polymyxin ointment.
455.510d  Fibrinolysin and desoxyribonuclease, combined (bovine) with 
          chloramphenicol ointment.
455.540  Mupirocin ointment.

    Authority:  21 U.S.C. 357.

[[Page 969]]


    Source:  39 FR 19166, May 30, 1974, unless otherwise noted.



                          Subpart A--Bulk Drugs



Sec. 455.4  Aztreonam.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Aztreonam is a practically odorless, 
white to slightly off-white fine powder. It is sparingly soluble in 
water of pH 2, and is very soluble at pH values above 4. Its solubility 
is slight to very slight in polar organic solvents such as methanol and 
ethanol and it is insoluble in nonpolar solvents such as hexane and 
heptane. It is so purified and dried that:
    (i) Its potency is not less than 900 micrograms of aztreonam per 
milligram on an ``as is'' basis.
    (ii) Its moisture content is not more than 2.0 percent.
    (iii) Its residue on ignition is not more than 0.1 percent.
    (iv) Its heavy metals content is not more than 30 parts per million.
    (v) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requirements for certification; samples. In addition to 
complying with the requirements of Sec. 431.1 of this chapter, each such 
request shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
residue on ignition, heavy metals, and identify.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research; 10 packages, each containing approximately 500 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 455.4a(b)(1).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (4) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
    (5) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 0.5 percent potassium bromide disc prepared as described in 
paragraph (b)(1) of that section, except prepare a solution containing 3 
milligrams of aztreonam per milliliter of methanol and use 0.5 
milliliter of the solution as the sample.
[54 FR 40385, Oct. 2, 1989]



Sec. 455.4a  Sterile aztreonam.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Aztreonam is a practically odorless, 
white to slightly off-white fine powder. It is sparingly soluble in 
water of pH 2, and is very soluble at pH values above 4. Its solubility 
is slight to very slight in polar organic solvents such as methanol and 
ethanol and it is insoluble in non-polar solvents such as hexane and 
heptane. It is so purified and dried that:
    (i) Its potency is not less than 900 micrograms of aztreonam per 
milligram on an ``as is'' basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 2.0 percent.
    (v) Its residue on ignition is not more than 0.1 percent.
    (vi) Its heavy metals content is not more than 30 parts per million.
    (vii) It passes the identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requirements for certification: samples. In addition to 
complying with the requirements of Sec. 431.1 of this chapter, each such 
request shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, residue on ignition, heavy metals, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.361 of this chapter, except in lieu of the guard column 
described in paragraph (a)(4) of that section, use a 5- to 10-centimeter 
precolumn having an inside diameter of 2 millimeters and packed with 
octadecyl silane chemically bonded to silica gel of a controlled surface 
porosity

[[Page 970]]

that has been bonded to a solid spherical core (U.S.P. designation L-2) 
30 micrograms to 50 micrograms in diameter; and use the resolution test 
solution to determine resolution in lieu of the working standard 
solution. Perform the assay at ambient temperature, using an ultraviolet 
detection system operating at a wavelength of 270 nanometers (or 254 
nanometers fixed mercury source), a column packed with octadecyl silane 
chemically bonded to porous silica or ceramic microparticles (U.S.P. 
designation L-1) 5 micrograms to 10 micrograms in diameter or 
equivalent, a flow rate of 1.5 milliliters per minute, and a known 
injection volume of 20 microliters. Reagents, working standard solution, 
sample solution, resolution test solution, system suitability 
requirements, and calculations are as follows:
    (i) Reagents--(a) 0.05M potassium phosphate buffer, pH 3.0. Prepare 
a solution containing 6.8 grams of potassium phosphate monobasic per 
liter of distilled water. Adjust the solution to pH 3.0 with 1M 
phosphoric acid.
    (b) Mobile phase. 0.05M potassium phosphate buffer, pH 3.0: methanol 
(4:1).
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(a) Working standard solution. Transfer approximately 25 
milligrams of aztreonam working standard, accurately weighed, to a 25-
milliliter volumetric flask. Dissolve and dilute to volume with mobile 
phase.
    (b) Sample solution. Transfer approximately 25 milligrams of the 
sample, accurately weighed, to a 25-milliliter volumetric flask. 
Dissolve and dilute to volume with mobile phase.
    (c) Resolution test solution. Dissolve 10 milligrams of [2S-
[2alpha,3beta(E]]-2-[[[1-(2-amino-4-thiazolyl)-2-[(2-methyl-4-oxo-1-
sulfo-3-azetidinyl)amino]-2-oxoethylidene] amino]oxy]-2-methylpropanoic 
acid (E isomer) in 10 milliliters of working standard solution and 
dilute to 50-milliliters with mobile phase.
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 2 at 5 percent 
of peak height.
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,000 theoretical plates.
    (c) Resolution. The resolution (R) between the peak for aztreonam 
and the E isomer is satisfactory if it is not less than 2.0.
    (d) Coefficient of variation. The coefficient of variation 
(SRin percent) of 5 replicate injections is satisfactory if 
it is not more than 2.0 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.361(b) of this chapter. Alternate chromatographic 
conditions are acceptable provided reproducibility and resolution are 
comparable to the system. However, the sample preparation described in 
paragraph (b)(1)(ii)(b) of this section should not be changed.
    (iv) Calculation. Calculate the micrograms of aztreonam per 
milligram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.316

where:

Au=Area of the aztreonam peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the aztreonam peak in the chromatogram of the 
          aztreonam working standard;
Ps=Aztreonam activity in the aztreonam working standard 
          solution in micrograms per milliliter; and
Cu=Milligrams of sample per milliliter of sample solution.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use diluting fluid I in lieu of diluting fluid A.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of aztreonam and 39 milligrams 
of pyrogen-free L-arginine base per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (6) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
    (7) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 0.5

[[Page 971]]

percent potassium bromide disc prepared as described in paragraph (b)(1) 
of that section, except prepare a solution containing 3 milligrams of 
aztreonam per milliliter of methanol and use 0.5 milliliter of the 
solution as the sample.
[52 FR 4614, Feb. 13, 1987, as amended at 55 FR 11584, Mar. 29, 1990]



Sec. 455.10  Chloramphenicol.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chloramphenicol is a white to grayish-
white or yellowish-white powder, occurring as needles or elongated 
plates. It is neutral, slightly soluble in water, but freely soluble in 
alcohol. It has the chemical formula D-(--)-threo--1-p--nitrophenyl-2 - 
dichloracetamido-1,3-propanediol. It is so purified and dried that:
    (i) Its potency is not less than 900 micrograms per milligram.
    (ii) [Reserved]
    (iii) Its pH in a saturated aqueous solution is not less than 4.5 
nor more than 7.5.
    (iv) Its specific rotation in absolute ethyl alcohol at 20 deg. C. 
is +20 deg.plus-minus1.5 deg., and at 25 deg. C. is 
+18.5 deg.plus-minus1.5 deg..
    (v) Its melting range is 151 deg.plus-minus2 deg. C.
    (vi) Its absorptivity at 278 nanometers is 100 
plus-minus3 percent of that of the chloramphenicol working 
standard similarly treated.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, pH, 
specific rotation, melting range, absorptivity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological turbidimetric assay shall be conclusive.
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 95 
percent ethyl alcohol to obtain a solution containing 10,000 micrograms 
of chloramphenicol per milliliter (estimated). Add sufficient distilled 
water to obtain a concentration of 1,000 micrograms of chloramphenicol 
per milliliter (estimated). Further dilute an aliquot of the stock 
solution with distilled water to the reference concentration of 2.5 
micrograms of chloramphenicol per milliliter (estimated).
    (ii) Spectrophotometric method. Dissolve approximately 50 milligrams 
each of the sample and working standard in 100 milliliters of distilled 
water. Warm if necessary to hasten dissolution. Transfer 10 milliliters 
into a 250-milliliter volumetric flask and fill to volume with distilled 
water. Using a suitable spectrophotometer equipped with a 1-centimeter 
cell and distilled water as the blank, determine the absorbance of each 
solution at 278 nanometers. Calculate the potency of chloramphenicol as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.317

    (2) [Reserved]
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
saturated aqueous solution.
    (4) Specific rotation. Accurately weigh approximately 1.25 grams of 
the sample into a 25-milliliter glass-stoppered volumetric flask and 
dissolve in about 15

[[Page 972]]

milliliters of absolute alcohol, warming if necessary. Dilute the 
solution to 25 milliliters with absolute alcohol and mix thoroughly. 
Proceed as directed in Sec. 436.210 of this chapter, using a 2.0-
decimeter polarimeter tube.
    (5) Melting range. Proceed as directed in Sec. 436.209 of this 
chapter.
    (6) Absorptivity. Proceed as directed in paragraph (b)(1)(ii) of 
this section, except calculate the percent relative absorptivity as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.318

    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19166, May 30, 1974, as amended at 45 FR 16476, Mar. 14, 1980; 48 
FR 3960, Jan. 28, 1983; 50 FR 19921, May 13, 1985]



Sec. 455.10a  Sterile chloramphenicol.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile chloramphenicol is a white to 
grayish-white or yellowish-white powder, occurring as needles or 
elongated plates. It is neutral, slightly soluble in water, but freely 
soluble in alcohol. It has the chemical formula D - (--) - threo - 1 - 
p--nitrophenyl - 2 - dichloracetamido - 1,3 - propanediol. It is so 
purified and dried that:
    (i) Its potency is not less than 900 micrograms per milligram.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv)--(v) [Reserved]
    (vi) Its pH in a saturated aqueous solution is not less than 4.5 nor 
more than 7.5.
    (vii) Its specific rotation in absolute ethyl alcohol at 20 deg. C. 
is +20 deg.plus-minus1.5 deg., and at 25 deg. C. is 
+18.5 deg.plus-minus1.5 deg..
    (viii) Its melting range is 151 deg.plus-minus2 deg. C.
    (ix) Its absorptivity at 278 nanometers is 100 
plus-minus3 percent of that of the chloramphenicol working 
standard similarly treated.
    (x) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, pH, specific rotation, melting range, absorptivity, and 
crystallinity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 50 milligrams.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods; however, the results obtained from the 
microbiological turbidimetric assay shall be conclusive.
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed portion of the sample in sufficient 95 
percent ethyl alcohol to obtain a solution containing 10,000 micrograms 
of chloramphenicol per milliliter (estimated). Add sufficient distilled 
water to obtain a concentration of 1,000 micrograms of chloramphenicol 
per milliliter (estimated). Further dilute an aliquot of the stock 
solution with distilled water to the reference concentration of 2.5 
micrograms of chloramphenicol per milliliter (estimated).
    (ii) Spectrophotometric method. Dissolve approximately 50 milligrams 
each of the sample and working standards in 100 milliliters of distilled 
water. Warm if necessary to hasten dissolution. Transfer 10 milliliters 
into a 250-milliliter volumetric flask and fill to volume with distilled 
water. Using a suitable spectrophotometer equipped with a 1-centimeter 
cell and distilled water

[[Page 973]]

as the blank, determine the absorbance of each solution at 278 
nanometers. Calculate the potency of chloramphenicol as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.319

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use 50 milligrams in lieu of 300 milligrams.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 5 milligrams of chloramphenicol per 
milliliter. Apply sufficient heat to dissolve the chloramphenicol.
    (4)--(5) [Reserved]
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
saturated aqueous solution.
    (7) Specific rotation. Accurately weigh approximately 1.25 grams of 
the sample in a 25-milliliter glass-stoppered volumetric flask and 
dissolve in about 15 milliliters of absolute alcohol, warming if 
necessary. Dilute the solution to 25 milliliters with absolute alcohol 
and mix thoroughly. Proceed as directed in Sec. 436.210 of this chapter, 
using a 2.0 decimeter polarimeter tube.
    (8) Melting range. Proceed as directed in Sec. 436.209 of this 
chapter.
    (9) Absorptivity. Proceed as directed in paragraph (b)(1)(ii) of 
this section except calculate the percent relative absorptivity as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.320

    (10) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19166, May 30, 1974, as amended at 45 FR 16476, Mar. 14, 1980; 45 
FR 64568, Sept. 30, 1980; 48 FR 3960, Jan. 28, 1983; 50 FR 19921, May 
13, 1985]



Sec. 455.11  Chloramphenicol palmitate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chloramphenicol palmitate is the white to 
grayish-white, tasteless palmitic acid ester of chloramphenicol. It is 
so urified and dried that:
    (i) It contains not less than 555 micrograms nor more than 595 
micrograms of chloramphenicol per milligram.
    (ii) [Reserved]
    (iii) Its melting range is 91 deg.plus-minus4 deg. C.
    (iv) Its specific rotation in absolute ethyl alcohol at 25 deg. C. 
is +23 deg.plus-minus2 deg..
    (v) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for chloramphenicol 
content, melting range, specific rotation, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Chloramphenicol content. Proceed 
as directed in Sec. 436.335 of this chapter.
    (2) [Reserved]

[[Page 974]]

    (3) Melting range. Proceed as directed in Sec. 436.209 of this 
chapter.
    (4) Specific rotation. Accurately weigh approximately 1.25 grams of 
sample in a 25-milliliter, glass-stoppered volumetric flask and dissolve 
in about 15 milliliters of absolute alcohol, warming if necessary to 
effect solution. Bring the solution to 25 deg. C. Dilute the solution to 
25 milliliters with absolute alcohol and mix thoroughly. Proceed as 
directed in Sec. 436.210 of this chapter, using a 2.0-decimeter 
polarimeter tube.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19166, May 30, 1974, as amended at 49 FR 6093, Feb. 17, 1984; 50 
FR 19921, May 13, 1985]



Sec. 455.12a  Sterile chloramphenicol sodium succinate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chloramphenicol sodium succinate is the 
light-yellow, water-soluble, ethanol-soluble sodium salt of the 3-
monosuccinate ester of chloramphenicol. It is so purified and dried 
that:
    (i) Its potency is not less than 650 and not more than 765 
micrograms per milligram. If it is packaged for dispensing, its potency 
when reconstituted as directed in the labeling is satisfactory if it is 
not less than 90 percent and not more than 115 percent of the number of 
milligrams of chloramphenicol per milliliter that it is represented to 
contain.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv)--(v) [Reserved]
    (vi) Its moisture content is not more than 5.0 percent.
    (vii) Its pH in an aqueous solution containing 250 milligrams of 
chloramphenicol per milliliter is not less than 6.4 and not more than 
7.0.
    (viii) Its specific rotation in an aqueous solution containing 50 
milligrams per milliliter at 25 deg. C. is 
+6.5 deg.plus-minus1.5 deg..
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, and specific rotation.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 500 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 8 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency--(i) Working standard. 
Dissolve an accurately weighed portion of the chloramphenicol working 
standard in sufficient distilled water to give a solution containing 20 
micrograms per milliliter. Using a suitable spectrophotometer and 
distilled water as the blank, determine the absorbance of this solution 
in a 1-centimeter cell at a wavelength of 278 nanometers.
    (ii) Procedure. Dissolve an accurately weighed portion of the sample 
to be tested in sufficient distilled water to give a solution containing 
30 micrograms of the sample per milliliter (estimated); and also if it 
is packaged for dispensing, reconstitute as directed in the labeling. 
Remove an accurately measured representative portion from each container 
and further dilute this portion with sufficient distilled water to give 
a concentration of 20 micrograms of chloramphenicol per milliliter 
(estimated). Using a suitable spectrophotometer and distilled water as 
the blank, determine the absorbance of this solution in a 1-centimeter 
cell at a wave length of 276 nanometers. Calculate the micrograms per 
milligram of the dry powder as follows:

[[Page 975]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.321



Calculate the milligrams per milliliter of the reconstituted solution in 
the dispensing container as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.322

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 5 milligrams of chloramphenicol per 
milliliter.
    (4)-(5) [Reserved]
    (6) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (7) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 250 milligrams of chloramphenicol per 
milliliter.
    (8) Specific rotation. Dilute the sample with sufficient distilled 
water to give a solution containing approximately 50 milligrams per 
milliliter. Proceed as directed in Sec. 436.210 of this chapter, using a 
1.0-decimeter polarimeter tube. Calculate the specific rotation on the 
anhydrous basis.
[39 FR 19166, May 30, 1974, as amended at 39 FR 37486, Oct. 22, 1974; 45 
FR 64568, Sept. 30, 1980; 50 FR 1504, Jan. 11, 1985; 50 FR 19921, May 
13, 1985]



Sec. 455.15  Clavulanate potassium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clavulanate potassium is the potassium 
salt of Z-(2R,5R)-3-(2-hydroxyethylidene)-7-oxo-4-oxa-1-
azabicyclo[3.2.0]heptane-2-carboxylic acid. It is so purified and dried 
that:
    (i) It is equivalent to not less than 755 micrograms and not more 
than 920 micrograms of clavulanic acid per milligram on an anhydrous 
basis.
    (ii) Its moisture content is not more than 1.5 percent.
    (iii) Its pH in an aqueous solution containing 10 milligrams per 
milliliter is not less than 5.5 and not more than 8.0.
    (iv) It gives a positive identity test.
    (v) Its content of the potassium salt of [3R,5S]-7-oxo-4-oxa-1-
azabicyclo[3.2.0]heptane-3-carboxylic acid (clavam-2-carboxylate) is 
satisfactory if it is not greater than .01 percent.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, identity, and clavam-2-carboxylate content.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 12 packages, each containing approximately 300 
milligrams.
    (b) Tests and methods of assay--(1) Clavulanic acid content. Proceed 
as directed in Sec. 436.351 of this chapter, using ambient temperature, 
an ultraviolet

[[Page 976]]

detection system operating at a wavelength between 220 and 230 
nanometers, and a column packed with microparticulate (3 to 10 
micrometers in diameter) reversed phase packing material such as 
octadecyl silane bonded silica. Reagents, working standard and sample 
solutions, system suitability requirements, and calculations are as 
follows:
    (i) Reagents--(a) 0.05M Sodium phosphate buffer solution, pH 4.4. 
Transfer 7.8 grams of monobasic sodium phosphate to a 1-liter volumetric 
flask and dissolve in 900 milliliters of distilled water. Adjust the pH 
to 4.40.1 with 18N phosphoric acid or 10N sodium hydroxide. 
Dilute to volume with distilled water. Mix well.
    (b) Mobile phase. Mix methanol: 0.05M sodium phosphate buffer, pH 
4.4 (5:95 v/v) and stir or ultrasonicate for no less than 2 minutes. 
Degas by passing through a 0.5-micrometer filter with vacuum. The mobile 
phase may be sparged with helium through a 2-micrometer metal filter for 
the duration of the analysis. Adjust the ratio of methanol to aqueous 
buffer as necessary to obtain satisfactory retention of the peaks.
    (ii) Preparation of clavulanic acid working standard and sample 
solutions. Accurately weigh and transfer into volumetric flasks 
sufficient clavulanic acid working standard or clavulanate potassium 
sample to obtain a final concentration of 250 micrograms per milliliter. 
To the clavulanic acid working standard, add sufficient amoxicillin 
trihydrate to provide a final concentration of 500 micrograms per 
milliliter. (The amoxicillin serves as an internal marker for system 
suitability testing.) Dissolve in water by shaking or ultrasonicating 
until solution becomes clear. Dilute the solutions as required to final 
volume with water. Use within 8 hours.
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.5.
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 550 theoretical plates.
    (c) Resolution factor. The resolution factor (R) between the 
clavulanic acid and amoxicillin peaks is satisfactory if it is not less 
than 3.5.
    (d) Coefficient of variation. The coefficient of variation 
(SRin percent) is satisfactory if it is not more than 2.0 
percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.351(b) of this chapter.
    (iv) Calculations. Calculate the micrograms of clavulanic acid per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.323

where:
Au=The clavulanic acid peak response in the chromatogram of 
          the sample (at a retention time equal to that observed for the 
          standard);
As=The clavulanic acid peak response in the chromatogram of 
          the clavulanic acid working standard);
Ps=Potency of the clavulanic acid working standard in 
          micrograms per milligram;
Wu=Milligrams of sample;
Ws=Milligrams of standard; and
m=Percent moisture content of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter.
    (4) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation described in paragraph (b)(2) of that 
section.
    (5) Clavam-2-carboxylate content. Proceed as directed in 
Sec. 436.352 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 210 nanometers, and a 
column packed with microparticulate (3 to 10 micrometers in diameter) 
reversed phase packing materials such as octadecyl silane bonded silica. 
Mobile phase, working standard and sample solutions, system suitability 
requirements, and calculations are as follows:
    (i) Mobile phase. 0.1M Sodium phosphate buffer solution, pH 4.0. 
Prepare a 0.1M aqueous solution of monobasic sodium phosphate and adjust 
to pH 4.0 with phosphoric acid.
    (ii) Working standard and sample solutions--(a) Preparation of 
working standard solution. Accurately weigh and

[[Page 977]]

transfer into a 50-milliliter volumetric flask approximately 16 
milligrams of clavam-2-carboxylate authentic sample. Dilute to volume 
and transfer 10 milliliters into a 100-milliliter flask. Dilute to 
volume with water.
    (b) Preparation of sample solution. Accurately weigh 100 milligrams 
of the sample into a 10-milliliter flask. Dilute to volume with water.
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) for the clavulanate standard peak is satisfactory if 
it is not more than 1.5.
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 4,000 theoretical plates.
    (c) Resolution factor. The resolution factor (R) between the 
clavulanic acid and clavam-2-carboxylic acid peaks is satisfactory if it 
is greater than 1.0.
    (d) Coefficient of variation (Relative standard deviation). The 
coefficient of variation (SRin percent) is satisfactory if it 
is not more than 2.0 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.352(b) of this chapter.
    (iv) Calculations. Calculate the percent of clavam-2-carboxylate 
content as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.074

where:
P=Percent clavam-2-carboxylic acid in the standard.
[49 FR 39674, Oct. 10, 1984, as amended at 55 FR 11584, Mar. 29, 1990]



Sec. 455.15a  Sterile clavulanate potassium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clavulanate potassium is the potassium 
salt of Z-(2R,5R)-3-(2-hydroxyethylidene)-7-oxo-4-oxa-1-
azabicyclo[3.2.0]heptane-2-carboxylic acid. It is so purified and dried 
that:
    (i) It is equivalent to not less than 755 micrograms and not more 
than 920 micrograms of clavulanic acid per milligram on an anhydrous 
basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 1.5 percent.
    (v) Its pH of an aqueous solution containing 10 milligrams per 
milliliter is not less than 5.5 and not more than 8.0.
    (vi) It gives a positive identity test.
    (vii) Its [3R,5S]-7-oxo-4-oxa-1-azabicyclo[3.2.0]heptane-3-
carboxylic acid (clavam-2-carboxylate) content is satisfactory if it is 
not greater than .01 percent.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, identity, and clavam-2-carboxylate content.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 12 packages, each containing approximately 300 
milligrams.
    (b) Tests and methods of assay--(1) Clavulanic acid content. Proceed 
as directed in Sec. 455.15(b)(1) of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 10 milligrams per milliliter of clavulanate 
potassium.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 10 milligrams per milliliter.
    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation described in paragraph (b)(2) of that 
section.
    (7) Clavam-2-carboxylate content. Proceed as directed in 
Sec. 455.15(b)(5) of this chapter.
[50 FR 33519, Aug. 20, 1985, as amended at 54 FR 11584, Mar. 29, 1990]



Sec. 455.20  Cycloserine.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cycloserine is a white to slightly 
yellowish compound. It has the

[[Page 978]]

chemical structure D-4-amino-3-isoxazolidone. It is so purified that:
    (i) Its potency is not less than 900 micrograms per milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 1.0 percent.
    (iv) Its pH in a 10 percent aqueous solution is not less than 5.5 
and not more than 6.5.
    (v) Its residue on ignition is not more than 0.5 percent.
    (vi) It gives a positive identity for cycloserine.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, residue on ignition, crystallinity, and identity.
    (ii) Samples of the batch: 10 packages, each containing 
approximately 500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Using the cycloserine 
working standard as the standard of comparison, assay for potency by 
either of the following methods; however, the results obtained from the 
microbiological turbidimetric assay shall be conclusive.
    (i) Colorimetric assay--(a) Stockstandard solution. Dry 
approximately 100 milligrams of the working standard for 3 hours at 
60 deg. C. and a pressure of 5 millimeters or less. Determine the dry 
weight and dissolve the dried working standard in sufficient distilled 
water to give a solution containing 1,000 micrograms per milliliter. 
This solution may be used for 1 month if kept under refrigeration.
    (b) Standard curve solutions. Pipette accurately 0.0, 1.0, 5.0, 
10.0, 15.0, and 20.0 milliliters of the stock standard solution to each 
of six 100-milliliter volumetric flasks, dilute to 100 milliliters with 
0.1N sodium hydroxide and mix thoroughly.
    (c) Reagents:
    (1) Acetic acid--1.0N solution.
    (2) Sodium hydroxide--4.0N and 0.1N solutions.
    (3) Sodium nitroprusside--4.0 percent solution: Dissolve 4.0 grams 
in sufficient distilled water to make 100.0 milliliters. Mix well. Store 
in amber bottle.
    (4) Oxidized nitroprusside reagent--Mix equal parts of 4 percent 
sodium nitroprusside solution and 4.0N sodium hydroxide, and let stand 
for 1 hour before using. Prepare daily, and store in an amber bottle.
    (d) Procedure. Transfer approximately 100 milligrams of sample, 
accurately weighed, to a 100 milliliter volumetric flask. Dissolve in 
sufficient 0.1N sodium hydroxide to measure exactly 100 milliliters. Mix 
thoroughly and transfer 10 milliliters to a second 100-milliliter 
volumetric flask, and mix thoroughly. Transfer exactly 1.0 milliliter of 
each of the standard curve solutions and of the sample solution to 
respective test tubes. Add exactly 3.0 milliliters of 1.0N acetic acid 
to each of the test tubes. Mix thoroughly. Add exactly 1.0 milliliter of 
oxidized nitroprusside reagent to each test tube and mix thoroughly. 
Allow the tubes to stand at room temperature for at least 10 minutes in 
order that maximum color intensity may develop. Using the solution 
containing 0.0 milliliter of working standard as a blank, determine the 
absorbances of the solutions at 625 nanometers in a suitable 
spectrophotometer. Plot concentration versus absorbance on linear graph 
paper. The curve may deviate slightly from a straight line. The standard 
curve solutions equal 0, 10, 50, 100, 150, and 200 micrograms of 
cycloserine, respectively.
    (e) Calculations:

Micrograms cycloserine per milligram = (Concentration in micrograms from 
          calibration curve  x  1,000)/Weight of original sample in 
          milligrams.

    (ii) Microbiological turbidimetric assay. Proceed as described in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient sterile distilled 
water to give a stock solution of convenient concentration. Further 
dilute the stock solution with sterile distilled water to the reference 
concentration of 50 micrograms of cycloserine per milliliter 
(estimated).
    (2) [Reserved]

[[Page 979]]

    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution with a concentration 100 milligrams per milliliter.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (6) Residue on ignition. Proceed as directed in Sec. 436.207(a) of 
this chapter.
    (7) Identity. Proceed as directed in paragraph (b)(1)(i) of this 
section.
[39 FR 19166, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 455.40  Mupirocin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Mupirocin is nonanoic acid, 9-[[3-methyl-
1-oxo-4-[tetrahydro-3,4-dihydroxy-5-[[3-(2-hydroxy-l-
methylpropyl)oxiranyl]methyl]-2H-pyran-2-yl]-2-butenyl]oxy]-,[2S-
[2(E),3B,4B,5[2R*,3R*(1R*,2R*)]]]-. It is a white to 
off-white crystalline solid. It is so purified and dried that:
    (i) Its potency is not less than 920 micrograms per milligram on an 
anhydrous basis.
    (ii) Its moisture content is not more than 1.0 percent.
    (iii) The pH of a saturated aqueous solution of mupirocin is not 
less than 3.5 and not more than 4.0.
    (iv) It is crystalline.
    (v) It gives a positive identity test for mupirocin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, crystallinity, and identity.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research: 10 packages, each containing approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 229 nanometers, a column 
packed with microparticulate (3 to 10 micrometers in diameter) reversed 
phase packing material such as an octadecylsilane, a flow rate of not 
more than 2.0 milliliters per minute, and a known injection volume of 
between 10 and 20 microliters. Use the resolution test solution to 
determine resolution in lieu of the working standard solution. Reagents, 
working standard and sample solutions, resolution test solution, system 
suitability requirements, and calculations are as follows:
    (i) Reagents--(A) Acetonitrile. Distilled in glass. Ultraviolet 
grade.
    (B) Phosphate buffer, pH 6.3. Prepare a 0.05M sodium monobasic 
phosphate solution and adjust to pH 6.3 with 1.0N sodium hydroxide.
    (C) Mobile phase. To 750 milliliters of 0.05M, pH 6.3 phosphate 
buffer, add 250 milliliters of acetonitrile. Filter through a suitable 
filter capable of removing particulate matter to 0.5 micron in diameter. 
Degas the mobile phase just prior to its introduction into the 
chromatograph.
    (ii) Preparation of working standard, sample, and resolution, test 
solutions--(A) Working standard solution. Accurately weigh approximately 
11 milligrams of the mupirocin working standard into a 100-milliliter 
volumetric flask. Dissolve the standard in about 20 milliliters of 
acetonitrile and dilute to volume with pH 6.3 phosphate buffer. Mix 
well.
    (B) Sample solution. Transfer approximately 11 milligrams of sample, 
accurately weighed, to a 100-milliliter volumetric flask. Dissolve the 
sample in about 20 milliliters of acetonitrile and dilute to volume with 
pH 6.3 phospate buffer. Mix well.
    (C) Resolution test solution. Acidify approximately 10 milliliters 
of the working standard solution with 6N hydrochloric acid to pH 2.0. 
Allow to stand at room temperature for about 2 hours. Neutralize this 
solution. Use this solution to determine the resolution requirement for 
the chromatographic system.
    (iii) System suitability requirements--(A) Asymmetry factor. 
Calculate the asymmetry factor (As), measured at a point 5 
percent of the peak height from the baseline as follows:

[[Page 980]]

[GRAPHIC] [TIFF OMITTED] TR01JA93.324


where:

a=Horizontal distance from point of ascent to point of maximum peak 
          height; and
b=Horizontal distance from the point of maximum peak height to point of 
          descent.

    The asymmetry factor (As) is satisfactory if it is not 
more than 1.5.

    (B) Efficiency of the column. From the number of theoretical plates 
(n) calculated as described in Sec. 436.216(c)(2) of this chapter, 
calculate the reduced plate height (hr) as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.325

where:

L=Length of the column in centimeters;
n=Number of theoretical plates; and
dP=Average diameter of the particles in the analytical column 
          packing in micrometers.

The absolute efficiency (hr) is satisfactory if it is not 
more than 20.0, equivalent to 1,500 theoretical plates for a 30-
centimeter column of 10 micrometer particles.

    (C) Resolution factor. The resolution factor (Rs) between 
the peak for mupirocin and its nearest eluting peak produced from its 
acid degradation is satisfactory if it is not less than 2.0. The 
chromatogram of the resolution test solution should show a significantly 
reduced mupirocin peak immediately preceded by a peak due to mupirocin 
degradation products. This degradation peak may appear as a single peak 
or be partially resolved showing a shoulder or two overlapping peaks.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent of 5 replicate 
injections) is satisfactory if it is not more than 2.0 percent.

If the system suitability parameters have been met, then proceed as 
described in Sec. 436.216(b) of this chapter.
    (iv) Calculations. Calculate the micrograms of mupirocin per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.326

where:

Au=Area of the mupirocin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the mupirocin peak in the chromatogram of the 
          mupirocin working standard;
Ps=Mupirocin activity in the mupirocin working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of mupirocin sample per milliliter of sample 
          solution;
m=Percent moisture content of the sample.

    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter using a 
saturated aqueous solution.
    (4) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (5) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation method described in Sec. 436.211(b)(2).
[55 FR 2641, Jan. 26, 1990; 55 FR 11110, Mar. 26, 1990; 55 FR 14378, 
Apr. 17, 1990]



Sec. 455.50  Calcium novobiocin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Calcium novobiocin is the calcium salt of 
a kind of novobiocin or a mixture of two or more such salts. It is so 
purified and dried that:
    (i) Its potency is not less than 840 micrograms per milligram, 
expressed in terms of novobiocin on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 10 percent.
    (iv) Its pH in a saturated aqueous suspension containing 25 
milligrams per milliliter is not less than 6.5 and not more than 8.5.
    (v) Its specific rotation in an acidmethyl alcohol solution at 
25 deg. C. is not less than -50 deg. and not more than -58 deg..
    (vi) It demonstrates a positive color identity test.
    (vii) It is crystalline.

[[Page 981]]

    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, specific rotation, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in 5 milliliters of absolute ethyl 
alcohol and then dilute with sufficient 0.1M potassium phosphate buffer, 
pH 8.0 (solution 3), to give a stock solution of 1,000 micrograms 
(estimated) per milliliter. Further dilute with 10 percent potassium 
phosphate buffer, pH 6.0 (solution 6), to the reference concentration of 
0.5 microgram of novobiocin per milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
saturated aqueous suspension prepared by suspending 25 milligrams of 
calcium novobiocin per milliliter.
    (5) Specific rotation. Proceed as directed in Sec. 455.51a(b)(8).
    (6) Identity. Proceed as directed in Sec. 455.51(b)(7).
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19166, May 30, 1974, as amended at 41 FR 10886, Mar. 15, 1976; 43 
FR 9801, Mar. 10, 1978; 50 FR 19921, May 13, 1985]



Sec. 455.51  Sodium novobiocin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sodium novobiocin is the monosodium salt 
of a kind of novobiocin or a mixture of two or more such salts. It is so 
purified and dried that:
    (i) Its potency is not less than 850 micrograms of novobiocin per 
milligram, calculated on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 6.0 percent.
    (iv) Its pH in a solution containing 25 milligrams per milliliter is 
not less than 6.5 and not more than 8.5.
    (v) Its residue on ignition is not less than 10.5 percent and not 
more than 12.0 percent, calculated on an anhydrous basis.
    (vi) Its specific rotation in an acidmethyl alcohol solution at 
25 deg. C. is not less than -50 deg. and not more than -58 deg..
    (vii) It demonstrates a positive color identity test.
    (viii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification: samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, residue on ignition, specific rotation, identity and 
crystallinity.
    (ii) Samples required on the batch; 10 packages, each containing 
approximately 600 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), to give a stock solution of 
convenient concentration. Further dilute with 10 percent potassium 
phosphate buffer, pH 6.0 (solution 6), to the reference concentration of 
0.5 microgram of novobiocin per milliliter (estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 25 milligrams of sodium novobiocin per milliliter.
    (5) Residue on ignition. Proceed as directed in Sec. 436.207(b) of 
this chapter, calculating on the basis of an anhydrous sample weight.
    (6) Specific rotation. Accurately weigh approximately 1.25 grams of 
the sample in a 25-milliliter glass-stoppered volumetric flask. Prepare 
an acid-methyl

[[Page 982]]

alcohol solution by diluting 1.0 milliliter of concentrated hydrochloric 
acid to a volume of 100 milliliters with absolute methyl alcohol and mix 
well. Dissolve the sample in about 15-milliliters of the acid-methyl 
alcohol solution. Adjust to volume with the acid-methyl alcohol solution 
and mix well. Proceed as directed in Sec. 436.210 of this chapter, using 
a 2.0-decimeter polarimeter tube. Calculate the specific rotation on the 
anhydrous basis.
    (7) Identity. (i) Using 0.1M aqueous sodium borate as a diluent, 
prepare 10 milliliters of a solution containing the equivalent of 1 
milligram (approximate) of novobiocin per milliliter.
    (ii) Prepare a saturated aqueous solution of N,2,6-
trichloroquinoneimine by shaking continuously for 30 minutes in a dark 
bottle 25 milligrams of N,2,6-trichloroquinoneimine in 100 milliliters 
of distilled water. Let stand 2 hours after shaking. Store in the dark 
bottle.
    (iii) Add 2.0 milliliters of the saturated N,2,6-
trichloroquinoneimine solution to 4 milliliters of the novobiocin 
solution. Mix well and heat in a water bath at 37 deg. C. for 10 
minutes. The development of a blue color is a positive test for the 
presence of novobiocin. To 2 milliliters of the blue solution, add 2 
milliliters of N-butyl alcohol and shake well. A green color should 
develop in the butyl alcohol layer. To the other 2-milliliter portion of 
the blue solution, add 2 milliliters of benzene (c.p.), and shake well. 
A pink color should be developed in the benzene layer.
    (8) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19166, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 455.51a  Sterile sodium novobiocin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sodium novobiocin is the crystalline 
monosodium salt at a kind of novobiocin or a mixture of two or more such 
salts. It is so purified and dried that:
    (i) Its potency is not less than 850 micrograms of novobiocin per 
milligram, calculated on an anhydrous basis.
    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) [Reserved]
    (v) Its loss on drying is not more than 6.0 percent.
    (vi) Its pH in a solution containing 25 milligrams per milliliter is 
not less than 6.5 and not more than 8.5.
    (vii) Its residue on ignition is not less than 10.5 percent and not 
more than 12.0 percent calculated on an anhydrous basis.
    (viii) Its specific rotation in an acidmethyl alcohol solution at 
25 deg. C. is not less than -50 deg. and not more than -58 deg..
    (ix) It demonstrates a positive color identity test.
    (x) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, loss on drying, pH, residue on ignition, specific rotation, 
identity, and crystallinity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 600 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), to give a stock solution of 
convenient concentration. Further dilute with 10 percent potassium 
phosphate buffer, pH 6.0 (solution 6), to the reference concentration of 
0.5 microgram of novobiocin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 10 milligrams of novobiocin per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.

[[Page 983]]

    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 25 milligrams of sodium novobiocin per milliliter.
    (7) Residue on ignition. Proceed as directed in Sec. 436.207(b) of 
this chapter, calculating on the basis of an anhydrous sample weight.
    (8) Specific rotation. Accurately weigh approximately 1.25 grams of 
the sample in a 25-milliliter glass-stoppered volumetric flask. Prepare 
an acid-methyl alcohol solution by diluting 1.0 milliliter of 
concentrated hydrochloric acid to a volume of 100 milliliters with 
absolute methyl alcohol and mix well. Dissolve the sample in about 15-
milliliters of the acid-methyl alcohol solution. Adjust to volume with 
the acid-methyl alcohol solution and mix well. Proceed as directed in 
Sec. 436.210 of this chapter, using a 2.0-decimeter polarimeter tube. 
Calculate the specific rotation on an anhydrous basis.
    (9) Identity. (i) Using 0.1M aqueous sodium borate as a diluent, 
prepare 10 milliliters of a solution containing the equivalent of 1 
milligram (approximate) of novobiocin per milliliter.
    (ii) Prepare a saturated aqueous solution of N,2,6-
trichloroquinoneimine by shaking continuously for 30 minutes in a dark 
bottle 25 milligrams of N,2,6-trichloroquinoneimine in 100 milliliters 
of distilled water. Let stand 2 hours after shaking. Store in the dark 
bottle.
    (iii) Add 2.0 milliliters of the saturated N,2,6-
trichloroquinoneimine solution to 4 milliliters of the novobiocin 
solution. Mix well and heat in a water bath at 37 deg. C. for 10 
minutes. The development of a blue color is a positive test for the 
presence of novobiocin. To 2 milliliters of the blue solution, add 2 
milliliters of N-butyl alcohol and shake well. A green color should 
develop in the butyl alcohol layer. To the other 2-milliliter portion of 
the blue solution, add 2 milliliters of benzene (c.p.), and shake well. 
A pink color should develop in the benzene layer.
    (10) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19166, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 455.70  Rifampin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Rifampin is a red-brown powder. It is 3-
(4-methylpiperazinyliminomethyl) rifamycin SV. It is very slightly 
soluble in water, soluble in ethyl acetate and in methyl alcohol, and 
freely soluble in chloroform. It is so purified and dried that:
    (i) Its potency is not less than 900 micrograms per milligram.
    (ii) [Reserved]
    (iii) Its loss on drying is not more than 2 percent.
    (iv) Its pH is not less than 4.0 and not more than 6.0 in a 1 
percent aqueous suspension.
    (v) When calculated on the anhydrous basis, its absorptivity at 475 
nanometers is 100plus-minus4 percent of that of the rifampin 
working standard, similarly treated.
    (vi) It passes the identity test.
    (vii) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5(b) of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, loss on 
drying, pH, absorptivity, identity, and crystallinity.
    (ii) Samples required: 10 packages, each containing approximately 
300 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample in sufficient methyl alcohol to 
give a stock solution containing 1.0 milligram of rifampin per 
milliliter (estimated). Further dilute an aliquot of the stock solution 
with 1 percent potassium phosphate buffer, pH 6.0 (solution 1), to the 
reference concentration of 5.0 micrograms of rifampin per milliliter 
(estimated).
    (2) [Reserved]
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter, except dry the sample for 4 hours.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
1 percent aqueous suspension.
    (5) Absorptivity. Determine the absorbance of the sample and 
standard

[[Page 984]]

solutions in the following manner: Dissolve approximately 100 milligrams 
each of the sample and standard in a 100-milliliter volumetric flask 
containing 50 milliliters of absolute methyl alcohol, and dilute to 
volume with absolute methyl alcohol. Transfer a 2-milliliter aliquot to 
a 100-milliliter volumetric flask, and dilute to volume with 1 percent 
potassium phosphate buffer, pH 6.0, as listed in Sec. 436.101(a)(1) of 
this chapter. Using a suitable spectrophotometer equipped with a 1-
centimeter cell, immediately determine the absorption of each solution 
at 475 nanometers with the blank containing the same proportion of 
solution 1 and methyl alcohol as the sample and standard solutions. 
Calculate the absorptivity as follows:
[GRAPHIC] [TIFF OMITTED] TC01AP94.075

where:
m1=percent moisture in standard;
m2=percent moisture in sample.

    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the sample preparation method described in paragraph (b)(3) of 
that section, except use a 4 percent solution of the sample in 
chloroform and 0.1-millimeter matched absorption cells.
    (7) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19166, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 455.80a  Sterile spectinomycin hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile spectinomycin hydrochloride is 
the pentahydrated dihydrochloride salt of decahydro-4a, 7, 9-trihydroxy-
2-methyl-6,8-bis(methyl-amino)4H-pyrano[2,3-b][1,4]benzodioxin-4-one. It 
is so purified and dried that:
    (i) Its spectinomycin content is not less than 603 micrograms per 
milligram. If it is packaged for dispensing, its spectinomycin content 
is satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of spectinomycin that it is 
represented to contain.
    (ii) Its microbiological activity is not less than 603 micrograms of 
spectinomycin per milligram.
    (iii) It is sterile.
    (iv) It is nonpyrogenic.
    (v) [Reserved]
    (vi) It contains no depressor substances.
    (vii) Its moisture content is not less than 16 percent nor more than 
20 percent.
    (viii) Its pH is an aqueous solution containing 10 milligrams per 
milliliter is not less than 3.8 nor more than 5.6. If it is packaged for 
dispensing, when reconstituted as directed in the labeling, its pH is 
not less than 4.0 nor more than 7.0.
    (ix) It passes the identity test.
    (x) Its residue on ignition is less than 1 percent.
    (xi) It is crystalline.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for spectinomycin 
content, microbiological activity, sterility, pyrogens, depressor 
substances, moisture, pH, identity, residue on ignition, and 
crystallinity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use in the 
manufacture of another drug:
    (1) For all tests except sterility: eight packages, each containing 
approximately 300 milligrams and two containing not less than 3 grams.

[[Page 985]]

    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Spectinomycin content (vapor 
phase chromatography). Proceed as directed in Sec. 436.307 of this 
chapter; and also, if the batch is packaged for dispensing prepare the 
sample for assay as follows: Reconstitute the vial as directed in the 
labeling. Then using a suitable hypodermic needle and syringe, remove 
all of the withdrawable contents if it is represented as a single dose 
container, or if the labeling specifies the amount of spectinomycin 
content in a given volume of the resultant preparation remove an 
accurately measured representative portion from the container. Dilute 
the sample with water to a concentration equivalent to about 20 
milligrams per milliliter of spectinomycin. Transfer 1.0 milliliter of 
the diluted sample to a 25-milliliter glass-stoppered Erlenmeyer flask 
and dry by lyophilization. Proceed as directed in Sec. 436.307(d)(1)(ii) 
of this chapter. Calculate the spectinomycin content as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.327

where:
[GRAPHIC] [TIFF OMITTED] TR01JA93.328

Ws=Weight of the spectinomycin working standard in 
milligrams;
D=Dilution of the spectinomycin dose;
f=Potency of the spectinomycin working standard in milligrams of 
spectinomycin per milligram.

    (2) Microbiological activity (microbiological turbidimetric assay). 
Proceed as directed in Sec. 436.106 of this chapter, preparing the 
sample for assay as follows: Dissolve an accurately weighed sample in 
sufficient sterile distilled water to give a stock solution of 
convenient concentration. Further dilute an aliquot of the stock 
solution with sterile distilled water to the reference concentration of 
30.0 micrograms of spectinomycin per milliliter (estimated).
    (3) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (4) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 50 milligrams of spectinomycin base per 
milliliter.
    (5) [Reserved]
    (6) Depressor substances. Proceed as directed in Sec. 436.35 of this 
chapter.
    (7) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (8) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 10 milligrams per milliliter, except, if 
it is packaged for dispensing, use the suspension obtained after 
reconstituting the drug as directed in the labeling.
    (9) Identity test. Proceed as directed in Sec. 436.211 of this 
chapter, using the method described in paragraph (b)(2) of that section.
    (10) Residue on ignition. Proceed as directed in Sec. 436.207 of 
this chapter, using the method described in paragraph (b) of that 
section.
    (11) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
[39 FR 19166, May 30, 1974, as amended at 46 FR 60568, Dec. 11, 1981; 50 
FR 19921, May 13, 1985]



Sec. 455.82a  Sterile sulbactam sodium.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile sulbactam sodium is sodium 
(2S,5R)-3,3-dimethyl-7-oxo-4-thia-1-azabicyclo[3.2.0]heptane-2-
carboxylate 4,4 dioxide. It is so purified and dried that:
    (i) Its sulbactam potency is not less than 886 micrograms and not 
more than 941 micrograms per milligram on an anhydrous basis.

[[Page 986]]

    (ii) It is sterile.
    (iii) It is nonpyrogenic.
    (iv) Its moisture content is not more than 1 percent.
    (v) It is crystalline.
    (vi) It passes the identity test for sulbactam sodium.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, crystallinity, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 30 packages, each containing approximately 300 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 230 nanometers, a column 
packed with microparticulate (3 to 10 micrometers in diameter) reversed 
phase packing material such as octadecyl hydrocarbon bonded silica, a 
flow rate of 2.0 milliliters per minute, and a known injection volume of 
10 microliters. Reagents, working standard and sample solutions, system 
suitability requirements, and calculations are as follows:
    (i) Reagents--(A) 1.0M Phosphoric acid. Prepare by dissolving 67.5 
milliliters of reagent grade phosphoric acid (85 percent) in distilled 
water and dilute to 1 liter.
    (B) 0.005M Tetrabutylammonim hydroxide. Dilute 6.6 milliliters of 
tetrabutylammonium hydroxide (40 percent) to 1,800 milliliters with 
distilled water. Adjust the pH to 5.0 with 1.0M phosphoric acid and 
dilute with distilled water to 2 liters.
    (C) Mobile phase. Mix 350 milliliters of acetonitrile with 1,650 
milliliters of 0.005M tetrabutylammonium hydroxide. Filter and degas the 
mobile phase just prior to its introduction into the chromatographic 
pumping system. (Slight adjustments in pH and/or acetonitrile content 
may be made to achieve the system suitability parameters defined in 
paragraph (b)(1)(iii) of this section.)
    (ii) Preparation of working standard and sample solutions--(A) 
Working standard solution. Dissolve an accurately weighed portion of 
sulbactam working standard in sufficient mobile phase to give a stock 
solution of a known concentration containing about 1 milligram of 
sulbactam per milliliter.
    (B) Sample solution. Dissolve an accurately weighed portion of the 
sample in sufficient mobile phase to give a stock solution containing 1 
milligram of sulbactam per milliliter (estimated).
    (iii) System suitability requirements--(A) Tailing factor. The 
tailing factor (T) is satisfactory if it is not more than 1.5 at 10 
percent of peak height in lieu of 5 percent of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory for sulbactam if it is greater than 3,500 theoretical 
plates for a 30-centimeter column.
    (C) Resolution. The resolution (R) between the peaks for sulbactam 
and penicillanic acid is satisfactory if it is not less than 3.8.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent) of 5 replicate 
injections is satisfactory if it is not more than 2.0 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.216(b) of this chapter. Alternate chromatographic 
conditions are acceptable provided reproducibility and resolution are 
comparable to the system. However, the sample preparation described in 
paragraph (b)(1)(ii)(B) of this section should not be changed.
    (iv) Calculations. Calculate the micrograms of sulbactam per 
milligram of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.329

where:
Au=Area of the sulbactam peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the sulbactam peak in the chromatogram of the 
          sulbactam working standard;
Ps=Sulbactam activity in the sulbactam working standard 
          solution in micrograms per milliliter;
Cu=Milligrams of sample per milliliter of

[[Page 987]]

          sample solution; and
m=Percent moisture content of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 20 milligrams of sulbactam per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) Crystallinity. Proceed as directed in Sec. 436.203(a) of this 
chapter.
    (6) Identity. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the sulbactam working standard.
[52 FR 42290, Nov. 4, 1987; 52 FR 45281, Nov. 25, 1987, as amended at 54 
FR 47205, Nov. 13, 1989; 55 FR 11585, Mar. 29, 1990]



Sec. 455.85  Vancomycin hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Vancomycin hydrochloride is the 
hydrochloride salt of a kind of vancomycin or a mixture of two or more 
such salts. It is soluble in water and moderately soluble in dilute 
methyl alcohol. It is insoluble in higher alcohols, acetone, and ether. 
It is so purified and dried that:
    (i) It contains not less than 900 micrograms of vancomycin per 
milligram, calculated on an anhydrous basis.
    (ii) [Reserved]
    (iii) Its moisture content is not more than 5 percent.
    (iv) Its pH in an aqueous solution containing 50 milligrams per 
milliliter is not less than 2.5 and not more than 4.5.
    (v) It contains not more than 15 percent of factor A.
    (vi) It gives a positive identity test for vancomycin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, moisture, 
pH, factor A content, and identity.
    (ii) Samples required: 12 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample of approximately 30 milligrams in 
sufficient sterile distilled water to give a stock solution of 1 
milligram per milliliter (estimated). Further dilute an aliquot of the 
stock solution with 0.1M potassium phosphate buffer, pH 4.5 (solution 
4), to the reference concentration of 10 micrograms of vancomycin per 
milliliter (estimated).
    (2) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 50 milligrams per milliliter.
    (5) Identity and factor A content. Proceed as directed in 
Sec. 455.85a(b)(7).
[39 FR 19166, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 455.85a  Sterile vancomycin hydrochloride.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sterile vancomycin hydrochloride is the 
hydrochloride salt of a kind of vancomycin or a mixture of two or more 
such salts. It is soluble in water and moderately soluble in dilute 
methyl alcohol. It is insoluble in higher alcohols, acetone, and ether. 
It is so purified and dried that:
    (i) It contains not less than 900 micrograms of vancomycin per 
milligram, calculated on an anhydrous basis. If it is packaged for 
dispensing, its potency is satisfactory if it is not less than 90 
percent and not more than 115 percent of the number of milligrams of 
vancomycin that it is represented to contain.
    (ii) It is sterile.
    (iii) [Reserved]
    (iv) It is nonpyrogenic.
    (v) Its moisture content is not more than 5 percent.
    (vi) Its pH in an aqueous solution containing 50 milligrams per 
milliliter

[[Page 988]]

is not less than 2.5 and not more than 4.5.
    (vii) Its heavy metals content is not more than 30 parts per 
million.
    (viii) It contains not more than 15 percent of factor A.
    (ix) It gives a positive identity test for vancomycin.
    (2) Packaging. In addition to the requirements of Sec. 432.1 of this 
chapter, if it is packaged for dispensing, the vancomycin content of 
each immediate container is 500 milligrams of vancomycin.
    (3) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (4) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on the batch for potency, sterility, 
pyrogens, moisture, pH, heavy metals, factor A content, and identity.
    (ii) Samples required:
    (a) If the batch is packaged for repacking or for use as an 
ingredient in the manufacture of another drug:
    (1) For all tests except sterility: 12 packages, each containing 
approximately 500 milligrams.
    (2) For sterility testing: 20 packages, each containing 
approximately 300 milligrams.
    (b) If the batch is packaged for dispensing:
    (1) For all tests except sterility: A minimum of 12 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Dissolve an accurately weighed sample of approximately 30 milligrams in 
sufficient sterile distilled water to give a stock solution of 1 
milligram per milliliter; and also if it is packaged for dispensing, 
reconstitute as directed in the labeling. Then using a suitable 
hypodermic needle and syringe, remove all of the withdrawable contents 
if it is represented as a single-dose container; or if the labeling 
specifies the amount of potency in a given volume of the resultant 
preparation, remove an accurately measured representative portion from 
each container. Dilute with 0.1M potassium phosphate buffer, pH 4.5 
(solution 4), to give a stock solution of 1 milligram per milliliter. 
Further dilute an aliquot of the stock solution with solution 4 to the 
reference concentration of 10.0 micrograms of vancomycin per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use sterile distilled water in lieu of diluting fluid A.
    (3) [Reserved]
    (4) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 5 milligrams of vancomycin per milliliter.
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 50 milligrams per milliliter.
    (7) Identity and factor A content--(i) Preparation of the 
chromatogram--(a) Equipment. (1) Chromatographic paper (Whatman No. 1 
untreated filter paper).
    (2) Equipment for descending paper chromatography (Mitchell tank).
    (b) Preparations of solutions--(1) Factor A. Prepare a solution in 
distilled water to contain 1.33 milligrams of factor A per milliliter 
and further dilute with distilled water to prepare solutions containing 
0.1 and 0.2 milligram of factor A per milliliter.
    (2) Vancomycin working standard solution. Prepare a solution in 
distilled water to contain 1.33 milligrams of vancomycin per milliliter.
    (3) Known mixture of factor A and vancomycin. Prepare a solution in 
distilled water to contain 0.2 milligram of factor A and 1.13 milligrams 
of vancomycin (estimated) per milliliter.
    (4) Sample. Prepare two solutions of the sample in distilled water, 
each to contain 1.33 milligrams of vancomycin (estimated) per 
milliliter.
    (5) Solvent mixture. Mix 300 milliliters of butyl alcohol, 150 
milliliters of pyridine, and 200 milliliters of water in a large 
separatory funnel and shake well for 3 minutes. Let stand at room 
temperature. There should be no separation of layers.
    (c) Procedure. Saturate the atmosphere in the tank with vapors of 
the

[[Page 989]]

solvent mixture by placing 10 milliliters of the mixture in a trough in 
the bottom of the tank and closing tightly for 15 minutes. Prepare a 
sheet of chromatographic paper (8 inches x 8 inches) by carefully 
drawing a line of origin with a pencil 2 inches from one of the edges. 
Fold the paper along a straight line 1\1/2\ inches from the same edge of 
the paper. Starting 1 inch from the left-hand edge, establish points at 
1-inch intervals along the line of origin on which to apply the 
solutions. Using a micropipette, apply the factor A solutions, the 
vancomycin solution, the known mixture solution, and the sample 
solutions by placing 5 microliters of each on separate spots. Properly 
identify the locations of the spots but avoid unnecessary handling of 
the paper. Allow the spots to dry spontaneously. Suspend the paper in 
the chamber so that the edge nearest the fold can be conveniently 
immersed in the solvent mixture contained in the top trough. Immerse the 
paper across its entire width to a depth sufficient to assure contact 
with the solvent mixture during the entire development time. Close the 
chamber tightly and allow the chromatograph to develop at room 
temperature for 6\1/2\ to 7 hours. Remove the paper and allow it to dry 
completely.
    (ii) Development by bioautograph--(a) Preparation of test organism 
(spore suspension). The test organism is Bacillus subtilis (ATCC 6633), 
1 test organism H, prepared as described in Sec. 436.103 of 
this chapter, using the method described in paragraph (b)(2) of that 
section.
---------------------------------------------------------------------------

    \1\ Available from: American Type Culture Collection, 12301 Parklawn 
Drive, Rockville, MD 20852.
---------------------------------------------------------------------------

    (b) Preparation of plates--(1) Baselayer. Add 42 milliliters of 
medium 2 described in Sec. 436.102(b)(2) of this chapter to each Petri 
dish (25 millimeters x 150 millimeters) and allow to harden on a flat, 
level surface. To prevent condensation of excess moisture, raise the 
tops slightly while the agar hardens.
    (2) Seed layer. Melt nutrient agar medium 2 described in 
Sec. 436.102(b)(2) of this chapter. Accurately measure a sufficient 
quantity of the melted agar, cool to 48 deg. C., and add the appropriate 
quantity of the spore suspension prepared as described in paragraph 
(b)(7)(ii)(a) of this section. Swirl the flask of inoculated agar to 
obtain a homogeneous suspension. Add 8 milliliters of this inoculated 
agar to each plate, spread evenly, and allow to harden on a flat, level 
surface. For accurate results, it is necessary to obtain uniform 
distribution of the agar over the entire surface of the plates.
    (c) Assay. For each spot on the paper described in paragraph 
(b)(7)(i)(c) of this section, cut a strip 1.5 centimeters by 
approximately 14 centimeters with the center of each strip centered 
about the line of descent of the spot. Place all strips on plates with 
the aid of forceps within as short a period of time as possible. Use 
maximum spacing between strips. Insure complete contact so that the 
entire strip becomes uniformly moistened. Allow to stand for 30 minutes. 
Remove the strips and identify each strip location on the Petri dish. 
Incubate the plates for 16-18 hours at 37 deg. C. Any zone of inhibition 
corresponding to factor A in the sample must not be greater than that of 
the 0.2 milligram-per-milliliter factor A standard. Also, the two areas 
of inhibition for the sample due to the presence of factor A and 
vancomycin must compare to the corresponding two areas of inhibition of 
the known mixture in their respective distances from their origins.
    (8) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
[39 FR 19166, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 455.86  Vancomycin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Vancomycin is a tricyclic glycopeptide. 
It is a free flowing white to off-white colored powder. It is so 
purified and dried that:
    (i) It contains not less than 925 micrograms of vancomycin per 
milligram, calculated on the anhydrous basis.
    (ii) It contains not less than 92 percent vancomycin factor B and 
not more than 3 percent of any individual vancomycin related factor.
    (iii) Its moisture content is not more than 20 percent.

[[Page 990]]

    (iv) Its heavy metals content is not more than 30 parts per million.
    (v) It gives a positive identity test for vancomycin.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency, 
chromatographic purity, moisture, heavy metals, and identity.
    (ii) Samples required: 12 packages, each containing approximately 
500 milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed sample of approximately 100 milligrams in a 
100-milliliter volumetric flask and dissolve in approximately 50 
milliliters of distilled water and 1.0 milliliter of 0.1N hydrochloric 
acid. Swirl or sonicate to dissolve the sample and bring to volume with 
distilled water. Further dilute an aliquot of this solution with 0.1M 
potassium phosphate buffer, pH 4.5 (solution 4), to the reference 
concentration of 10 micrograms of vancomycin per milliliter (estimated).
    (2) Chromatographic purity. Proceed as directed in Sec. 436.366 of 
this chapter. The relative amount of vancomycin B is not less than 92 
percent, and the relative amount of any related substance is not more 
than 3 percent.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
    (5) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 0.5 percent potassium bromide disc preparation as described in 
Sec. 436.211(b)(1).
[59 FR 8400, Feb. 22, 1994]



Sec. 455.88  Rifabutin.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Rifabutin is an amorphous red-violet 
powder. It is (9S, 12E, 14S, 15R, 16S, 17R, 18R, 19R, 20S, 21S, 22E, 
24Z)-6,16,18, 20-tetrahydroxy-1'-isobutyl-14-methoxy-7, 9,15,17,19,21, 
25-heptamethylspiro[9,4-(epoxypentadeca[1,11,13]trienimino)-2H-furo[2', 
3':7,8] naphth[1,2-d]imidazole-2, 4'-piperidine]-5,10, 26-(3H, 9H)-
trione-16-acetate. It is very slightly soluble in water, sparingly 
soluble in ethanol, and soluble in chloroform and methanol. It is so 
purified and dried that:
    (i) Its potency is not less than 950 micrograms and not more than 
1,020 micrograms of rifabutin activity per milligram on an anhydrous 
basis.
    (ii) Its content for the four major related substances detected by 
high-performance liquid chromatography (HPLC) is not more than 1.0 
percent each. All other unknown related substances are not more than 0.5 
percent. The total of all related substances is not more than 3.0 
percent.
    (iii) Its moisture content is not more than 2.5 percent.
    (iv) Its N-isobutylpiperidone content is not more than 0.5 percent.
    (v) It gives a positive identity test.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for rifabutin potency, 
related substances, moisture, N-isobutylpiperidone, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research: 10 packages each containing approximately 300 
milligrams.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.216 of this chapter, using ambient temperature, an ultraviolet 
detection system operating at a wavelength of 254 # 1 
nanometers, an 11 centimeters X 4.7 millimeters (i.d.) column packed 
with microparticulate (5 to 7 micrometers in diameter) packing material 
such as octylsilane chemically bonded to porous silica (U.S. 
Pharmacopeia designation L7), a flow rate of about 1.0 milliliter per 
minute, and a manual or automatic injector capable of injecting 10 
microliters. The retention time for rifabutin is between 9 and 11 
minutes. Reagents; working standard, sample,

[[Page 991]]

and resolution solutions; system suitability requirements; and 
calculations are as follows:
    (i) Reagents--(A) Hydrochloric acid, 2N. Dilute 85 milliliters of 
hydrochloric acid (37 percent) with distilled water to 500 milliliters.
    (B) Potassium dihydrogen phosphate, 0.1M. Prepare a solution 
containing 15.4 grams of potassium dihydrogen phosphate monohydrate 
(potassium phosphate monobasic) per liter of distilled water.
    (C) Sodium hydroxide, 2N. Dissolve 8 grams of sodium hydroxide 
pellets in 100 milliliters of distilled water.
    (D) Mobile phase. Acetonitrile:phosphate buffer, pH 6.5, 50:50. Mix 
equal quantities of acetonitrile and 0.1M potassium dihydrogen phosphate 
and adjust to an apparent pH of 6.5 # 0.1 by dropwise 
addition of 2N sodium hydroxide. Filter through a suitable filter 
capable of removing particulate matter 0.5 micron in diameter and degas 
it just prior to its introduction into the chromatograph. Slight 
adjustments of the mobile phase components ratio may be made in order to 
meet the system suitability requirements described in the system 
suitability tests in paragraph (b)(1)(iii) of this section.
    (ii) Preparation of working standard, sample, and resolution test 
solution--(A) Working standard solution. Accurately weigh approximately 
25 milligrams of the rifabutin working reference standard into a 50-
milliliter volumetric flask. Add 5 milliliters of acetonitrile. Dissolve 
and dilute to volume with mobile phase and mix to obtain a solution 
having a known concentration of about 0.5 milligram of rifabutin per 
milliliter.
    (B) Sample solution. Accurately weigh approximately 25 milligrams of 
sample into a 50-milliliter volumetric flask. Add 5 milliliters of 
acetonitrile. Dissolve and dilute to volume with mobile phase and mix to 
obtain a solution containing 0.5 milligram of rifabutin per milliliter 
(estimated).
    (C) Resolution test solution. Dissolve approximately 10 milligrams 
of rifabutin in 2 milliliters of methanol and add 1 milliliter of 2N 
sodium hydroxide. Allow to stand for 3 to 4 minutes and then add 1 
milliliter of 2N hydrochloric acid. Mix and dilute to 50 milliliters 
with mobile phase. Store aliquots of this solution in the frozen state 
for future use.
    (iii) System suitability requirements. Using the apparatus and 
conditions described in this section, test the chromatographic system by 
injecting the resolution test solution. The chromatogram shows one major 
degradation peak and two minor degradation peaks eluting at relative 
retention times (RRT) of 0.5-0.6, 0.65-0.75, and 0.8-0.9, respectively, 
followed by the rifabutin peak.
    (A) Asymmetry factor. The asymmetry factor (AS) is 
satisfactory if it is not less than 1.0 and not more than 4.0 forther if 
a butin peak.
    (B) Efficiency of the column. The absolute efficiency 
(hr) is satisfactory if it is not more than 11 for the 
rifabutin peak, equivalent to 2,000 theoretical plates for a 11-
centimeter column of 5-micrometer particles.
    (C) Resolution factor. The resolution factor (R) between the peak 
for rifabutin and its closest eluting degradation product (generated in 
situ as described in paragraph (b)(1)(iii) of this section and eluting 
at RRT of 0.8-0.9) is satisfactory if it is not less than 1.3.
    (D) Coefficient of variation (relative standard deviation). The 
coefficient of variation (SR in percent of 5 replicate 
injections of the rifabutin working standard solution) is satisfactory 
if it is not more than 2.0 percent. If the system suitability parameters 
have been met, then proceed as described in Sec. 436.216(b) of this 
chapter.
    (iv) Calculations. Calculate the micrograms of rifabutin per 
milligram of sample on an anhydrous basis as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.330

where:
AU = Area of the rifabutin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
AS = Area of the rifabutin peak in the chromatogram of the 
          rifabutin working standard;
PS = Rifabutin activity in the rifabutin working standard 
          solution in micrograms per milliliter;
CU = Milligrams of sample per milliliter of

[[Page 992]]

          sample solution; and
m = Percent moisture content of the sample.

    (2) Related substances. Proceed as directed in paragraph (b)(1) of 
this section for potency using the sample prepared as described in 
paragraph (b)(1)(ii)(B) of this section and calculating the amounts of 
related substances as follows.
    (i) Calculations. Calculate the percentage of related substances as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.331

where:
Ai = Area of the individual related substance peak;
A = The sum of areas of all peaks minus the area due to the rifabutin 
          peak and solvent front peak; and
At = The sum of areas of all peaks in the chromatogram 
          excluding the solvent peak.

    (ii) [Reserved]
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) N-Isobutylpiperidone. Proceed as directed in Sec. 436.369 of 
this chapter.
    (5) Identity. (i) Proceed as directed in Sec. 436.211 of this 
chapter, using the sample preparation method described in paragraph 
(b)(1) of that section using a 1 to 2 percent mixture in potassium 
bromide.
    (ii) The identity of rifabutin is confirmed by the qualitative 
comparison of the HPLC of the sample to the rifabutin working standard 
as directed in paragraph (b)(1) of this section.
[59 FR 40807, Aug. 10, 1994; 59 FR 46479, Sept. 8, 1994]



Sec. 455.90a  Sterile vidarabine monohydrate.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Vidarabine monohydrate is the monohydrate 
form of 9- - D - arabinofuranosyl - 9H - purin - 6-amine. It is 
a white to off-white powder. It is so purified and dried that:
    (i) Its vidarabine content is not less than 845 micrograms and not 
more than 985 micrograms of vidarabine per milligram.
    (ii) It is sterile.
    (iii) [Reserved]
    (iv) Its loss on drying is not less than 5 percent and not more than 
7 percent.
    (v) Its specific rotation in dimethylformamide at 25 deg. C is --
60.5 deg.plus-minus4.5 deg..
    (vi) It passes the identity test for vidarabine.
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5(b) of this chapter, this drug shall be labeled 
``vidarabine''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for vidarabine content, 
sterility, loss on drying, specific rotation, and identity.
    (ii) Samples required:
    (a) For all tests except sterility: 10 packages, each containing 
approximately 500 milligrams.
    (b) For sterility testing: 20 packages, each containing 
approximately 200 milligrams.
    (b) Tests and methods of assay--(1) Vidarabine content. Proceed as 
directed in Sec. 436.325 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section, except 
use 100 milligrams in lieu of 300 milligrams.
    (3) [Reserved]
    (4) Loss on drying. Proceed as directed in Sec. 436.200(e) of this 
chapter.
    (5) Specific rotation. Using a solution containing 10 milligrams of 
vidarabine per milliliter in dimethylformamide and a polarimeter tube 
1.0 decimeter in length, proceed as directed in Sec. 436.210 of this 
chapter, except determine the specific rotation at 365 nanometers.
    (6) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 0.5 percent potassium bromide disc prepared as described in 
paragraph (b)(1) of that section.
[42 FR 44224, Sept. 2, 1977; 43 FR 9802, Mar. 10, 1978, as amended at 44 
FR 30334, May 25, 1979; 50 FR 19921, May 13, 1985]

[[Page 993]]



                      Subpart B--Oral Dosage Forms



Sec. 455.110  Chloramphenicol capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chloramphenicol capsules are composed of 
chloramphenicol with or without one or more suitable and harmless 
diluents and lubricants. Each capsule contains 50, 100, or 250 
milligrams of chloramphenicol. Its potency is satisfactory if it is not 
less than 90 percent and not more than 120 percent of the number of 
milligrams of chloramphenicol that it is represented to contain. The 
chloramphenicol used conforms to the standards prescribed by 
Sec. 455.10(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The chloramphenicol used in making the batch for potency, pH, 
specific rotation, melting range, absorptivity, and crystallinity.
    (b) The batch for potency.
    (ii) Samples required:
    (a) The chloramphenicol used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay; potency. Use either of the following 
methods; however, the results obtained from the microbiological 
turbidimetric assay shall be conclusive.
    (1) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing 100 milliliters of 95 percent ethyl alcohol. 
Blend for 2 minutes. Then add 400 milliliters of distilled water and 
blend again for 2 minutes. Remove an aliquot and further dilute with 
distilled water to the reference concentration of 2.5 micrograms of 
chloramphenicol per milliliter (estimated).
    (2) Spectrophotometric assay--(i) Preparation of working standard 
solution. Dissolve approximately 50 milligrams of the working standard 
in 100 milliliters of distilled water. Warm if necessary to hasten 
dissolution. Transfer 10 milliliters into a 250-milliliter volumetric 
flask and fill to volume with distilled water.
    (ii) Procedure. Place the contents of 10 capsules into a 250-
milliliter volumetric flask. Add 50 milliliters of pure methyl alcohol 
to the flask and shake for at least 1 minute. Fill to volume with 
distilled water and mix thoroughly. Withdraw an aliquot and dilute with 
sufficient distilled water to give a concentration of 20 micrograms per 
milliliter. Using a suitable spectrophotometer equipped with a 1.0-
centimeter cell and distilled water as the blank, determine the 
absorbance of the working standard and sample solutions at 278 
nanometers. Calculate the potency as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.332

[39 FR 19149, May 30, 1974, as amended at 48 FR 3960, Jan. 28, 1983; 50 
FR 19921, May 13, 1985]



Sec. 455.111  Chloramphenicol palmitate oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chloramphenicol palmitate oral suspension 
is chloramphenicol palmitate and one or more suitable and harmless 
buffer substances, suspending agents, preservatives, colorings, and 
flavorings suspended in a suitable and harmless vehicle. Each milliliter 
contains chloramphenicol palmitate equivalent to 30.0 milligrams of 
chloramphenicol. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
chloramphenicol that it is represented to contain. Its pH is not less 
than 4.5 nor more than 7.0. Its content of polymorph A crystals does not 
exceed 10 percent. The chloramphenicol palmitate used conforms to the 
standards prescribed by Sec. 455.11(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.

[[Page 994]]

    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The chloramphenicol palmitate used in making the batch for 
chloramphenicol content, melting range, specific rotation, and 
crystallinity.
    (b) The batch for chloramphenicol content, pH, and content of 
polymorph A crystals.
    (ii) Samples required:
    (a) The chloramphenicol palmitate used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Chloramphenicol content (high-
pressure liquid chromatography). Proceed as directed in Sec. 436.335 of 
this chapter, except prepare the sample solution and calculate the 
chloramphenicol content as follows:
    (i) Preparation of sample solution. Transfer a portion of the sample 
equivalent to 150 milligrams of chloramphenicol into a 200-milliliter 
volumetric flask. Add 100 milliliters of methanol and 4 milliliters of 
glacial acetic acid. Shake and dilute to volume with methanol. Filter 
the solution through a glass fiber filter or equivalent that is capable 
of removing particulate contamination to 1 micron in diameter.
    (ii) Calculations. Calculate the chloramphenicol content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.333
    
where:

A=Area of the chloramphenicol palmitate sample peak (at a retention time 
equal to that observed for the standard);
B=Area of the working standard peak;
Ws=Weight of standard in milligrams;
f=Micrograms of chloramphenicol activity per milligram of 
chloramphenicol palmitate working standard; and
V=Volume of sample in milliliters.

    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted sample.
    (3) Content of polymorph A crystal.--(i) Preparation of standards--
(a) Standard containing 20 percent of polymorph A. Prepare a thoroughly 
mixed, dry powder composed by weight of 1 part of polymorph A crystals 
of chloramphenicol palmitate and 4 parts of nonpolymorph A crystals of 
chloramphenicol palmitate.
    (b) Standard containing 10 percent of polymorph A. Prepare a 
thoroughly mixed, dry powder composed by weight of 1 part of polymorph A 
crystals of chloramphenicol palmitate and 9 parts of nonpolymorph A 
crystals of chloramphenicol palmitate.
    (ii) Preparation of sample. Place 20 milliliters of thoroughly mixed 
oral suspension into a 50-milliliter centrifuge tube. Add 20 milliliters 
of water and mix. Centrifuge for 10 to 15 minutes at a speed not less 
than 18,000 revolutions per minute. Decant the supernatant liquid. Wash 
the residue as follows: Add 2 milliliters of water to the residue, mix 
to make paste, add 18 milliliters of water, and mix thoroughly. 
Centrifuge, decant the supernatant liquid, and wash the residue two more 
times. Remove the washed residue from the centrifuge tube and dry it at 
least 14 hours in a vacuum desiccator at room temperature.
    (iii) Procedure. Weigh 150 to 200 milligrams of liquid petrolatum 
into an agate mortar and add about 100 milligrams of standard or sample. 
Mix with a small spatula and then mull thoroughly with a pestle until a 
uniform consistency is obtained. Adjust a suitable infrared 
spectrophotometer so that 100 percent transmittance is recorded over the 
range of 11.0 to 13.0 microns. Use two rock salt plates as an absorption 
cell. Place a small drop of the mull in the center of one of the plates. 
Gently put the other plate on the mull and slowly squeeze the plates 
together to spread the mull uniformly. Clamp the two plates firmly 
together in a metal cell holder. Examine the assembled cell by holding 
it up to the light. It should appear smooth and free of any air bubbles 
and when placed in the instrument it should give a percent transmittance 
of 20 to 30 percent at 12.3 microns. Place the cell in the infrared 
spectrophotometer and record the absorption spectrum from 11.0 to 13.0 
microns.
    (iv) Treatment of spectra--(a) Standard containing 20 percent of 
polymorph A.

[[Page 995]]

Determine by inspection of the recorded spectrum the exact wavelengths 
of minimum absorption at approximately 11.3 and 12.65 microns. Also 
determine by inspection the exact wavelengths of maximum absorption at 
approximately 11.65 and 11.86 microns. In the following subdivision, 
references to these four nominal wavelengths are to the exact 
wavelengths observed on the particular instrument being used.
    (b) Standard containing 10 percent of polymorph A. Draw a straight 
baseline between the minima occurring at 11.3 and 12.65 microns. Draw 
straight lines at 11.65 and 11.86 microns intersecting both the recorded 
spectrum and the baseline. Obtain the corrected absorbances at 11.65 and 
11.86 microns and calculate the absorbance ratios as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.334

where:
S11.65=Absorbance value of recorded spectrum at 11.65 
microns;
B11.65=Absorbance value at point of intersection of the 
11.65-micron line with the baseline;
S11.86=Absorbance value of recorded spectrum at 11.86 
microns;
B11.86=Absorbance value at point of intersection of the 
11.86-micron line with the baseline.

    (c) Sample. Proceed as described in paragraph (b)(3)(iv)(b) of this 
section.
    (v) Calculation. The absorbance ratio of the sample must be greater 
than the absorbance ratio of the standard containing 10 percent of 
polymorph A.
[39 FR 19166, May 30, 1974, as amended at 49 FR 6093, Feb. 17, 1984; 50 
FR 19921, May 13, 1985]



Sec. 455.120  Cycloserine capsules.

    (a) Requirements for certification--(1) Standards of identity, 
quality, and purity. Cycloserine capsules are capsules composed of 
crystalline cycloserine, with or without one or more suitable and 
harmless buffer substances, diluents, binders, and lubricants. Each 
capsule contains 250 milligrams of cycloserine. Its potency is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of cycloserine that it is 
represented to contain. The loss on drying is not more than 1.0 percent. 
The cycloserine used conforms to the standards prescribed by 
Sec. 455.20(a)(1).
    (2) Labeling. In addition to the labeling prescribed by Sec. 432.5 
of this chapter, the labeling of each package shall bear a warning to 
the effect that the drug is to be used in patients with tuberculosis who 
fail to respond to treatment with isoniazid, streptomycin, 
paraaminosalicylic acid, viomycin, pyrazinamide, or combinations of 
these drugs, and that the drug may cause serious reactions such as 
convulsive seizures and mental disturbances.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) Cycloserine used in making the batch for potency, loss on 
drying, pH, residue on ignition, crystallinity, and identity.
    (b) The batch for cycloserine content and loss on drying.
    (ii) Samples required:
    (a) Cycloserine used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch: Minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Using the cycloserine 
working standard as the standard of comparison, assay for potency by 
either of the following methods; however, the results obtained from the 
microbiological turbidimetric assay shall be conclusive.
    (i) Chemical colorimetric assay--(a) Reagents. (1) Acetic acid--1.0N 
solution.
    (2) Sodium hydroxide--4.0N and 0.1N solutions.
    (3) Sodium nitroprusside--4.0 percent solution: Dissolve 4.0 grams 
in sufficient distilled water to make 100.0 milliliters. Mix well. Store 
in amber bottle.
    (4) Oxidized nitroprusside reagent--Mix equal parts of the 4.0 
percent sodium nitroprusside solution and 4.0N sodium hydroxide, and let 
stand for 1 hour before using. Prepare daily and store in amber bottle.
    (5) Cycloserine standard solution--dilute an appropriate-sized 
aliquot of the

[[Page 996]]

stock standard solution, prepared as directed in 
Sec. 455.20(b)(1)(i)(a), in 0.1N sodium hydroxide to obtain a working 
standard solution containing 100 micrograms of cycloserine per 
milliliter.
    (b) Procedure. Transfer the contents of 10 capsules into a 1,000-
milliliter volumetric flask. Add 0.1N sodium hydroxide to dissolve the 
sample, and add sufficient 0.1N sodium hydroxide to measure 1,000 
milliliters. Mix well and filter. Dilute an alliquot of the filtrate 
with sufficient 0.1N sodium hydroxide to give a concentration of 0.1 
milligram per milliliter (estimated) and mix well. Pipette exactly 1.0 
milliliter of the working standard solution and 1.0 milliliter of the 
sample solution into separate test tubes. Add exactly 3.0 milliliters of 
1.0N acetic acid and exactly 1.0 milliliter of oxidized nitroprusside 
reagent to each of the test tubes; then mix thoroughly. Allow the tubes 
to stand at room temperature for 10 to 15 minutes, in order that maximum 
color intensity may develop. Using a reagent blank, determine the 
absorbance of the solutions at 625 nanometers in a suitable spectropho-
tometer.
    Calculation:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.335
    
    (ii) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules in a high-speed glass blender 
with sufficient sterile distilled water to give a stock solution of 
convenient concentration. Blend 3 to 5 minutes. Further dilute the stock 
solution with sterile distilled water to the reference concentration of 
50 micrograms of cycloserine per milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[39 FR 19166, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 455.150  Calcium novobiocin oral suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Calcium novobiocin oral suspension is a 
suspension containing calcium novobiocin and one or more suitable and 
harmless diluents, preservatives, suspending agents, surfactants, 
flavorings, and colorings in purified water. Each milliliter contains 25 
milligrams of novobiocin. Its potency is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the number of 
milligrams of novobiocin that it is represented to contain. The pH is 
not less than 6.0 and not more than 7.5. The calcium novobiocin used 
conforms to the standards prescribed by Sec. 455.50(a)(1) (i), (iv), 
(v), (vi), and (vii). If sodium novobiocin is reacted with a suitable 
calcium salt to form calcium novobiocin, the sodium novobiocin used 
conforms to the standards prescribed by Sec. 455.51(a)(1) (i), (iv), 
(v), (vi), (vii), and (viii).
    (2) Labeling. It shall be labeled in accordance with Sec. 432.5 of 
this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The calcium novobiocin used in making the batch for potency, pH, 
crystallinity, identity, and specific rotation. If sodium novobiocin is 
used in making the batch: Potency, pH, residue on ignition, specific 
rotation, identity, and crystallinity.
    (b) The batch for potency and pH.
    (ii) Samples required:
    (a) The calcium novobiocin or the sodium novobiocin used in making 
the batch: 10 packages, each containing approximately 500 milligrams.
    (b) The batch: Minimum of 5 immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Remove a representative sample of the sirup with a suitable

[[Page 997]]

syringe and place into a high-speed glass blender with sufficient 
absolute ethyl alcohol to give a concentration (estimated) of 1,000 
micrograms per milliliter. Blend for 3 to 5 minutes. Further dilute with 
10 percent potassium phosphate buffer, pH 1.0 (solution 6), to the 
reference concentration of 0.5 microgram of novobiocin per milliliter 
(estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted suspension.
[39 FR 19166, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 455.151  Sodium novobiocin oral dosage forms.



Sec. 455.151a  Sodium novobiocin tablets.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sodium novobiocin tablets are tablets 
that contain sodium novobiocin, with or without one or more suitable and 
harmless buffer substances, diluents, binders, and lubricants. Each 
tablet contains 125 milligrams or 250 milligrams of novobiocin. The 125-
milligram tablet contains 375 milligrams of sulfamethizole. Its potency 
is satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of novobiocin that it is represented 
to contain. Its loss on drying is not more than 3 percent. The tablets 
disintegrate within 1 hour. The sodium novobiocin used conforms to the 
standards prescribed by Sec. 455.51(a)(1).
    (2) Labeling. It shall be labeled in accordance with Sec. 432.5 of 
this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) Sodium novobiocin used in making the batch for potency, loss on 
drying, pH, residue on ignition, specific rotation, identity, and 
crystallinity.
    (b) The batch for potency, loss on drying, disintegration time.
    (ii) Samples required:
    (a) Sodium novobiocin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch: A minimum of 36 tablets.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Blend a representative number of tablets in a high-speed glass blender 
with sufficient 0.1M potassium phosphate buffer, pH 8.0 (solution 3), to 
give a stock solution of convenient concentration. Further dilute the 
stock solution with 10 percent potassium phosphate buffer, pH 6.0 
(solution 6), to the reference concentration of 0.5 microgram of 
novobiocin per milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (3) Disintegration time. Proceed as directed in Sec. 436.212 of this 
chapter, using the method described in paragraph (e)(1) of that section.
[39 FR 19166, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 455.151b  Sodium novobiocin capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sodium novobiocin capsules are gelatin 
capsules containing sodium novobiocin with a suitable and harmless 
filler and with or without a binder and a lubricant. Each capsule 
contains 100 milligrams or 250 milligrams of novobiocin. Its potency is 
satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of novobiocin that it is represented 
to contain. The loss on drying is not more than 6.0 percent. The sodium 
novobiocin used conforms to the standards prescribed by 
Sec. 455.51(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The sodium novobiocin used in making the batch for potency, loss 
on drying, pH, residue on ignition, specific rotation, crystallinity, 
and identity.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:
    (a) The sodium novobiocin used in making the capsules: 10 packages, 
each

[[Page 998]]

containing approximately 500 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules in a high-speed glass blender 
with 1.0 milliliter of polysorbate 80 and sufficient 0.1M potassium 
phosphate buffer, pH 8.0 (solution 3), to give a stock solution of 
convenient concentration. Blend 3 to 5 minutes. Further dilute with 10 
percent potassium phosphate buffer, pH 6.0 (solution 6), to the 
reference concentration of 0.5 microgram of novobiocin per milliliter 
(estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[39 FR 19166, May 30, 1974, as amended at 50 FR 19921, May 13, 1985]



Sec. 455.170  Rifampin oral dosage forms.



Sec. 455.170a  Rifampin capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Rifampin capsules are gelatin capsules 
containing rifampin with a suitable and harmless filler and with or 
without binders, lubricants, and stabilizers. Each sample contains 150 
milligrams or 300 milligrams of rifampin. Its potency is satisfactory if 
it is not less than 90 percent and not more than 130 percent of the 
number of milligrams of rifampin that it is represented to contain. Its 
loss on drying is not more than 3.0 percent. The rifampin used conforms 
to the standards prescribed by Sec. 455.70(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The rifampin used in making the batch for potency, loss on 
drying, pH, absorptivity, identity, and crystallinity.
    (b) The batch for potency and loss on drying.
    (ii) Samples required:
    (a) The rifampin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed glass 
blender jar containing 200 milliliters of methyl alcohol and blend for 3 
minutes. Add 300 milliliters of 1 percent potassium phosphate buffer, pH 
6.0 (solution 1), and blend for 3 to 5 minutes. Remove an aliquot and 
further dilute with solution 1 to the reference concentration of 5.0 
micrograms of rifampin per milliliter (estimated).
    (2) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[39 FR 19166, May 30, 1974. Redesignated at 40 FR 53997, Nov. 20, 1975, 
and amended at 46 FR 46314, Sept. 18, 1981; 50 FR 19921, May 13, 1985]



Sec. 455.170b  Rifampin-isoniazid capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Rifampin-isoniazid capsules contain 
rifampin and isoniazid with a suitable and harmless filler and with or 
without binders, lubricants, and stabilizers in a gelatin capsule. Each 
capsule contains 300 milligrams of rifampin and 150 milligrams of 
isoniazid. Its rifampin content is satisfactory if it is not less than 
90 percent and not more than 130 percent of the number of milligrams of 
rifampin that it is represented to contain. Its isoniazid content is 
satisfactory if it is not less than 90 percent and not more than 110 
percent of the number of milligrams of isoniazid that it is represented 
to contain. Its loss on drying is not more than 3.0 percent. The 
rifampin used conforms to the standards prescribed by Sec. 455.70(a)(1). 
The isoniazid used conforms to the standards prescribed by the U.S.P.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:

[[Page 999]]

    (a) The rifampin used in making the batch for potency, loss on 
drying, pH, absorptivity, identity, and crystallinity.
    (b) The isoniazid used in making the batch for all U.S.P. 
specifications.
    (c) The batch for rifampin content, isoniazid content, and loss on 
drying.
    (ii) Samples required:
    (a) The rifampin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 36 capsules.
    (b) Tests and methods of assay--(1) Rifampin content. Proceed as 
directed in Sec. 436.105 of this chapter, preparing the sample for assay 
as follows: Place a representative number of capsules into a high-speed 
glass blender jar containing 200 milliliters of methyl alcohol and blend 
for 3 minutes. Add 300 milliliters of 1 percent potassium phosphate 
buffer, pH 6.0 (solution 1), and blend for 3 to 5 minutes. Remove an 
aliquot and further dilute with solution 1 to the reference 
concentration of 5.0 micrograms of rifampin per milliliter (estimated).
    (2) Isoniazid content--(i) Equipment--(a) Electronic voltmeter. A 
vacuum tube voltmeter or pH meter capable of measuring potentials from 0 
to 1,400 millivolts.
    (b) Platinum electrodes. Use twin platinum electrodes.
    (c) Constant current potential source. Polarize the platinum 
electrodes by means of a battery and a suitable resistance in series 
with the electrodes, or by a stable electronic power supply, so that the 
current flow is about 2.5 microamperes.
    (d) Titration vessel. Use a 100-milliliter beaker.
    (ii) Reagents--(a) Concentrated hydrochloric acid, reagent grade.
    (b) 0.1N Bromine solution. Dissolve 3.0 grams of potassium bromate 
and 15.0 grams of potassium bromide in sufficient water to make 1 liter. 
Preserve in dark amber-colored, glass-stoppered bottles.
    (c) 1.0N Potassium iodide. Dissolve 16.5 grams of potassium iodide 
in 100 milliliters of water.
    (d) Starch iodide paste, T.S. (U.S.P.).
    (e) 0.1N Sodium thiosulfate (U.S.P.).
    (f) 0.1N Hydrochloric acid.
    (g) Chloroform, reagent grade.
    (iii) Standardization of 0.1N bromine solution. Measure accurately 
about 25 milliliters of the bromine solution into a 500-milliliter 
iodine flask and dilute with 120 milliliters of water. Add 5 milliliters 
of hydrochloric acid, insert the stopper in the flask, and shake it 
gently. Then add 5 milliliters of potassium iodide T.S., insert the 
stopper, shake the mixture, and allow it to stand for 5 minutes. Titrate 
the liberated iodine with standard 0.1N sodium thiosulfate U.S.P., 
adding starch iodide paste T.S./U.S.P. as the endpoint is approached. 
Calculate the normality of the bromine solution.
    (iv) Preparation of sample solution. Empty the contents of not less 
than 10 capsules into a tared weighing bottle. Mix and weigh the powder. 
Calculate the average capsule weight content and accurately weigh a 
sample equivalent to approximately 100 milligrams of isoniazid. Transfer 
the sample to a 125-milliliter separatory funnel. Add 20 milliliters of 
0.1N hydrochloric acid and shake well. Extract the acidic solution with 
six 25-milliliter portions of chloroform, combining any interfacial 
emulsion with the aqueous phase throughout the extraction procedure. 
Discard the chloroform extracts. Quantitatively transfer the acidic 
aqueous layer to a 100-milliliter volumetric flask and dilute to volume 
with 0.1N hydrochloric acid.
    (v) Titration procedure. Pipet 25 milliliters of the sample solution 
into the titration vessel and add 10 milliliters of concentrated 
hydrochloric acid. Adjust the volume to approximately 50 milliliters 
with water. Titrate potentiometrically at constant current with 0.1N 
bromine solution to a dead stop endpoint. Calculate the isoniazid 
content for the sample used and determine the isoniazid content for the 
average capsule weight as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.336

where:
V=Volume in milliliters of 0.1N bromine solution used to titrate the 
sample;
N=Normality of bromine solution;
W=Average capsule weight content in milligrams;

[[Page 1000]]

S=Weight of sample in milligrams.

    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
[40 FR 53997, Nov. 20, 1975, as amended at 50 FR 19921, May 13, 1985]



Sec. 455.185  Vancomycin hydrochloride oral dosage forms.



Sec. 455.185a  Vancomycin hydrochloride for oral solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Vancomycin hydrochloride for oral 
solution is vancomycin hydrochloride packaged in a suitable dispensing 
container. It may contain a suitable stabilizing agent. Its potency is 
satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of grams of vancomycin that it is represented to 
contain. Its moisture content is not more than 5 percent. When 
reconstituted as directed in the labeling, its pH is not less than 2.5 
and not more than 4.5. The vancomycin hydrochloride used conforms to the 
standards prescribed by Sec. 455.85.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assay on:
    (a) The vancomycin hydrochloride used in making the batch for 
potency, moisture, pH, factor A content, and identity.
    (b) The batch for potency, moisture, and pH.
    (ii) Samples required:
    (a) The vancomycin hydrochloride used in making the batch: 12 
packages, each containing approximately 500 milligrams.
    (b) The batch: A minimum of six immediate containers.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Empty the contents into an accurately measured volume of distilled water 
as directed in the labeling of the drug. Further dilute an aliquot with 
0.1M passium phosphate buffer, pH 4.5 (solution 4), to the reference 
concentration of 10 micrograms of vancomycin per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
[39 FR 19166, May 30, 1974, as amended at 50 FR 19921, May 13, 1985. 
Redesignated at 51 FR 22072, June 18, 1986, and amended at 59 FR 8399, 
Feb. 22, 1994]



Sec. 455.185b  Vancomycin hydrochloride capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Vancomycin hydrochloride capsules contain 
vancomycin hydrochloride dispersed in polyethylene glycol. Each capsule 
contains either 125 milligrams or 250 milligrams of vancomycin. Its 
potency is satisfactory if it is not less than 90 percent and not more 
than 115 percent of the number of milligrams of vancomycin that it is 
represented to contain. Its moisture is not more than 8 percent. It 
passes the dissolution test. The vancomycin hydrochloride used conforms 
to the standards prescribed by Sec. 455.85(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The vancomycin hydrochloride used in making the batch for 
potency, moisture, pH, factor A content, and identity.
    (b) The batch for potency, moisture, and dissolution.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The vancomycin hydrochloride used in making the batch: 12 
packages, each containing approximately 500 milligrams.
    (b) The batch: A minimum of 100 capsules.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Place a representative number of capsules into a high-speed

[[Page 1001]]

glass blender jar with sufficient distilled water to obtain a stock 
solution of convenient concentration. Blend for 3 to 5 minutes. Further 
dilute an aliquot of the stock solution with 0.1M potassium phosphate 
buffer, pH 4.5 (solution 4) to the reference concentration of 10 
micrograms of vancomycin per milliliter (estimated).
    (2) Moisture. Proceed as directed in Sec. 436.201 of this chapter, 
using the titration procedure described in paragraph (e)(1) of that 
section, except:
    (i) Remove gelatin coating before grinding the capsules; and
    (ii) Use solvent C in lieu of solvent A.
    (3) Dissolution. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity Q (the amount of vancomycin dissolved) is 85 
percent within 45 minutes.
[51 FR 22072, June 18, 1986, as amended at 55 FR 11585, Mar. 29, 1990]



Sec. 455.188  Rifabutin capsules.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Rifabutin capsules are gelatin capsules 
containing rifabutin with a suitable and harmless filler and with or 
without binders, lubricants, and stabilizers. Each capsule contains 
rifabutin equivalent to 150 milligrams of rifabutin. Its rifabutin 
content is satisfactory if it is not less than 90 percent and not more 
than 110 percent of the number of milligrams of rifabutin that it is 
represented to contain. Its content of the four major related substances 
detected by high-performance liquid chromatography (HPLC) is not more 
than 1.0 percent each. All other unknown related substances are not more 
than 0.5 percent. The total of all related substances is not more than 
4.5 percent. It passes the dissolution test if the quantity (Q) 
dissolved is 75 percent at 45 minutes. It passes the identity test. The 
rifabutin used conforms to the standards prescribed by 
Sec. 455.88(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The rifabutin used in making the batch for potency, related 
substances, moisture, N-isobutylpiperidone, and identity.
    (B) The batch for content, related substances, dissolution, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The rifabutin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (B) The batch: A minimum of 30 capsules.
    (b) Tests and methods of assay--(1) Rifabutin content. Proceed as 
directed in Sec. 455.88(b)(1), preparing the sample solution and 
calculating the rifabutin content as follows:
    (i) Preparation of sample solution. Empty 20 capsules, collecting 
the contents quantitatively. Weigh the powder and determine the average 
capsule fill weight. Mix the powder and accurately weigh a portion 
containing the equivalent of about 25 milligrams of rifabutin into a 50-
milliliter volumetric flask. Add 5 milliliters of acetonitrile. Dilute 
to volume with mobile phase and mix to yield a solution containing 0.5 
milligram of rifabutin per milliliter (estimated). Filter through a 
suitable filter capable of removing particulate matter 0.5 micron in 
diameter prior to injection into the chromatographic system.
    (ii) Calculations. Calculate the rifabutin content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.337
    
where:
AU = Area of the rifabutin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
AS = Area of the rifabutin peak in the chromatogram of the 
          rifabutin working standard;
CS = Milligrams of rifabutin working standard per milliliter 
          of standard solution;
CU = Milligrams of sample per milliliter of sample solution;
PS = Rifabutin activity in the rifabutin working standard 
          solution in micrograms per milliliter; and
Wa = Average capsule fill weight in milligrams.


[[Page 1002]]


    (2) Related substances. Proceed as directed in paragraph (b)(1) of 
this section for rifabutin content using the sample prepared as 
described in paragraph (b)(1)(i) of this section and calculating the 
amounts of related substances as follows.
    (i) Calculations. Calculate the percentage of related substances as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.338

where:
Ai = Area of the individual related substance peak;
A = The sum of areas of all peaks minus the area due to the rifabutin 
          peak and solvent front peak; and
At = The sum of areas of all peaks in the chromatogram 
          excluding the solvent peak.

    (ii) [Reserved]
    (3) Dissolution test. Proceed as directed in Sec. 436.215 of this 
chapter. The quantity (Q) (the amount of rifabutin activity dissolved) 
is 75 percent within 45 minutes.
    (4) Identity. (i) The retention time of the rifabutin response in 
the HPLC procedure described in paragraph (b)(1) of this section as 
applied to the sample solution compares qualitatively to that of the 
rifabutin reference standard.
    (ii) The identity of rifabutin capsules is also confirmed by the 
spectrophotometric identity test described in Sec. 436.370 of this 
chapter.
[59 FR 40808, Aug. 10, 1994]



                   Subpart C--Injectable Dosage Forms



Sec. 455.204  Aztreonam injectable dosage forms.



Sec. 455.204a  Aztreonam for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Aztreonam for injection is a dry mixture 
of aztreonam and arginine. Its potency is satisfactory if each milligram 
of aztreonam for injection contains not less than 900 micrograms and not 
more than 1,050 micrograms of aztreonam when corrected for arginine 
content and moisture content. Its aztreonam immediate container fill 
(content) is satisfactory if it is not less than 90 percent and not more 
than 120 percent of the number of milligrams of aztreonam that it is 
represented to contain. It is sterile. It is nonpyrogenic. Its moisture 
content is not more than 2.0 percent. Its pH in an aqueous solution 
containing 100 milligrams of aztreonam per milliliter is not less than 
4.5 and not more than 7.5. The aztreonam used conforms to the standards 
prescribed by Sec. 455.4a(a)(1), except if the aztreonam for injection 
is manufactured by lyophilization, in which case the aztreonam need not 
be sterile.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The aztreonam used in making the batch for potency, sterility, 
pyrogens, moisture, residue on ignition, heavy metals, and identity. If 
the aztreonam for injection is made by lyophilization, the aztreonam 
need not be tested for sterility.
    (b) The batch for aztreonam potency, aztreonam content, sterility, 
pyrogens. moisture. and pH.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (a) The aztreonam used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency and content. Determine 
both micrograms of aztreonam per milligram of sample and milligrams of 
aztreonam per container. Proceed as

[[Page 1003]]

directed in Sec. 436.361 of this chapter, except in addition to the 
column described in paragraph (a)(4) of that section, use a 5- to 50-
centimeter saturator column having an inside diameter of 2 to 4.6 
millimeters and packed with approximately 37 micrometer silica; and use 
the resolution test solution to determine resolution in lieu of the 
working standard solution. Perform the assay at ambient temperature, 
using an ultraviolet detection system operating at a wavelength of 206 
nanometers, and a column packed with Chromegabond Diol (dihydroxypropane 
chemically bonded to porous silica), 5 to 10 micrometers or equivalent. 
Mobile phase, working standard solution, sample solution, resolution 
test solution, system suitability requirements, and calculations are as 
follows:
    (i) Mobile phase. Acetonitrile:0.01M ammonium phosphate, pH 2.0. 
Transfer 1.15 grams of ammonium phosphate monobasic to a 1-liter 
volumetric flask. Add about 800 milliliters of distilled water and 
sonicate to aid dissolution. Adjust the solution to pH 2.0 with o-
phosphoric acid, 85 percent. Dilute the solution to volume with 
distilled water and mix well. Transfer about 250 milliliters of this 
solution and 750 milliliters of acetonitrile to a suitably sized 
container and mix well.
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(a) Working standard solution. Transfer approximately 25 
milligrams each of the aztreonam working standard and the arginine 
working standard, accurately weighed, to a 25-milliliter volumetric 
flask. Dissolve and dilute to volume with mobile phase (primary working 
standard solution). Further dilute with mobile phase to 0.2 milligram of 
aztreonam per milliliter (estimated).
    (b) Sample solutions. Use separate containers for preparation of 
each sample solution as described in paragraph (b)(1)(ii)(b)(1) and (2) 
of this section.
    (1) Potency (micrograms of aztreonam per milligram). Accurately 
weigh the container contents by difference and quantitatively transfer 
it to a 100-milliliter volumetric flask. Dissolve and dilute to volume 
with mobile phase. Further dilute in mobile phase to 0.2 milligram of 
aztreonam per milliliter (estimated).
    (2) Content (milligrams of aztreonam per container). If packaged in 
containers with capacities of less than 100 milliliters, reconstitute 
the sample as directed in the labeling, using distilled water in lieu of 
reconstituting fluid. If packaged in bottles with capacities of 100 
milliliters or greater, reconstitute with 10 milliliters of distilled 
water. Withdraw the total contents of each container or bottle and 
dilute with mobile phase to a concentration of 0.2 milligram of 
aztreonam per milliliter (estimated).
    (c) Resolution test solution. Dissolve 10 milligrams of open ring 
aztreonam, [[(2-amino-4-thiazolyl)[(1-carboxy-1-
methylethoxy)imino]acetyl]amino]-3-(sulfoamino)-butanoic acid, in 10.0 
milliliters of primary standard solution. Further dilute 5 milliliters 
of this solution to 25.0 milliliters with mobile phase.
    (iii) System suitability requirements--(a) Tailing factor. The 
tailing factor (T) of the aztreonam peak is satisfactory if it is not 
more than 2 at 5 percent of peak height.
    (b) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,000 theoretical plates.
    (c) Resolution. The resolution (R) between aztreonam peak and open 
ring aztreonam is satisfactory if it is not less than 2.0.
    (d) Coefficient of variation. The coefficient of variation 
(SRin percent) of 5 replicate injections is satisfactory if 
it is not more than 2.0 percent.


If the system suitability requirements have been met, then proceed as 
described in Sec. 436.361(b) of this chapter. Alternate chromatographic 
conditions are acceptable provided reproducibility and resolution are 
comparable to the system. However, the sample preparation described in 
paragraph (b)(1)(ii)(b) of this section should not be changed.
    (iv) Calculations--(a) Potency (micrograms per milligram). (1) 
Calculate the micrograms of aztreonam per milligram (uncorrected) as 
follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.340

where:


[[Page 1004]]


Au=Area of the aztreonam peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the aztreonam peak in the chromatogram of the 
          working standard;
Ps=Aztreonam activity in the working standard solution in 
          micrograms per milliliter; and
Cu=Milligrams of sample per milliliter of sample solution.

    (2) Calculate the micrograms of arginine per milligram as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.341
    
where:

Au=Area of the arginine peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the arginine peak in the chromatogram of the 
          working standard;
Ps=Arginine activity in the working standard solution in 
          micrograms per milliliter; and
Cu=Milligrams of sample per milliliter of sample solution.

    (3) Calculate the micrograms of aztreonam per milligram (corrected) 
as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.342

    (b) Content (milligrams of aztreonam per container). Calculate the 
aztreonam content of the container as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.339

where:

Au=Area of the aztreonam peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the aztreonam peak in the chromatogram of the 
          working standard;
Ps=Aztreonam activity in the aztreonam working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of aztreonam per milliliter.
    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams of aztreonam per 
milliliter.
[52 FR 4615, Feb. 13, 1987; 52 FR 8550, Mar. 18, 1987. Redesignated at 
54 FR 40385, Oct. 2, 1989, and amended at 54 FR 41824, Oct. 12, 1989; 55 
FR 11585, Mar. 29, 1990]



Sec. 455.204b  Aztreonam injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Aztreonam injection is a frozen aqueous 
iso-osmotic solution of aztreonam and arginine. Each milliliter contains 
aztreonam equivalent to either 10 milligrams, 20 milligrams, or 40 
milligrams. Its aztreonam content is satisfactory if it is not less than 
90 percent and not more than 120 percent of the number of milligrams of 
aztreonam that it is represented to contain. It is sterile. It is 
nonpyrogenic. Its pH is not less than 4.5 and not more than 7.5. It 
passes the identity test. The aztreonam used conforms to the standards 
prescribed by Sec. 455.4(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:

[[Page 1005]]

    (A) The aztreonam used in making the batch for potency, moisture, 
residue on ignition, heavy metals, and identity.
    (B) The batch for aztreonam potency, sterility, pyrogens, pH, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The aztreonam used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Potency. Proceed as directed in Sec. 436.361 of this chapter, 
except in addition to the column described in paragraph (a)(4) of that 
section, use a 5- to 50-centimeter saturator column having an inside 
diameter of 2 to 4.6 millimeters and packed with approximately 37 
micrometer silica, and use the resolution test solution to determine 
resolution in lieu of the working standard solution. Perform the assay 
at ambient temperature, using an ultraviolet detection system operating 
a wavelength of 206 nanometers, and a column packed with Chromegabond 
Diol (dihydroxypropane chemically bonded to porous silica), 5 to 10 
micrometers or equivalent. Mobile phase, working standard solution, 
sample solution, resolution test solution, system suitability 
requirements, and calculations as follows:
    (i) Mobile phase. Acetonitrile: 0.01M pH 2.0 ammonium phosphate 
(75:25). Transfer 1.15 grams of ammonium phosphate monobasic to a 1-
liter volumetric flask. Add about 800 milliliters of distilled water and 
sonicate to aid dissolution. Adjust the solution to pH 2.0 with o-
phosphoric acid, 85 percent. Dilute the solution to volume with 
distilled water and mix well. Transfer about 250 milliliters of this 
solution and 750 milliliters of acetonitrile to a suitable-sized 
container and mix well. Filter the mobile phase through a suitable glass 
fiber filter or equivalent that is capable of removing particulate 
contamination to 1 micron in diameter. Degas the mobile phase just prior 
to its introduction into the chromatograph pumping system.
    (ii) Preparation of working standard, sample, and resolution test 
solutions--(A) Working standard solution. Transfer approximately 25 
milligrams each of the aztreonam working standard and the arginine 
working standard, accurately weighed, to a 25-milliliter volumetric 
flask. Dissolve and dilute to volume with mobile phase (primary working 
standard solution). Further dilute with mobile phase to obtain a 
solution containing 0.2 milligram of aztreonam per milliliter.
    (B) Sample solution. Using a suitable hypodermic needle and syringe, 
remove an accurately measured representative portion from each container 
and dilute with sufficient mobile phase to obtain a solution containing 
0.2 milligram of aztreonam per milliliter (estimated).
    (C) Resolution test solution. Dissolve 10 milligrams of open ring 
aztreonam, 2-[[(2-amino-4-thiazolyl)[(1-carboxy-1-
methylethoxy)imino]acetyl]amino]-3-(sulfoamino)-butanoic acid, in 10.0 
milliliters of primary standard solution. Further dilute 5 milliliters 
of this solution to 25.0 milliliters with mobile phase.
    (iii) System suitability requirements--(A) Tailing factor. The 
tailing factor (T) of the aztreonam peak is satisfactory if it is not 
more than 2 at 5 percent of peak height.
    (B) Efficiency of the column. The efficiency of the column (n) is 
satisfactory if it is greater than 1,000 theoretical plates.
    (C) Resolution. The resolution (R) between the aztreonam peak and 
open ring aztreonam is satisfactory if it is not less than 2.0.
    (D) Coefficient of variation. The coefficient of variation (SR 
in percent) of 5 replicate injections is satisfactory if it is not more 
than 2.0 percent.

If the system suitability requirements have been met, then proceed as 
described in Sec. 436.361(b) of this chapter. Alternative 
chromatographic conditions are acceptable, provided reproducibility and 
resolution are comparable to the system. However, the

[[Page 1006]]

sample preparation described in paragraph (b)(1)(ii)(B) of this section 
should not be changed.
    (iv) Calculations: Calculate the milligrams of aztreonam per 
milliliter of sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.343

where:

Au=Area of the aztreonam peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the aztreonam peak in the chromatogram of the 
          working standard;
Ps=Aztreonam activity in the aztreonam working standard 
          solution in micrograms per milliliter; and
d=Dilution factor of the sample.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
except inject a sufficient volume of the undiluted solution to deliver 
50 milligrams of aztreonam per kilogram.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (5) Identity. The high-performance liquid chromatogram of the sample 
is determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the aztreonam working standard.
[54 FR 40385, Oct. 2, 1989]



Sec. 455.210  Chloramphenicol injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chloramphenicol injection is 
chloramphenicol, with or without one or more suitable and harmless 
buffer substances, dissolved in one or more suitable and harmless 
solvents. Each milliliter contains 250 milligrams of chloramphenicol. 
Its potency is satisfactory if it is not less than 90 percent and not 
more than 130 percent of the number of milligrams of chloramphenicol 
that it is represented to contain. It is sterile. It is nonpyrogenic. 
Its pH is not less than 4.7 and not more than 5.0. The chloramphenicol 
used conforms to the standards prescribed by Sec.  455.10a(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The chloramphenicol used in making the batch for potency, pH, 
specific rotation, melting range, absorptivity, and crystallinity.
    (b) The batch for potency, sterility, pyrogens, and pH.
    (ii) Samples required:
    (a) The chloramphenicol used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of eight immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dilute an accurately measured representative portion of the sample in 
sufficient distilled water to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of the stock solution with 
distilled water to the reference concentration of 2.5 micrograms of 
chloramphenicol per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
add the contents of each container directly to the dry filter, thus 
eliminating the preliminary solubilization step.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 5 milligrams per milliliter.
    (4)-(5) [Reserved]
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted drug.
[39 FR 19166, May 30, 1974, as amended at 45 FR 64568, Sept. 30, 1980; 
48 FR 3961, Jan. 28, 1983; 48 FR 7440, Feb. 22, l983; 50 FR 19921, May 
13, 1985]

[[Page 1007]]



Sec. 455.212  Sterile chloramphenicol sodium succinate.

    The requirements for certification and the tests and methods of 
assay for sterile chloramphenicol sodium succinate packaged for 
dispensing are described in Sec. 455.12a.
[43 FR 9801, Mar. 10, 1978]



Sec. 455.230  Moxalactam disodium for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Moxalactam disodium for injection is a 
dry mixture of moxalactam disodium and mannitol. Its moxalactam content 
is satisfactory if it is not less than 90 percent and not more tha 120 
percent of the number of milligrams of moxalactam that it is represented 
to contain. The moxalactam content of the dry mixture is not less than 
722 micrograms of moxalactam per milligram. The ratio of R-isomer to S-
isomer is not less than 0.8 and not more than 1.4. It is sterile. It is 
nonpyrogenic. Its moisture content is not more than 3.0 percent. Its pH 
is not less than 4.5 and not more than 7.0. It passes the identity test 
for moxalactam.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for moxalactam content, 
isomer ratio, sterility, pyrogens, moisture, pH, and identity.
    (ii) Samples required on the batch:
    (a) For all tests except sterility: A minimum of 10 immediate 
containers.
    (b) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Moxalactam content; isomer 
ratio. Proceed as directed in Sec. 436.332 of this chapter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 50 milligrams of moxalactam.
    (4) [Reserved]
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 100 milligrams per milliliter.
    (7) Identity. Proceed as directed in Sec. 436.333 of this chapter.
[46 FR 61070, Dec. 15, 1981, as amended at 50 FR 19921, May 13, 1985]



Sec. 455.251  Sodium novobiocin for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sodium novobiocin for injection is sodium 
novobiocin with or without one or more suitable solubilizing agents, 
preservatives, and diluents. Each vial contains 500 milligrams of 
novobiocin. Its potency is satisfactory if it is not less than 90 
percent and not more than 120 percent of the number of milligrams of 
novobiocin that it is represented to contain. It is sterile and 
nonpyrogenic. Its loss on drying is not more than 6.0 percent. Its pH, 
when reconstituted as directed in the labeling, is not less than 6.5 and 
not more than 8.5. The sodium novobiocin used conforms to the standards 
prescribed by Sec. 455.51a(a)(1) (i), (iii), (v), (vi), (vii), and 
(viii).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (a) The sodium novobiocin used in making the batch for potency, loss 
on drying, pH, residue on ignition, specific rotation, crystallinity, 
and identity.
    (b) The batch for potency, sterility, pyrogens, loss on drying, and 
pH.
    (ii) Samples required:
    (a) The sodium novobiocin used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.

[[Page 1008]]

    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Then using a suitable 
hypodermic needle and syringe, remove all of the withdrawable contents 
if it is represented as a single dose container; or if the labeling 
specifies the amount of potency in a given volume of the resultant 
preparation, remove an accurately measured representative portion from 
each container. Dilute with 0.1M potassium phosphate buffer, pH 8.0 
(solution 3), to give a stock solution of convenient concentration. 
Further dilute an aliquot of the stock solution with 10 percent 
potassium phosphate buffer, pH 6.0 (solution 6), to the reference 
concentration of 0.5 microgram of novobiocin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32 of this chapter, 
using a solution containing 10 milligrams of novobiocin per milliliter.
    (4) [Reserved]
    (5) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the sample after reconstituting as directed in the labeling.
[39 FR 19166, May 20, 1974, as amended at 46 FR 25608, May 8, 1981; 50 
FR 19921, May 13, 1985]



Sec. 455.270  Rifampin for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Rifampin for injection is a dry mixture 
of rifampin, sodium formaldehyde sulfoxylate, and sodium hydroxide. Its 
potency is 600 milligrams per vial. Its potency is satisfactory if it is 
not less than 90 percent and not more than 115 percent of the number of 
milligrams of rifampin that it is represented to contain. It is sterile. 
It is nonpyrogenic. Its moisture content is not more than 3.0 percent. 
Its pH is not less than 7.8 and not more than 8.8. It passes the 
identity test. The rifampin used conforms to the standards prescribed by 
Sec. 455.70(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (A) The rifampin used in making the batch for potency, loss on 
drying, pH, absorptivity, identity, and crystallinity.
    (B) The batch for potency, sterility, pyrogens, moisture, pH, and 
identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research.
    (A) The rifampin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Using a suitable hypodermic 
needle and syringe, remove the withdrawable contents from each container 
represented as a single-dose container; or if the labeling specifies the 
amount of potency in a given volume of the preparation, withdraw an 
accurately measured volume from each container. Dilute with 1 percent 
potassium phosphate buffer, pH 6.0 (solution 1) to give a stock solution 
of 1.0 milligram of rifampin per milliliter (estimated). Further dilute 
an aliquot of the stock solution with solution 1 to the reference 
concentration of 5.0 micrograms of rifampin per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(b) of this chapter, 
using a solution containing 10 milligrams of rifampin per milliliter.

[[Page 1009]]

    (4) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
concentration of 60 milligrams of rifampin per milliliter.
    (6) Identity. Proceed as directed in Sec. 436.365 of this chapter.
[54 FR 38375, Sept. 18, 1989]



Sec. 455.280a  Sterile spectinomycin hydrochloride.

    The requirements for certification and the tests and methods of 
assay for sterile spectinomycin hydrochloride packaged for dispensing 
are described in Sec. 455.80a.



Sec. 455.285  Vancomycin hydrochloride injectable dosage forms.



Sec. 455.285a  Sterile vancomycin hydrochloride.

    The requirements for certification and the tests and methods of 
assay for sterile vancomycin hydrochloride packaged for dispensing are 
described in Sec. 455.85a.



Sec. 455.285b  Vancomycin hydrochloride for injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Vancomycin hydrochloride for injection is 
a dry mixture of vancomycin hydrochloride and a suitable stabilizing 
agent. It contains not less than 925 micrograms of vancomycin per 
milligram, calculated on an anhydrous basis. Its vancomycin content is 
satisfactory if it is not less than 90 percent and not more than 115 
percent of the number of milligrams of vancomycin that it is represented 
to contain. It contains not less than 88 percent vancomycin factor B. It 
contains not more than 4 percent of any individual vancomycin related 
factor. It is sterile. It is nonpyrogenic. Its moisture content is not 
more than 5 percent. The pH of an aqueous solution containing 50 
milligrams per milliliter is not less than 2.5 and not more than 4.5. 
Its heavy metals content is not more than 30 parts per million. It gives 
a positive identity test. The vancomycin hydrochloride used conforms to 
the standards prescribed by Sec. 455.85(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Request for certification; samples. In addition to the 
requirements of Sec. 431.1 of this chapter, each such request shall 
contain:
    (i) Results of tests and assays on:
    (A) The vancomycin hydrochloride used in making the batch for 
potency, moisture, pH, factor A content, and identity.
    (B) The batch for vancomycin potency, vancomycin content, 
chromatographic purity, sterility, pyrogens, moisture, pH, heavy metals, 
and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The vancomycin used in making the batch: 10 packages, each 
containing approximately 500 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 10 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Test and methods of assay--(1) Vancomycin potency and content. 
Determine both micrograms of vancomycin per milligram of sample and 
milligrams of vancomycin per container. Proceed as directed in 
Sec. 435.105 of this chapter, preparing the sample solution as follows:
    (i) Preparation of sample solution. Use separate containers for 
preparation of each sample solution as described in paragraphs (b)(1)(i) 
(A) and (B) of this section.
    (A) Micrograms of vancomycin per milligram. Dissolve an accurately 
weighed sample of approximately 30 milligrams in sufficient distilled 
water to obtain a stock solution of 1 milligram per milliliter. Further 
dilute an aliquot of the stock solution with 0.1M potassium phosphate 
buffer, pH 4.5 (solution 4) to the reference concentration of 10.0 
micrograms of vancomycin per milliliter (estimated).
    (B) Milligrams of vancomycin per container. Reconstitute as directed 
in the labeling. Using a suitable hypodermic needle and syringe, remove 
all of the withdrawable contents if it is represented as a single-dose 
container; or,

[[Page 1010]]

if the labeling specifies the amount of vancomycin content in a given 
volume of the resultant preparation, remove an accurately measured 
representative portion from each container. Dilute with 0.1M potassium 
phosphate buffer, pH 4.5 (solution 4) to the reference concentration of 
10.0 micrograms of vancomycin per milliliter (estimated).
    (2) Chromatographic purity. Proceed as directed in Sec. 436.366 of 
this chapter. The relative amount of vancomycin B is not less than 88 
percent and the relative amount of any related substance is not more 
than 4 percent.
    (3) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section, except 
use sterile distilled water in lieu of diluting fluid A.
    (4) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 5 milligrams of vancomycin per milliliter.
    (5) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (6) pH. Proceed as directed in Sec. 436.202 of this chapter, using a 
solution containing 50 milligrams per milliliter.
    (7) Heavy metals. Proceed as directed in Sec. 436.208 of this 
chapter.
    (8) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using the 0.5 percent potassium bromide disc preparation as described in 
paragraph (b)(1) of that section.
[54 FR 20384, May 11, 1989; 54 FR 22838, May 28, 1989]



Sec. 455.285c  Vancomycin hydrochloride injection.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Vancomycin hydrochloride injection is a 
frozen, aqueous, iso-osmotic solution of vancomycin hydrochloride and a 
tonicity adjusting agent. Each milliliter contains vancomycin 
hydrochloride equivalent to 5 milligrams of vancomycin. Its vancomycin 
content is satisfactory if it is not less than 90 percent and not more 
than 115 percent of the number of milligrams of vancomycin that it is 
represented to contain. It contains not less than 88 percent vancomycin 
factor B. It contains not more than 4 percent of any individual 
vancomycin related factor. It is sterile. It contains not more than 0.33 
U.S.P. Endotoxin Unit per milligram of vancomycin hydrochloride. Its pH 
is not less than 3.0 and not more than 5.0. The vancomycin used conforms 
to the standards prescribed by Sec. 455.86.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter. In addition, this drug shall 
be labeled ``vancomycin hydrochloride injection.''
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The vancomycin used in making the batch for vancomycin potency, 
chromatographic purity, moisture, heavy metals, and identity.
    (B) The batch for vancomycin content, chromatographic purity, 
sterility, bacterial endotoxins, pH, and identity.
    (ii) Samples, if required by the Director, Center for Drug 
Evaluation and Research:
    (A) The vancomycin used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (B) The batch:
    (1) For all tests except sterility: A minimum of 12 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay. Thaw the sample as directed in the 
labeling. The sample solution used for testing must be at room 
temperature.
    (1) Vancomycin content. Proceed as directed in Sec. 436.105 of this 
chapter, preparing the sample solution as follows: Using a suitable 
hypodermic needle and syringe, remove an accurately measured 
representative portion from each container immediately after thawing and 
reaching room temperature. Dilute with 0.1M potassium phosphate buffer, 
pH 4.5 (solution 4), to the reference concentration of 10 micrograms of 
vancomycin per milliliter (estimated).
    (2) Chromatographic purity. Proceed as directed in Sec. 436.366 of 
this chapter. The relative amount of vancomycin B is not less than 88 
percent and the relative amount of any related substance is not more 
than 4 percent.

[[Page 1011]]

    (3) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in Sec. 436.20(e)(1), except use sterile 
distilled water in lieu of diluting fluid A.
    (4) Bacterial endotoxins. Proceed as directed in the U.S.P. 
bacterial endotoxins test. The specimen under test contains not more 
than 0.33 U.S.P. Endotoxin Unit per milligram of vancomycin 
hydrochloride.
    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
    (6) Identity. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(2) of this section compares 
qualitatively to that of the vancomycin working standard.
[59 FR 8400, Feb. 22, 1994]



Sec. 455.290  Vidarabine monohydrate for infusion.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Vidarabine monohydrate for infusion 
contains in each milliliter vidarabine monohydrate equivalent to 187.4 
milligrams of vidarabine in an aqueous suspension containing suitable 
and harmless buffers and preservatives. Its potency is satisfactory if 
it is not less than 90 percent and not more than 120 percent of the 
number of milligrams of vidarabine that it is represented to contain. It 
is sterile. It is nonpyrogenic. It contains no histamine or histamine-
like substances. Its pH is not less than 5.0 and not more than 6.2. The 
vidarabine monohydrate used conforms to the standards prescribed by 
Sec. 455.90a (a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``vidarabine for 
infusion''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The vidarabine monohydrate used in making the batch for 
vidarabine content, loss on drying, specific rotation, and identity.
    (b) The batch for vidarabine content, sterility, pyrogens, 
histamine, and pH.
    (ii) Samples required:
    (a) The vidarabine monohydrate used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 16 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Vidarabine content. Proceed as 
directed in Sec. 436.325 of this chapter, except prepare the sample 
solution and calculate the vidarabine content as follows:
    (i) Preparation of sample solution. Using a suitable hypodermic 
needle and syringe, transfer 2 milliliters of the well-shaken suspension 
to a 500-milliliter volumetric flask. Add approximately 50 milliliters 
of distilled water and 5 milliliters of glacial acetic acid. Warm on a 
steam bath for 15 minutes to dissolve the vidarabine. Cool to room 
temperature and dilute to volume with distilled water. Transfer 4 
milliliters to a 25-milliliter volumetric flask and dilute to volume 
with distilled water.
    (ii) Calculations. Calculate the vidarabine content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.344
    
where:
    A=Area of the vidarabine sample peak (at a retention time equal to 
that observed for the standard);
    B=Area of the standard peak;
    Ws=Weight of the standard in milligrams; and
    f=Potency of standard in micrograms per milligram.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(2) of that section.
    (3) Pyrogens. Proceed as directed in Sec. 436.32(a) of this chapter, 
using a solution containing 10 milligrams of vidarabine per milliliter.
    (4) Histamine. Proceed as directed in Sec. 436.35 of this chapter. 
Apply sufficient heat to dissolve the vidarabine.

[[Page 1012]]

    (5) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted suspension.
[44 FR 1374, Jan. 5, 1979, as amended at 44 FR 30334, May 25, 1979; 50 
FR 19921, May 13, 1985]



                   Subpart D--Ophthalmic Dosage Forms



Sec. 455.310  Chloramphenicol ophthalmic dosage forms.



Sec. 455.310a  Chloramphenicol ophthalmic solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chloramphenicol ophthalmic solution 
contains in each milliliter 5 milligrams of chloramphenicol with or 
without one or more suitable and harmless preservatives, buffer 
substances, and surfactants, in an aqueous solution. Its potency is 
satisfactory if it is not less than 90 percent and not more than 130 
percent of the number of milligrams of chloramphenicol that it is 
represented to contain. It is sterile. Its pH is not less than 3 nor 
more than 6; however, if the solution is buffered, its pH is not less 
than 7.0 nor more than 7.5. The chloramphenicol used conforms to the 
standards prescribed by Sec. 455.10(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The chloramphenicol used in making the batch for potency, pH, 
specific rotation, melting range, absorptivity, and crystallinity.
    (b) The batch for potency, sterility, and pH.
    (ii) Samples required:
    (a) The chloramphenicol used in making the batch: 10 containers, 
each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of five immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dilute an accurately measured representative portion of the sample with 
sufficient distilled water to obtain a stock solution of convenient 
concentration. Further dilute an aliquot of the stock solution with 
distilled water to the reference concentration of 2.5 micrograms of 
chloramphenicol per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the undiluted solution.
[39 FR 19166, May 30,1974, as amended at 43 FR 59057, Dec. 19, 1978; 46 
FR 46313, Sept. 18, 1981; 48 FR 3961, Jan. 28, 1983; 50 FR 19921, May 
13, 1985]



Sec. 455.310b  Chloramphenicol for ophthalmic solution.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chloramphenicol for ophthalmic solution 
contains 25 milligrams of chloramphenicol with one or more suitable and 
harmless buffer substances. When reconstituted as directed in the 
labeling, its potency is not less than 90 percent and not more than 130 
percent of the number of milligrams of chloramphenicol that it is 
represented to contain. It is sterile. Its pH is not less than 7.1 and 
not more than 7.5. The chloramphenicol used conforms to the standards 
prescribed by Sec. 455.10(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The chloramphenicol used in making the batch for potency, pH, 
specific rotation, melting range, absorptivity, and crystallinity.
    (b) The batch for potency, sterility, and pH.
    (ii) Samples required:
    (a) The chloramphenicol used in making the batch: 10 packages, each

[[Page 1013]]

containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of five immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods:
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an accurately measured 
representative aliquot of the sample with sufficient distilled water to 
obtain a stock solution of convenient concentration. Further dilute an 
aliquot of the stock solution with distilled water to the reference 
concentration of 2.5 micrograms of chloramphenicol per milliliter 
(estimated).
    (ii) Spectrophotometric assay. Reconstitute the sample as directed 
in the labeling and dilute a 1.0-milliliter aliquot in sufficient 
distilled water to obtain a solution containing 20 micrograms of 
chloramphenicol per milliliter. Dissolve an accurately weighed portion 
of the working standard in sufficient distilled water to obtain a 
solution containing 20 micrograms per milliliter. Using a suitable 
spectrophotometer and distilled water as the blank, determine the 
absorbance of the sample and standard solutions at 278 nanometers. 
Calculate the potency of the sample as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.345

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 5 milligrams per milliliter.
[49 FR 6093, Feb. 17, 1984, as amended at 50 FR 19921, May 13, 1985]



Sec. 455.310c  Chloramphenicol ointment (chloramphenicol cream).

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chloramphenicol ointment is 
chloramphenicol in a suitable and harmless ointment base, with or 
without suitable and harmless buffer substances, dispersing and 
suspending agents. It may contain cortisone or a suitable derivative of 
cortisone. If such base is water-miscible, it shall contain a suitable 
and harmless preservative. Its potency is not less than 1.0 milligram 
per gram. If it is intended for ophthalmic use, it is sterile. The 
chloramphenicol used conforms to the requirements of Sec. 455.10a(a)(1), 
except paragraphs (a)(1) (ii), (iii), and (v) of that section. The 
chloramphenicol used in making the chloramphenicol ophthalmic ointment 
conforms to the requirements of Sec. 455.10a(a)(1), except paragraphs 
(a)(1) (iii) and (v) of that section. Each other substance used, if its 
name is recognized in the U.S.P. or N.F., conforms to the standards 
prescribed therefor by such official compendium.
    (2) Packaging. Unless it is packaged in a single dose container, 
chloramphenicol ointment shall be packaged in collapsible tubes, which 
shall be well-closed containers as defined by the U.S.P., and shall not 
be larger than the \1/8\-ounce size if such ointment is represented for 
ophthalmic use, and in no case larger than the 2-ounce size, except that 
if it is labeled solely for hospital use it may be packaged in immediate 
containers of glass which meet the test for tight containers as defined 
by the U.S.P. The composition of the immediate container and closure 
shall be such as will not cause any change in the strength, quality, or 
purity of the contents beyond any limit therefor in

[[Page 1014]]

applicable standards, except that minor changes so caused which are 
normal and unavoidable in good packaging, storage, and distribution 
practice shall be disregarded.
    (3) Labeling. In addition to the labeling requirements prescribed by 
Sec. 201.100 of this chapter (regulations issued under section 502(f) of 
the act), each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On the outside wrapper or container and the immediate container 
the statement ``Expiration date ----------'', the blank being filled in 
with the date that is 60 months, or 24 months if it is packaged in an 
immediate container other than tin or glass, or 12 months if the 
ointment base is water miscible, after the month during which the batch 
was certified.
    (ii) If it contains one of the active ingredients specified in 
paragraph (a)(1) of this section, after the name ``chloramphenicol 
ointment'', wherever it appears, the name of the active ingredient, in 
juxtaposition with such name.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The chloramphenicol used in making the batch for potency, pH, 
specific rotation, melting point, and absorptivity.
    (b) The batch for potency and for sterility if the ointment is 
intended for ophthalmic use.
    (ii) Samples required:
    (a) The chloramphenicol used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 5 immediate 
containers if it is packaged in immediate containers of tin or glass; a 
minimum of 20 immediate containers if it is packaged in immediate 
containers other than tin or glass.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows:
    (i) If the ointment is water miscible. Place an accurately weighed 
representative portion of the sample into a high-speed glass blender jar 
containing 1.0 milliliter polysorbate 80 and sufficient distilled water 
to obtain a stock solution of convenient concentration. Blend for 3 to 5 
minutes. Further dilute an aliquot of the stock solution with distilled 
water to the reference concentration of 2.5 micrograms of 
chloramphenicol per milliliter (estimated).
    (ii) If the ointment is not water miscible. Place an accurately 
weighed representative portion of the sample into a separatory funnel 
containing approximately 50 milliliters of petroleum ether. Shake the 
sample and ether until homogeneous. Add 20 to 25 milliliters of 
distilled water and shake well. Allow the layers to separate. Remove the 
aqueous layer and repeat the extraction procedure with each of three 
more 20- to 25-milliliter quantities of distilled water. Combine the 
aqueous extractives in a suitable volumetric flask and dilute to volume 
with distilled water. Remove an aliquot and further dilute with 
distilled water to the reference concentration of 2.5 micrograms of 
chloramphenicol per milliliter (estimated). The potency of 
chloramphenicol ointment is satisfactory if it contains not less than 90 
percent and not more than 130 percent of the number of milligrams of 
chloramphenicol that it is represented to contain.
    (2) Sterility. If the ointment is intended for ophthalmic use, 
proceed as directed in Sec. 436.20 of this chapter, using the method 
described in paragraph (e)(3) of that section. However, if the ointment 
is not soluble in isopropyl myristate proceed as directed in Sec. 436.20 
of this chapter, using the method described in Sec. 436.20(e)(2), except 
use 100 milligrams in lieu of 300 milligrams of solids.
[39 FR 19166, May 30, 1974, as amended at 41 FR 10886, Mar. 15, 1976; 44 
FR 10380, Feb. 20, 1979; 48 FR 3961, Jan. 28, 1983; 50 FR 19921, May 13, 
1985]

[[Page 1015]]



Sec. 455.310d  Chloramphenicol-polymyxin ointment.

    (a) Requirements for certification. Chloramphenicol-polymyxin 
ointment conforms to all requirements and is subject to all procedures 
prescribed by Sec. 455.310c(a) for chloramphenicol ointment, except 
that:
    (1) It contains not less than 10,000 units of polymyxin B per gram. 
The polymyxin B used conforms to the requirements prescribed for 
polymyxin B by Sec. 444.170a(a)(1) of this chapter.
    (2) In lieu of the labeling prescribed by Sec. 455.310c(a)(3)(i)(a), 
each package shall bear on the outside wrapper or container and the 
immediate container, the statement ``Expiration date ----------'', the 
blank being filled in with the date that is 24 months after the month 
during which the batch was certified, except that the blank may be 
filled in with the date that is 36 months, 48 months, or 60 months after 
the month during which the batch was certified if the person who 
requests certification has submitted to the Commissioner results of 
tests and assays showing that after having been stored for such period 
of time such drug as prepared by him complies with the standards 
prescribed by this section: Provided however, That such expiration date 
may be omitted from the immediate container if it contains a single dose 
and it is packaged in an individual wrapper or container.
    (3) In addition to complying with the requirements of 
Sec. 455.310c(a)(4), a person who requests certification of a batch 
shall submit with his request a statement showing the batch mark and 
(unless previously submitted) the results and date of the latest tests 
and assays of the polymyxin used in making the batch for potency. He 
shall also submit in connection with his request a sample consisting of 
not less than 6 packages of ointment and (unless it was previously 
submitted) a sample consisting of 5 packages containing approximately 
equal portions of not less than 0.5 gram each of the polymyxin used in 
making the batch.
    (b) Tests and methods of assay--(1) Potency--(i) Chloramphenicol 
content. Proceed as directed in Sec. 455.310c(b). Its chloramphenicol 
content is satisfactory if it contains not less than 90 percent and not 
more than 120 percent of the number of milligrams per gram that it is 
represented to contain.
    (ii) Polymyxin content. Proceed as directed in 
Sec. 444.170a(b)(2)(i) of this chapter, except in lieu of the directions 
in Sec. 444.170a(b)(2)(i)(g) of this chapter for the preparation of the 
sample, prepare the sample as follows: Place an accurately weighed 
sample (usually approximately 1.0 gram) in a separatory funnel 
containing approximately 50 milliliters of peroxide-free ether, and 
shake the sample and ether until homogeneous. Add 25 milliliters of 10-
percent potassium phosphate buffer, pH 6.0 and shake. Remove the buffer 
layer and repeat the extraction with three additional 25-milliliter 
portions of buffer. Combine the extractives and make the proper 
estimated dilutions, using the buffer solution, except that, if the 
sample contains a water-soluble base, place an accurately weighed 
representative sample in a blending jar containing 1.0 milliliter of 
polysorbate 80 and sufficient 10 percent potassium phosphate buffer, pH 
6.0, to give a final volume of 200 milliliters. Using a highspeed 
blender, blend the mixture for 2 minutes to 3 minutes and then make the 
proper estimated dilutions with 10 percent phosphate buffer pH 6.0. Its 
content of polymyxin is satisfactory if it contains not less than 90 
percent and not more than 125 percent of the number of units per gram 
that it is represented to contain.
    (2) Sterility. If the ointment is intended for ophthalmic use, 
proceed as directed in Sec. 436.20 of this chapter, using the method 
described in paragraph (e)(3) of that section. However, if the ointment 
is not soluble in isopropyl myristate proceed as directed in Sec. 436.20 
of this chapter, using the method described in paragraph (e)(2) of that 
section, except use 100 milligrams in lieu of 300 milligrams of solids.
[39 FR 19166, May 30, 1974, as amended at 47 FR 23443, May 28, 1982; 50 
FR 19921, May 13, 1985]



Sec. 455.310e  Chloramphenicol - hydrocortisone acetate for ophthalmic suspension.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality,

[[Page 1016]]

and purity. Chloramphenicol-hydrocortisone acetate for ophthalmic 
suspension contains 12.5 milligrams of chloramphenicol and 25 milligrams 
of hydrocortisone acetate with one or more suitable and harmless buffer 
substances, preservatives, and diluents. When reconstituted as directed 
in the labeling, its potency is not less than 90 percent and not more 
than 130 percent of the number of milligrams of chloramphenicol that it 
is represented to contain. It is sterile. Its pH is not less than 7.1 
and not more than 7.5. The chloramphenicol used conforms to the 
standards prescribed by Sec. 455.10(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The chloramphenicol used in making the batch for potency, pH, 
specific rotation, melting range, absorptivity, and crystallinity.
    (b) The batch for potency, sterility, and pH.
    (ii) Samples required:
    (a) The chloramphenicol used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of five immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Use either of the 
following methods:
    (i) Microbiological turbidimetric assay. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an accurately measured 
representative aliquot of the sample with sufficient distilled water to 
obtain a stock solution of convenient concentration. Further dilute an 
aliquot of the stock solution with distilled water to the reference 
concentration of 2.5 micrograms of chloramphenicol per milliliter 
(estimated).
    (ii) Spectrophotometric assay. Reconstitute the sample as directed 
in the labeling and dilute a 1.0-milliliter aliquot in sufficient 
distilled water to obtain a solution containing 20 micrograms of 
chloramphenicol per milliliter. Dissolve an accurately weighed portion 
of the working standard in sufficient distilled water to obtain a 
solution containing 20 micrograms per milliliter. Using a suitable 
spectrophotometer and distilled water as the blank, determine the 
absorbance of the sample and standard solutions at 278 nanometers. 
Calculate the potency of the sample as follows:

Milligrams of chloramphenicol per milliliter=Absorbance of sample X 
labeled potency per milliliter in milligrams/Absorbance of standard.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
an aqueous solution containing 5 milligrams per milliliter.
[49 FR 6093, Feb. 17, 1984]



Sec. 455.390  Vidarabine monohydrate ophthalmic ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Vidarabine monohydrate ophthalmic 
ointment contains in each gram vidarabine monohydrate equivalent to 
28.11 milligrams of vidarabine in a suitable and harmless base. Its 
potency is satisfactory if it is not less than 90 percent and not more 
than 120 percent of the number of milligrams of vidarabine that it is 
represented to contain. It is sterile. It passes the test for metal 
particles. The vidarabine monohydrate used conforms to the standards 
prescribed by Sec. 455.90a(a)(1).
    (2) Labeling. In addition to the labeling requirements prescribed by 
Sec. 432.5 of this chapter, this drug shall be labeled ``vidarabine 
ophthalmic ointment''.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:k

[[Page 1017]]

    (a) The vidarabine monohydrate used in making the batch for 
vidarabine content sterility, loss on drying, specific rotation, and 
identity.
    (b) The batch for vidarabine content, sterility, and metal 
particles.
    (ii) Samples required:
    (a) The vidarabine monohydrate used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 16 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Vidarabine content. Proceed as 
directed in Sec. 436.325 of this chapter, except prepare the sample 
solution and calculate the vidarabine content as follows:
    (i) Preparation of sample solution. Accurately weigh a portion of 
the sample containing the equivalent of approximately 12 milligrams of 
vidarabine (estimated) into a 100-milliliter volumetric flask. Add 
approximately 80 milliliters of distilled water and heat for 15 minutes 
on a steam bath. Shake to dissolve the vidarabine and, while the 
solution is still hot, add 10 milliliters of heptane to dissolve the 
ointment base. Swirl gently until the ointment base is dissolved. Cool 
to room temperature and dilute the aqueous phase to volume with 
distilled water. Discard the heptane phase and mix the solution.
    (ii) Calculations. Calculate the vidarabine content as follows:
    [GRAPHIC] [TIFF OMITTED] TR01JA93.346
    
where:

    A=Area of the vidarabine sample peak (at a retention time equal to 
that observed for the standard);
    B=Area of the standard peak;
    Ws=Weight of standard in milligrams;
    Wu=Weight of sample in milligrams; and
    f=Potency of standard in micrograms per milligram.

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(3) of that section.
    (3) Metal particles. Proceed as directed in Sec. 436.206 of this 
chapter.
[42 FR 44224, Sept. 2, 1977, as amended at 44 FR 30335, May 25, 1979; 50 
FR 19921, May 13, 1985]



                      Subpart E--Otic Dosage Forms



Sec. 455.410  Chloramphenicol otic.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chloramphenicol otic is a solution of 
chloramphenicol in a suitable and harmless vehicle. Each milliliter 
contains 5.0 milligrams of chloramphenicol. Its potency is satisfactory 
if it is not less than 90 percent and not more than 130 percent of the 
number of milligrams of chloramphenicol that it is represented to 
contain. It is sterile. Its moisture content is not more than 2 percent. 
Its pH is not less than 4 and not more than 8. The chloramphenicol used 
conforms to the standards prescribed by Sec. 455.10(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain the following:
    (i) Results of tests and asays on--
    (a) The chloramphenicol used in making the batch for potency, pH, 
specific rotation, melting range, absorptivity, and crystallinity; and
    (b) The batch for potency, sterility, moisture, and pH.
    (ii) Samples required:
    (a) The chloramphenicol used in making the batch: 10 packages, each 
containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Dilute an accurately measured representative portion of the sample with 
distilled water to obtain a stock solution of convenient concentration. 
Further dilute an aliquot of

[[Page 1018]]

the stock solution with distilled water to the reference concentration 
of 2.5 micrograms of chloramphenicol per milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the sample diluted with an equal volume of distilled water.
[44 FR 5881, Jan. 30, 1979, as amended at 48 FR 3961, Jan. 28, 1983; 50 
FR 19921, May 13, 1985]



                  Subpart F--Dermatologic Dosage Forms



Sec. 455.510  Chloramphenicol dermatologic dosage forms.



Sec. 455.510a  Chloramphenicol ointment (chloramphenicol cream).

    The requirements for certification and the tests and methods of 
assay for chloramphenicol ointment (chloramphenicol cream) are described 
in Sec. 455.310c.



Sec. 455.510b  [Reserved]



Sec. 455.510c  Chloramphenicol-polymyxin ointment.

    The requirements for certification and the tests and methods of 
assay for chloramphenicol-polymyxin ointment are described in 
Sec. 455.310d.



Sec. 455.510d  Fibrinolysin and desoxyribonuclease, combined (bovine) with chloramphenicol ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Fibrinolysin and desoxyribonuclease, 
combined (bovine) with chloramphenicol ointment is fibrinolysin, 
desoxyribonuclease, and chloramphenicol in a suitable and harmless 
ointment base. It contains a suitable and harmless preservative. Each 
gram contains 1 unit of fibrinolysin, 666 units of desoxyribonuclease, 
and 10 milligrams of chloramphenicol. Its chloramphenicol content is 
satisfactory if it is not less then 90 percent and not more than 120 
percent of the number of milligrams of chloramphenicol that it is 
represented to contain. The chloramphenicol used conforms to the 
standards prescribed by Sec. 455.10, except paragraph (b)(2) of that 
section. In addition to the requirements prescribed by this paragraph, 
the drug satisfies the requirements designated therefor by the Center 
for Biologics Evaluation and Research, Food and Drug Administration, 
Department of Health and Human Services.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The chloramphenicol used in making the batch for potency, pH, 
specific rotation, melting range, absorptivity, and crystallinity.
    (b) The batch for potency.
    (ii) Samples required:
    (a) The chloramphenicol used in making the batch: 10 packages each 
containing approximately 300 milligrams.
    (b) The batch: A minimum of 5 containers if it is packaged in 
immediate containers of tin or glass, and a minimum of 20 immediate 
containers if it is packaged in immediate containers other than tin or 
glass.
    (b) Tests and methods of assay; potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Place an accurately weighed representative portion of the sample into a 
separatory funnel containing approximately 50 milliliters of petroleum 
ether. Shake the sample and ether until homogeneous. Add 20 to 25 
milliliters of distilled water and shake well. Allow the layers to 
separate. Remove the aqueous layer and repeat the extraction procedure 
with each of three more 20- to 25-milliliter quantities of distilled 
water. Combine the aqueous extractives in a suitable volumetric flask 
and dilute to volume with distilled water. Remove an aliquot and further 
dilute with distilled water to the reference concentration of 2.5

[[Page 1019]]

micrograms of chloramphenicol per milliliter (estimated).
[44 FR 10380, Feb. 20, 1979, as amended at 48 FR 3961, Jan. 28, 1983; 50 
FR 19921, May 13, 1985; 55 FR 11585, Mar. 29, 1990]



Sec. 455.540  Mupirocin ointment.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Mupirocin ointment is mupirocin in a 
suitable and harmless ointment base. Each gram of ointment contains 20 
milligrams of mupirocin. Its mupirocin content is satisfactory if it is 
not less than 90 percent and not more than 110 percent of the number of 
milligrams of mupirocin that it is represented to contain. It passes the 
identity test. The mupirocin used conforms to the standards prescribed 
by Sec. 455.40(a)(1).
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (A) The mupirocin used in making the batch for potency, moisture, 
pH, crystallinity, and identity.
    (B) The batch for mupirocin content and identity.
    (ii) Samples, if required by the Center for Drug Evaluation and 
Research:
    (A) The mupirocin used in making the batch: 10 packages, each 
containing not less than 300 milligrams.
    (B) The batch: A minimum of 10 immediate containers.
    (b) Tests and methods of assay--(1) Mupirocin content. Proceed as 
directed in Sec. 455.40(b)(1), preparing the sample solution and 
calculating the mupirocin content as follows:
    (i) Sample solution. Accurately weigh approximately 0.5 gram of 
ointment and dissolve in 20 milliliters of acetonitrile. Transfer to a 
100-milliliter volumetric flask with the aid of pH 6.3 phosphate buffer. 
Dilute to volume with pH 6.3 phosphate buffer. Mix well. The sample 
solution contains approximately 100 micrograms of mupirocin per 
milliliter (estimated).
    (ii) Calculations. Calculate the mupirocin content in milligrams per 
gram as follows:
[GRAPHIC] [TIFF OMITTED] TR01JA93.347

where:

Au=Area of the mupirocin peak in the chromatogram of the 
          sample (at a retention time equal to that observed for the 
          standard);
As=Area of the mupirocin peak in the chromatogram of the 
          mupirocin working standard;
As=Mupirocin activity in the mupirocin working standard 
          solution in micrograms per milliliter;
d=Dilution factor of the sample; and
n=Number of grams of sample assayed.

    (2) Identity. The high-performance liquid chromatogram of the sample 
determined as directed in paragraph (b)(1) of this section compares 
qualitatively to that of the mupirocin working standard.
[55 FR 2642, Jan. 26, 1990]



PART 460--ANTIBIOTIC DRUGS INTENDED FOR USE IN LABORATORY DIAGNOSIS OF DISEASE--Table of Contents




                     Subpart A--Susceptibility Discs

Sec.
460.1  Certification procedures for antibiotic susceptibility discs.
460.6  Tests and methods of assay for potency of antibiotic 
          susceptibility discs.
460.11  Certification procedures for antibiotic elution susceptibility 
          discs.
460.15  Streptomycin sulfate discs for use in culture media.
460.16  Rifampin discs for use in culture media.

                    Subpart B--Susceptibility Powders

460.25  Bacitracin diagnostic sensitivity powder.
460.28  Disodium carbenicillin diagnostic sensitivity powder.
460.33  Clindamycin hydrochloride hydrate sensitivity powder.
460.38  Sodium colistimethate diagnostic sensitivity powder.
460.42  Dihydrostreptomycin sulfate diagnostic sensitivity powder.
460.47  Doxycycline hyclate diagnostic sensitivity powder.
460.55  Lincomycin hydrochloride monohydrate diagnostic sensitivity 
          powder.
460.58  Methacycline hydrochloride diagnostic sensitivity powder.
460.64  Minocycline hydrochloride powder for

[[Page 1020]]

          microbial susceptibility testing.
460.66  Oleandomycin phosphate diagnostic sensitivity powder.
460.70  Oxytetracycline hydrochloride diagnostic sensitivity powder.
460.75  Potassium penicillin G diagnostic sensitivity powder.
460.79  Polymyxin B sulfate diagnostic sensitivity powder.
460.86  Spectinomycin hydrochloride powder for microbial susceptibility 
          testing.
460.89  Streptomycin sulfate diagnostic sensitivity powder.
460.93  Tetracycline hydrochloride diagnostic sensitivity powder.

                  Subpart C--Susceptibility Test Panels

460.100  Antimicrobial susceptibility test panels.
460.110  Ampicillin concentrated stock solutions for use in 
          antimicrobial susceptibility test panels.
460.113  Carbenicillin concentrated stock solutions for use in 
          antimicrobial susceptibility test panels.
460.116  Cephalothin concentrated stock solutions for use in 
          antimicrobial susceptibility test panels.
460.119  Chloramphenicol concentrated stock solutions for use in 
          antimicrobial susceptibility test panels.
460.122  Clindamycin concentrated stock solutions for use in 
          antimicrobial susceptibility test panels.
460.125  Colistin concentrated stock solutions for use in antimicrobial 
          susceptibility test panels.
460.128  Erythromycin concentrated stock solutions for use in 
          antimicrobial susceptibility test panels.
460.131  Gentamicin concentrated stock solutions for use in 
          antimicrobial susceptibility test panels.
460.134  Kanamycin concentrated stock solutions for use in antimicrobial 
          susceptibility test panels.
460.137  Methicillin concentrated stock solutions for use in 
          antimicrobial susceptibility test panels.
460.140  Penicillin G concentrated stock solutions for use in 
          antimicrobial susceptibility test panels.
460.146  Tetracycline concentrated stock solutions for use in 
          antimicrobial susceptibility test panels.
460.149  Tobramycin concentrated stock solutions for use in 
          antimicrobial susceptibility test panels.
460.152  Trimethoprim concentrated stock solutions for use in 
          antimicrobial susceptibility test panels.
460.153  Sulfamethoxazole concentrated stock solutions for use in 
          antimicrobial susceptibility test panels.

    Authority:  21 U.S.C. 357.

    Source:  39 FR 19181, May 30, 1974, unless otherwise noted.



                     Subpart A--Susceptibility Discs



Sec. 460.1  Certification procedures for antibiotic susceptibility discs.

    (a) Standards of identity, strength, quality, and purity. Antibiotic 
susceptibility discs are round flat discs that have a diameter of one-
fourth inch and are made of clear absorbent paper containing antibiotic 
compounds. They are capable of absorbing moisture rapidly and the 
antibiotic is evenly distributed. The thickness is sufficient to assure 
rigidity and to have permitted the complete absorption of an adequate 
volume of antibiotic solution (approximately 0.02 milliliter). The 
identity of each disc is signified either by a color or by means of an 
identifying sign. The absorbent paper and dye or ink used must not 
affect either bacterial growth or the antibiotic. Each disc shall have a 
uniform potency that is equivalent to that contained in a standard disc 
prepared with one of the following quantities of antibiotic drugs:

    Ampicillin: 10 mcg.
    Bacitracin: 10 units.
    Carbenicillin: 50 mcg.
    Cefamandole:30mcg.
    Cefoxitin: 30 mcg
    Cephalothin: 30 mcg.
    Chloramphenicol: 30 mcg.
    Clindamycin: 2 mcg.
    Colistin: 10 mcg.
    Erythromycin: 15 mcg.
    Gentamicin: 10 mcg.
    Kanamycin: 30 mcg.
    Methicillin: 5 mcg.
    Neomycin: 30 mcg.
    Novobiocin: 30 mcg.
    Oleandomycin: 15 mcg.
    Penicillin G: 10 units.
    Polymyxin B: 300 units.
    Rifampin: 5 mcg.
    Streptomycin: 10 mcg.
    Tetracycline: 30 mcg.
    Tobramycin: 10 mcg.
    Vancomycin: 30 mcg.


The standard discs used to determine the potency shall be made of paper 
as described in Sec. 460.6(d). Each antibiotic compound used to 
impregnate such standard discs shall be equilibrated in terms of the 
working standard designated by the Commissioner for use in determining 
the potency or purity of such antibiotic.

[[Page 1021]]

    (b) Packaging. The immediate container shall be a tight container as 
defined by the U.S.P. and shall be of such composition as will not cause 
any change in the strength, quality, or purity of the contents beyond 
any limit therefor in applicable standards, except that minor changes so 
caused that are normal and unavoidable in good packaging, storage, and 
distribution practice shall be disregarded. Each immediate container may 
contain a desiccant, and each may be packaged in combination with 
containers of suitable discs of drugs other than those described in 
paragraph (a) of this section. Such other discs shall be suitable only 
if the manufacturer and packer have submitted to the Commissioner 
information of the kind described in Sec. 431.17 of this chapter, and 
such information has been accepted by the Commissioner.
    (c) Labeling. Each package of discs shall bear on its label or 
labeling, as hereinafter indicated, the following:
    (1) On the outside wrapper or container and the immediate container:
    (i) The batch mark.
    (ii) The name and potency of each disc in the batch according to the 
following:

                                                                        
------------------------------------------------------------------------
                                              Content of antibiotic in  
               Name of disc                 micrograms or units per disc
------------------------------------------------------------------------
Ampicillin-class disc.....................  10 mcg. ampicillin.         
Bacitracin disc...........................  10 units bacitracin.        
Carbenicillin disc........................  50 mcg. carbenicillin.      
Cefamandole disc..........................  30 mcg. cefamandole.        
Cefoxitin disc............................  30 mcg. cefoxitin.          
Cephalosporin-class disc..................  30 mcg. cephalothin.        
Chloramphenicol disc......................  30 mcg. chloramphenicol.    
Colistin disc.............................  10 mcg. colistin.           
Erythromycin disc.........................  15 mcg. erythromycin.       
Gentamicin disc...........................  10 mcg. gentamicin.         
Kanamycin disc............................  30 mcg. kanamycin.          
Lincomycin-class disc.....................  2 mcg. clindamycin.         
Neomycin disc.............................  30 mcg. neomycin.           
Novobiocin disc...........................  30 mcg. novobiocin.         
Oleandomycin disc.........................  15 mcg. oleandomycin.       
Penicillin-class disc.....................  10 units penicillin G.      
Penicillinase-resistant penicillin-class    5 mcg. methicillin.         
 disc.                                                                  
Polymyxin B disc..........................  300 units polymyxin B.      
Rifampin disc.............................  5 mcg. rifampin.            
Streptomycin-class disc...................  10 mcg. streptomycin.       
Tetracycline-class disc...................  30 mcg. tetracycline.       
Tobramycin disc...........................  10 mcg. tobramycin.         
Vancomycin disc...........................  30 mcg. vancomycin.         
------------------------------------------------------------------------

    (iii) The statement ``Expiration date ----------'', the blank being 
filled in with the date that is 6 months after the month during which 
the batch was certified, except that the blank may be filled in with a 
date that is 12, 18, 24, 30, 36, 42, 48, 54, or 60 months after the 
month during which the batch was certified if the person who requests 
certification has submitted to the Commissioner results of tests and 
assays showing that such drug as prepared by him is stable for such 
longer period of time. If it is a packaged combination of discs of two 
or more drugs, its outside wrapper shall bear only one expiration date, 
and that date shall be the date that is required for the shortest dated 
discs contained in the package.
    (iv) The statement ``For laboratory use only''.
    (2) On the circular or other labeling within or attached to the 
package, adequate directions for the use of such discs, including the 
following recommended method:

                  Standardized Disc Susceptibility Test

                           directions for use

    Quantitative methods that require the measurement of zone sizes give 
the most precise estimates of antibiotic susceptibilities. The following 
outline describes such a procedure. Minor variations from this procedure 
may be used if the resulting procedure is standardized according to the 
results obtained in the laboratory from adequate studies with control 
cultures.

               a. preparation of culture medium and plates

    1. Melt previously prepared and sterilized Mueller-Hinton agar 
medium and cool to 45 deg.-50 deg. C.
    2. For the purpose of testing certain fastidious organisms such as 
streptococci and Haemophilus species, 5 percent defibrinated human, 
horse, or sheep blood may be added to the above medium which is 
``chocolatized'' when indicated.
    3. To prepare the plates, pour the melted medium into Petri dishes 
on a level surface to a depth of 4 millimeters.
    4. Let the medium harden and allow to stand long enough for excess 
moisture to evaporate. (For this purpose plates may be placed in an 
incubator at 35 deg.-37 deg. C. for 15-30 minutes or allowed to stand 
somewhat longer at room temperature.) There should be no moisture 
droplets on the surface of the medium or on the petri dish covers. The 
pH of the solidified medium should be 7.2-7.4. Satisfactory plates may 
be used immediately or refrigerated. Plates may be used as long

[[Page 1022]]

as the surface is moist and there is no sign of deterioration.
    Note: Commercially prepared agar plates meeting the above 
specifications may be used.

                       b. preparation of inoculum

    1. Select four or five similar colonies.
    2. Transfer these colonies (obtained by touching the top of each 
colony in turn with a wire loop) in turn to a test tube containing about 
5 milliliters of a suitable liquid medium such as soybean-casein digest 
broth, U.S.P.
    3. Incubate the tube at 35 deg.-37 deg. C. long enough (2 to 8 
hours) to produce an organism suspension with moderate cloudiness. At 
that point the inoculum density of the suspension should be controlled 
by diluting it, or a portion of it, with sterile saline to obtain a 
turbidity equivalent to that of a freshly prepared turbidity standard 
obtained by adding 0.5 milliliter of 1.175 percent barium chloride 
dihydrate (BaCl22H2O) solution to 99.5 milliliters 
of 0.36 N (1.0 percent) sulfuric acid. Other suitable methods for 
standardizing inoculum density may be used; for example, a photometric 
method. In some cases it may be possible to get an adequate inoculum 
density in the tube even without incubation.
    Note: Extremes in inoculum density should be avoided. Undiluted 
overnight broth culture should never be used for streaking plates.

                        c. inoculating the plates

    1. Dip a sterile cotton swab on a wooden applicator into the 
properly diluted inoculum. Remove excess inoculum from the swab by 
rotating it several times with firm pressure on the inside wall of the 
test tube above the fluid level.
    2. Streak the swab over the entire sterile agar surface of a plate. 
Streaking successively in three different directions is recommended to 
obtain an even inoculum.
    3. Replace the plate top and allow the inoculum to dry for 3 to 5 
minutes.
    4. Place the susceptibility discs on the inoculated agar surface and 
with sterile forceps, or needle tip flamed and cooled between each use, 
gently press down each disc to insure even contact. Space the discs 
evenly so that they are no closer than 10 to 15 millimeters to the edge 
of the petri dish and sufficiently separated from each other to avoid 
overlapping zones of inhibition. (Spacing may be accomplished by using a 
disc dispenser or by putting the plate over a pattern to guide the 
placement of discs.) Within 30 minutes, place the plate in an incubator 
under aerobic conditions at a constant temperature in the range of 
35 deg.-37 deg. C.
    5. Read the plate after overnight incubation or, if rapid results 
are desired, the diameters of the zone of inhibition may be readable 
after 6 to 8 hours incubation. In the latter case, the results should be 
confirmed by also reading the results after overnight incubation.
    Note: Microbial growth on the plate should be just or almost 
confluent. If only isolated colonies are present the inoculum was too 
light and the test should be repeated.
    Modifications of the inoculation procedure described in 1-3 above, 
such as the use of the agar overlay method described in Barry, A. L., 
Garcia, F., and Thrupp, L. D.: ``An Improved Single-disk Method for 
Testing the Antibiotic Susceptibility of Rapidly-growing Pathogens.'' 
Amer. J. Clin. Pathol. 53:149-58, 1970,* a copy of which is on file with 
the Office of the Federal Register, may be used if the procedure is 
standardized to produce results with the control cultures that are 
equivalent to those obtained with the recommended cotton swab streak 
method.

    *Copies may be obtained from: J. B. Lippincott Company, Attn: 
Circulation Manager, East Washington Square, Philadelphia, PA 19105.
---------------------------------------------------------------------------

                          d. reading the plates

    Measure and record the diameter of each zone (including the diameter 
of the disc) to the closest millimeter, reading to the point of complete 
inhibition as judged by the unaided eye. Preferably, read from the 
underside of the plate without removing the cover, using a ruler, 
calipers, transparent plastic gage, or other device. A mechanical zone 
reader may be used. If blood agar is used, measure the zones from the 
surface with the cover removed from the plate.

                     e. interpretation of zone sizes

    Interpret the susceptibility according to the following table:

                                                                                                                
----------------------------------------------------------------------------------------------------------------
                                                               Diameter (millimeters) of zone of inhibition     
            Antibiotic                  Disc content     -------------------------------------------------------
                                                               Resistant       Intermediate      Susceptible    
----------------------------------------------------------------------------------------------------------------
Ampicillin \1\ when testing gram-   10 mcg..............  11 or less.........        12-13   14 or more.        
 negative microorganisms and                                                                                    
 enterococci.                                                                                                   
Ampicillin \1\ when testing         10 mcg..............  20 or less.........        21-28   29 or more.        
 staphylococci and penicillin G--                                                                               
 susceptible micro-organisms.                                                                                   
Ampicillin \1\ when testing         10 mcg..............  19 or less.........  ............  20 or more.        
 Haemophilus species.                                                                                           

[[Page 1023]]

                                                                                                                
Bacitracin........................  10 units............  8 or less..........         9-12   13 or more.        
Carbenicillin when testing Proteus  50 mcg..............  17 or less.........        18-22   23 or more.        
 species and Escherichia coli.                                                                                  
Carbenicillin when testing          50 mcg..............  12 or less.........        13-14   15 or more.        
 Pseudomonas aeruginosa.                                                                                        
Cefamandole \12\..................  30mcg...............  14 or less.........        16-17   18 or more.        
Cefoxitin \11\....................  30 mcg..............  14 or less.........        15-17   18 or more.\11\    
Cephalothin when reporting          30 mcg..............  14 or less.........        15-17   18 or more.\2\     
 susceptibility to cephalothin,                                                                                 
 cephaloridine, and cephalexin.                                                                                 
Cephalothin when reporting          30 mcg..............  14 or less.........  ............  15 or more.        
 susceptibility to cephaloglycin.                                                                               
Chloramphenicol...................  30 mcg..............  12 or less.........        13-17   18 or more.        
Clindamycin \3\ when reporting      2 mcg...............  14 or less.........        15-16   17 or more.        
 susceptibility to clindamycin.                                                                                 
Clindamycin when reporting          2 mcg...............  16 or less.........        17-20   21 or more.        
 susceptibility to lincomycin.                                                                                  
Colistin..........................  10 mcg..............  8 or less..........         9-10   11 or more.        
Erythromycin......................  15 mcg..............  13 or less.........        14-17   18 or more.        
Gentamicin........................  10 mcg..............  12 or less.........  ............  13 or more.        
Kanamycin.........................  30 mcg..............  13 or less.........        14-17   18 or more.        
Methicillin \5\...................  5 mcg...............  9 or less..........        10-13   14 or more.        
Neomycin..........................  30 mcg..............  12 or less.........        13-16   17 or more.        
Novobiocin........................  30 mcg..............  17 or less.........        18-21   22 or more.\6\     
Oleandomycin \7\..................  15 mcg..............  11 or less.........        12-16   17 or more.        
Penicillin G when testing           10 units............  20 or less.........        21-28   29 or more.        
 staphylococci \8\.                                                                                             
Penicillin G when testing other     10 units............  11 or less.........    \9\ 12-21   22 or more.        
 micro-organisms \8\.                                                                                           
Polymyxin B.......................  300 units...........  8 or less..........         9-11   12 or more.\4\     
Rifampin when testing Neisseria     5 mcg...............  24 or less.........  ............  25 or more.        
 meningitidis susceptibility only.                                                                              
Streptomycin......................  10 mcg..............  11 or less.........        12-14   15 or more.        
Tetracycline \10\.................  30 mcg..............  14 or less.........        15-18   19 or more.        
Tobramycin........................  10 mcg..............  11 or less.........        12-13   14 or more.        
Vancomycin........................  30 mcg..............  9 or less..........        10-11   12 or more.        
----------------------------------------------------------------------------------------------------------------
\1\ The ampicillin disc is used for testing susceptibility to both ampicillin and hetacillin.                   
\2\ Staphylococci exhibiting resistance to the penicillinase-resistant penicillin class discs should be reported
  as resistant to cephalosporin class antibiotics. The 30 mcg. cephalothin disc cannot be relied upon to detect 
  resistance of methicillin-resistant staphylococci to cephalosporin class antibiotics.                         
\3\ The clindamycin disc is used for testing susceptibility to both clindamycin and lincomycin.                 
\4\ Colistin and polymyxin B diffuse poorly in agar and the accuracy of the diffusion method is thus less than  
  with other antibiotics. Resistance is always significant but when treatment of systemic infections due to     
  susceptible strains is considered, it is wise to confirm the results of a diffusion test with a dilution      
  method.                                                                                                       
\5\ The methicillin disc is used for testing susceptibility to all penicillinase-resistant penicillins; that is,
  methicillin, cloxacillin, dicloxacillin, oxacillin, and nafcillin.                                            
\6\ Not applicable to medium that contains blood.                                                               
\7\ The oleandomycin disc is used for testing susceptibility to oleandomycin and troleandomycin.                
\8\ The penicillin G disc is used for testing susceptibility to all penicillinase-susceptible penicillins except
  ampicillin and carbenicillin; that is, penicillin G, phenoxymethyl penicillin, and phenethicillin.            
\9\ This category includes some organisms such as enterococci and gram-negative bacilli that may cause systemic 
  infections treatable with high doses of penicillin G. Such organisms should only be reported susceptible to   
  penicillin G and not to phenoxymethyl penicillin or phenethicillin.                                           
\10\ The tetracycline disc is used for testing susceptibility to all tetracyclines; that is, chlortetracycline, 
  demeclocycline, doxycycline, methacycline, oxytetracycline, rolitetracycline, minocycline, and tetracycline.  
\11\ The cefoxitin disc should not be used for testing susceptibility of other cephalosporins.                  
\12\ The cefamandole disc should not be used for testing susceptibility of other cephalosporins.                

                         f. reference organisms

    1. Maintain stock cultures of Staphylococcus aureus (ATCC 25923) 
1 and Escherichia coli (ATCC 25922). \1\
    2. Test these reference organisms daily by the above procedure using 
antibiotic discs representative of those to be used in the testing of 
clinical isolates.
    3. The individual values of zone sizes for the control organisms can 
be expected to fall in the ranges indicated in the following table:

                                                                                                                
----------------------------------------------------------------------------------------------------------------
                                                                                       Individual tests         
                                                                             -----------------------------------
                                                                                 Zone diameter in               
                                                                                    millimeters        Permitted
                  Antibiotic                            Disc content         ------------------------ millimeter
                                                                                With S.               difference
                                                                                aureus      With E.   ATCC 25923-
                                                                              ATTC 25923   coli ATCC  ATCC 25922
                                                                                  \1\      25922 \1\            
----------------------------------------------------------------------------------------------------------------
Ampicillin...................................  10 mcg.......................       24-35       15-20        7-17
Bacitracin...................................  10 units.....................       17-22  ..........  ..........
Cefamandole..................................  30 mcg.......................       28-34       24-31      -1-6.8
Cefoxitin....................................  30 mcg.......................       24-32       25-30         3-4
Cephalothin..................................  30 mcg.......................       25-37       18-23        5-16

[[Page 1024]]

                                                                                                                
Chloramphenicol..............................  30 mcg.......................       19-26       21-27        -4-1
Clindamycin..................................  2 mcg........................       23-29  ..........  ..........
Colistin.....................................  10 mcg.......................  ..........       11-15  ..........
Erythromycin.................................  15 mcg.......................       22-30        8-14       10-19
Gentamicin...................................  10 mcg.......................       19-27       19-26        -2-3
Kanamycin....................................  30 mcg.......................       19-26       17-25        -1-4
Methicillin..................................  5 mcg........................       17-22  ..........  ..........
Neomycin.....................................  30 mcg.......................       18-26       17-23         0-3
Novobiocin...................................  30 mcg.......................       22-31  ..........  ..........
Oleandomycin.................................  15 mcg.......................       19-28  ..........  ..........
Penicillin G.................................  10 units.....................       26-37  ..........  ..........
Polymyxin B..................................  300 units....................        7-13       12-16       -7--2
Streptomycin.................................  10 mcg.......................       14-22       12-20        -1-5
Tobramycin...................................  10 mcg.......................       19-29       18-26  ..........
Tetracycline.................................  30 mcg.......................       19-28       18-25         0-6
Vancomycin...................................  30 mcg.......................       15-19  ..........  ..........
----------------------------------------------------------------------------------------------------------------
\1\ Available from: American Type Culture Collection, 12301 Parklawn Dr., Rockville, Md. 20852.                 

                      g. limitations of the method

    The method of interpretation described in E above applies to rapidly 
growing pathogens and should not be applied to slowly growing organisms. 
The latter show larger zones of inhibition than those given in the 
table. Susceptibility of gonococci to penicillin, and of slow-growing 
strains, e.g., Bacteroides species and fastidious anaerobes to any 
antibiotic, should be determined by the broth-dilution or agar-dilution 
method unless specifically standardized diffusion tests are used.

    (d) Requests for certification; samples. (1) In addition to 
complying with the requirements of Sec. 431.1 of this chapter, a person 
who requests certification of a batch of antibiotic susceptibility discs 
shall submit with his request a statement showing the batch mark, the 
number of packages of each size in such batch, and, unless it was 
previously submitted, the date on which the latest assay of the 
antibiotic used in making such batch was completed, the potency of each 
disc, the quantity of each ingredient used in making the batch, the date 
on which the latest assay of the drug comprising such batch was 
completed, and a statement that each ingredient used in making the batch 
conforms to the requirements prescribed therefor by this section.
    (2) Such person shall submit in connection with his request results 
of the tests and assays made by him on an accurately representative 
sample of the batch for potency.
    (3) Such person shall submit in connection with his request an 
accurately representative sample of the batch consisting of one disc for 
each 5,000 discs in the batch, but in no case less than 36 discs 
collected by taking single discs at intervals throughout the entire time 
of packaging the batch so that the quantities packaged during the 
intervals are approximately equal.
[39 FR 19181, May 30, 1974, as amended at 41 FR 7093, Feb. 17, 1976; 41 
FR 35061, Aug. 19, 1976; 44 FR 10376, Feb. 20, 1979; 44 FR 20666, Apr. 
6, 1979]



Sec. 460.6  Tests and methods of assay for potency of antibiotic susceptibility discs.

    (a) Culture media. Use ingredients that conform to the standards 
prescribed by the United States Pharmacopeia or The National Formulary. 
In lieu of preparing the media from the individual ingredients, they may 
be made from a dehydrated mixture which, when reconstituted with 
distilled water, has the same composition as such media. Minor 
modification of the specified individual ingredients is permissible if 
the resulting media possess growth-promoting properties at least equal 
to the media described.
    (1) Medium A:

                                                                        
                                                                        
                                                                        
Peptone.....................................................     6.0 gm.
Pancreatic digest of casein.................................     4.0 gm.
Yeast extract...............................................     3.0 gm.

[[Page 1025]]

                                                                        
Beef extract................................................     1.5 gm.
Dextrose....................................................     1.0 gm.
Agar........................................................    15.0 gm.
Distilled water, q.s........................................     1,000.0
                                                                     ml.
pH 6.5 to 6.6 after sterilization.                                      
                                                                        

    (2) Medium B. Same as medium A, except that it also contains 300 
milligrams of hydrated manganese sulfate per liter.
    (3) Medium C. Same as medium A except that the final pH is adjusted 
from 7.9 to 8.1 after sterilization.
    (4) Medium D:

                                                                        
                                                                        
                                                                        
Peptone.....................................................     5.0 gm.
Yeast extract...............................................     1.5 gm.
Beef extract................................................     1.5 gm.
Sodium chloride.............................................     3.5 gm.
Dextrose....................................................     1.0 gm.
Dipotassium phosphate.......................................    3.68 gm.
Potassium dihydrogen phosphate..............................    1.32 gm.
Disilled water, q.s.........................................     1,000.0
                                                                     ml.
pH 7.0 after sterilization                                              
                                                                        

    (5) Medium E:

                                                                        
                                                                        
                                                                        
Peptone.....................................................     6.0 gm.
Yeast extract...............................................     3.0 gm.
Beef extract................................................     1.5 gm.
Agar........................................................    15.0 gm.
Distilled water, q.s........................................     1,000.0
                                                                     ml.
pH 6.5 to 6.6 after sterilization.                                      
                                                                        

    (6) Medium F:

                                                                        
                                                                        
                                                                        
Pancreatic digest of casein.................................    17.0 gm.
Papaic digest of soybean....................................     3.0 gm.
Sodium chloride.............................................     5.0 gm.
Dipotassium phosphate.......................................     2.5 gm.
Dextrose....................................................     2.5 gm.
Agar........................................................    20.0 gm.
Distilled water, q.s........................................     1,000.0
                                                                     ml.
pH 7.3 after sterilization.                                             
                                                                        

    (7) Medium G. Same as medium F except for the following:

                                                                        
                                                                        
                                                                        
Agar........................................................    12.0 gm.
Polysorbate 80 (Sterile)....................................    10.0 gm.
Add polysorbate 80 after boiling.                                       
                                                                        

    (8) Medium H:

                                                                        
                                                                        
                                                                        
Peptone.....................................................     9.4 gm.
Yeast extract...............................................     4.7 gm.
Beef extract................................................     2.4 gm.
Sodium chloride.............................................    10.0 gm.
Dextrose....................................................    10.0 gm.
Agar........................................................    23.5 gm.
Distilled water, q.s........................................     1,000.0
                                                                     ml.
pH 6.0 to 6.2 after sterilization.                                      
                                                                        

    (9) Medium I:

                                                                        
                                                                        
                                                                        
Peptone.....................................................     6.0 gm.
Yeast extract...............................................     3.0 gm.
Beef extract................................................     1.5 gm.
Dextrose....................................................     1.0 gm.
Agar........................................................    15.0 gm.
Distilled water, q.s........................................     1,000.0
                                                                     ml.
pH 6.6 after sterilization.                                             
                                                                        

    (10) Medium J:

                                                                        
                                                                        
                                                                        
Pancreatic digest of casein.................................    15.0 gm.
Papaic digest of soybean....................................     5.0 gm.
Sodium chloride.............................................     5.0 gm.
Agar........................................................    15.0 gm.
Distilled water, q.s........................................     1,000.0
                                                                     ml.
pH 7.3 after sterilization.                                             
                                                                        

    (11) Medium K:

                                                                        
                                                                        
                                                                        
Pancreatic digest of casein.................................    17.0 gm.
Papaic digest of soybean....................................     3.0 gm.
Sodium chloride.............................................     5.0 gm.
Dipotassium phosphate.......................................     2.5 gm.
Dextrose....................................................     2.5 gm.
Distilled water, q.s........................................     1,000.0
                                                                     ml.
pH 7.3 after sterilization.                                             
                                                                        

    (12) Medium L:

                                                                        
                                                                        
                                                                        
Agar agar...................................................      15 gm.
Distilled water, q.s........................................     1,000.0
                                                                     ml.
                                                                        

    (13) Medium M:

                                                                        
                                                                        
                                                                        
Beef, inclusion from........................................     300 gm.
Acid hydrolysate of casein..................................    17.5 gm.
Soluble starch..............................................     1.5 gm.
Agar........................................................      15 gm.
Distilled water, q.s........................................     1,000.0
                                                                     ml.
pH 7.4 after sterilization.                                             
                                                                        

    (14) Medium N:

                                                                        
                                                                        
                                                                        
Infusion from beef..........................................   300.0 gm.
Acid hydrolysate of casein..................................    17.5 gm.
Starch......................................................     1.5 gm.
Distilled water, q.s........................................     1,000.0
                                                                     ml.
pH 7.4 after sterilization.                                             
                                                                        

    (15) Medium O:

                                                                        
                                                                        
                                                                        
Calf brains, infusion from..................................   200.0 gm.
Beef heart, infusion from...................................   250.0 gm.
Pancreatic digest of gelatin................................    10.0 gm.
Dextrose....................................................     2.0 gm.
Sodium chloride.............................................     5.0 gm.
Sodium phosphate dibasic (Na2HPO4)..........................     2.5 gm.
Distilled water, q.s........................................     1,000.0
                                                                     ml.
pH 7.4 after sterilization.                                             
                                                                        

    (16) Medium P. Same as medium J with 5 percent defibrinated sheep 
blood added.
    (17) Medium Q:

                                                                        
                                                                        
                                                                        
Pancreatic digest of gelatin................................    17.0 gm.
Pancreatic digest of casein plus equal part of peptic digest            
 of animal tissues..........................................     3.0 gm.
Lactose.....................................................    10.0 gm.
Bile salts mixture..........................................     1.5 gm.
Sodium chloride.............................................     5.0 gm.
Agar........................................................    13.5 gm.
Neutral red.................................................    0.03 gm.
Crystal violet..............................................   0.001 gm.
Distilled water, q.s........................................     1,000.0
                                                                     ml.
pH 7.1 after sterilization.                                             
                                                                        

    (b) Preparation of test organism suspensions--(1) Suspension 1. 
Staphylococcus aureus (ATCC 6538P)1 is maintained and

[[Page 1026]]

grown on medium A. Wash the organisms from an agar slant, incubated for 
24 hours at 32 deg. C. to 35 deg. C., with 3.0 milliliters of sterile 
sodium chloride solution onto the agar surface of a Roux bottle 
containing 300 milliliters of medium A. Spread the suspension of 
organisms over the entire agar surface with the aid of sterile glass 
beads. Incubate 24 hours at 32 deg. C. to 35 deg. C. Wash the resulting 
growth from the agar surface with about 50 milliliters of sterile sodium 
chloride solution. Standardize this stock suspension by determining the 
dilution that will permit 20 percent light transmission. Store the stock 
suspension in the refrigerator (1 week) and use the indicated dilution 
prepared daily.
---------------------------------------------------------------------------

    1 Available from: American Type Culture Collection, 12301 
Parklawn Drive, Rockville, MD 20852.
---------------------------------------------------------------------------

    (2) Suspension 2. Follow the procedure described for suspension 1, 
except standardize the bulk suspension so that a 1:10 dilution in saline 
solution gives 20 percent light transmission. In this case, the bulk 
suspension, and not the 1:10 dilution of it, is used for the inoculum.
    (3) Suspension 3. The test organism is Staphylococcus aureus (ATCC 
13150).1 Follow the procedure described for suspension 1, but 
determine how much the bulk suspension should be diluted to obtain a 
suspension permitting 80 percent light transmission. Use the indicated 
dilution prepared daily for the inoculum for the plates.
    (4) Suspension 4. Sarcina lutea (ATCC 9341) 1 is 
maintained on agar slants of medium A and transferred to fresh slants 
approximately every 2 weeks. This culture is incubated overnight at 
26 deg. C., and then stored in the refrigerator. Prepare an inoculum for 
the plates as follows: Streak an agar slant heavily with the test 
organism and incubate for 24 hours at 26 deg. C. Wash the growth from 
the slant with 3 milliliters to 4 milliliters of medium D, and transfer 
to the surface of a Roux bottle containing 300 milliliters of medium A. 
Spread the suspension evenly over the entire surface with the aid of 
sterile glass beads. Incubate for 24 hours at 26 deg. C. Wash the growth 
from the agar surface with 15 milliliters of medium D. If an aliquot of 
this bulk suspension when diluted 1:10 with medium D gives 10 percent 
light transmission, the bulk suspension is satisfactory for use. It may 
be necessary to adjust the bulk suspension by dilution so that an 
aliquot of the adjusted suspension when diluted 1:10 will give the 
desired 10 percent light transmission. The adjusted bulk suspension 
only, and not the 1:10 dilution of it, is used in preparing the 
inoculum. Store the stock suspension in the refrigerator and use for 2 
weeks.
    (5) Suspension 5. Bacillus subtilis (ATCC 6633) 1 is 
maintained on agar medium A and transferred to a fresh slant every 
month. To prepare the spore suspension, inoculate a fresh slant of agar 
medium A with the test organism and incubate at 37 deg. C. for 16 hours 
to 24 hours. Wash the culture from the slant with 3 milliliters of 
sterile sodium chloride solution onto the surface of a Roux bottle 
containing 300 milliliters of agar medium B. Incubate for 5 days at 
37 deg. C. Suspend the growth in 50 milliliters of sterile saline 
solution, centrifuge, and decant the supernatant liquid. Reconstitute 
the sediment and heat-shock the suspension by heating for 30 minutes at 
70 deg. C. Store the spore suspension in the refrigerator. It may be 
kept several months. Light transmission is not used for standardization.
    (6) Suspension 6. Staphylococcus epidermidis (ATCC 12228) 1 
is maintained on medium A and transferred to a fresh slant once a week. 
Inoculate a fresh slant of medium A with the test organism and incubate 
at 32 deg. C. to 35 deg. C. for 24 hours. Wash the culture from the 
slant with 3 milliliters of sterile sodium chloride solution onto the 
surface of a Roux bottle containing 300 milliliters of medium A. 
Incubate at 32 deg. C. to 35 deg. C. for 24 hours. Wash the resulting 
growth from the agar surface with about 50 milliliters of sterile sodium 
chloride solution. Standardize this stock suspension by determining the 
dilution that will give 80 percent light transmission. Store the stock 
suspension in the refrigerator (1 week) and use the indicated dilution 
prepared daily for the inoculum for the plates.
    (7) Suspension 7. Bordetella bronchiseptica (ATCC 4617) 1 
is maintained on medium F and transferred to a fresh slant every 2 
weeks. To prepare a stock suspension inoculate a fresh slant of medium F 
and incubate at 37 deg. C. for 16 hours to 24 hours. Wash the

[[Page 1027]]

culture from this slant with 3 milliliters of sterile distilled water 
onto the surface of a Roux bottle containing 300 milliliters of medium 
F, and incubate 24 hours at 37 deg. C. Wash off the growth with 50 
milliliters of sterile distilled water and standardize the resulting 
stock suspension by determining the dilution that will give 50 percent 
light transmission. Store the stock suspension in the refrigerator (2 
weeks), and use the indicated dilution prepared daily for the inoculum 
for the plates.
    (8) Suspension 8. Saccharomyces cerevisiae (ATCC 9763) 1 
is maintained on slants of medium H and transferred once a week. After 
transfer, the culture is incubated at 37 deg. C. for 24 hours and then 
kept refrigerated. Wash the organism from a freshly incubated agar slant 
with 3 milliliters of sterile saline solution onto the agar surface in a 
Roux bottle containing 300 milliliters of medium H. Spread the 
suspension of organisms over the entire agar surface with the aid of 
sterile glass beads. Incubate for 24 hours at 37 deg. C. and then wash 
the resulting growth from the agar surface with about 25 milliliters of 
sterile saline solution. Store the suspension in the refrigerator and 
use for 1 month.
    (9) Suspension 9. Follow the procedure described for suspension 1, 
except determine how much the bulk suspension should be diluted to 
obtain a suspension permitting 80 percent light transmission. Use the 
indicated dilution, prepared daily, for the inoculum for the plates.
    (10) Suspension 10: Klebsiella pneumoniae (ATCC 10031), 1 
noncapsulated, is maintained on medium A and transferred to a fresh 
slant once a week. Inoculate a fresh slant of medium A with the test 
organism and incubate overnight at 32 deg. C.-35 deg. C. Wash the 
culture from the slant with 3 milliliters of sterilized U.S.P. saline 
T.S. onto the surface of a Roux bottle containing 300 milliliters of 
medium A. Incubate at 32 deg. C.-35 deg. C. for 24 hours. Wash the 
resulting growth from the agar surface with about 50 milliliters of 
sterilized U.S.P. saline T.S. If an aliquot of this bulk suspension when 
diluted 1:9 with saline solution gives 40 percent light transmission, 
the bulk suspension is satisfactory for use. It may be necessary to 
adjust the bulk suspension by dilution so that an aliquot of the 
adjusted suspension when diluted 1:9 will give the desired 40 percent 
light transmission. The adjusted bulk suspension (not the 1:9 dilution) 
is used in preparing the inoculum. Store the suspension in the 
refrigerator and use for no more than 1 week.
---------------------------------------------------------------------------

    1 Available from: American Type Culture Collection, 12301 
Parklawn Drive, Rockville, MD 20852.
---------------------------------------------------------------------------

    (11) Suspension 11 Streptococcus fecalis (ATCC 14506) 1 
is maintained on medium E and transferred to a fresh agar slant once a 
week. After transfer, the culture is incubated at 37 deg. C. for 24 
hours and then kept refrigerated. Transfer from a freshly incubated agar 
slant to a tube containing 10 milliliters of culture medium described in 
Sec. 147.3(b)(1). Incubate the broth culture for 16 to 18 hours at 
37 deg. C. and store in the refrigerator. This culture may be used for 
no more than 1 week.

The light transmission values referred to in this paragraph were 
determined with a Lumetron Model 400-A photoelectric colorimeter at a 
wavelength of 650 millimicrons. If other instruments are used, different 
light transmission readings will probably be obtained. The values given 
are to be used as guides in this paragraph.
    (12) Suspension 12. Pseudomonas aeruginosa (ATCC 25619) 1 
is maintained and grown on medium J and transferred to a fresh agar 
slant once a week. Inoculate a fresh slant of medium J with the test 
organism and incubate at 37 deg. C. for 24 hours. Transfer the culture 
from this slant with sterile glass beads onto the agar surface of a Roux 
bottle containing 300 milliliters of medium J. Spread the organisms over 
the entire agar surface with the aid of the glass beads. Incubate 24 
hours at 37 deg. C. Wash the resulting growth from the agar surface with 
about 30 milliliters of medium K. Do not standardize the suspension. 
Store the stock suspension under refrigeration and use for 2 weeks.
    (13) Suspension 13. Escherichia coli (ATCC 29214) 1 is 
maintained and grown on medium M. Wash the organisms from an agar slant, 
incubated for 24 hours at 37 deg. C, with 3 milliliters of

[[Page 1028]]

sterilized U.S.P. saline T.S. onto the surface of a Roux bottle 
containing 250 milliliters of medium M. Spread the suspension of 
organisms over the entire agar surface with the aid of sterile glass 
beads. Incubate for 24 hours at 37 deg. C and then wash the resulting 
growth from the agar surface with 50 milliliters of sterilized U.S.P. 
saline T.S. Store the suspension in the refrigerator and use for 2 
weeks.
    (14) Suspension 14. The test organism is Staphylococcus aureus (ATCC 
29213).
    (i) Stock culture. Transfer a lyophilized culture into medium K in a 
sterile container and incubate at 37 deg. C for 24 hours. Streak the 
culture onto the solidified agar surface of a plate containing medium P 
and incubate the plate at 37 deg. C for 24 hours. Transfer 5 to 10 
colonies into 3 milliliters of medium O in a sterile container and 
incubate at 37 deg. C for 24 hours. Add 3 milliliters of sterile 
glycerol or 3 milliliters of sterile rabbit serum to the broth culture, 
mix well and pour the contents into a sterile flask containing a layer 
of sterile glass beads. Rotate the flask to coat the beads with the 
culture mixture and aseptically aspirate all the excess liquid from the 
flask. Store the flask containing the coated glass beads at -20 deg. C 
to -70 deg. C.
    (ii) Test suspension. Aseptically add a coated glass bead to 0.5 
milliliter of medium O and incubate at 37 deg. C for 24 hours. Streak 
the culture onto the solidified agar surface of a plate containing 
medium P and incubate at 37 deg. C for 24 hours. The streak plate may be 
used for 1 week if kept under refrigeration. On the day of test, 
transfer 4 to 10 colonies to a sterile tube containing 0.5 milliliter of 
medium O and incubate at 37 deg. C for 4 to 6 hours. Pipet 0.05 
milliliter of the test suspension into a screw-topped tube containing 25 
milliliters of sterile distilled water and 0.005 milliliter of sterile 
polysorbate 80 and mix well (do not shake). Use this test culture 
suspension as the daily inoculum source.
    (15) Suspension 15. The test organism is Escherichia coli (ATCC 
25922).1 Follow the procedure described for suspension 14 in paragraph 
(b)(14) of this section, except under paragraph (b)(14)(ii) of this 
section use medium Q in place of medium P.\1\
---------------------------------------------------------------------------

    \1\ Available from American Type Culture Collection, 12301 Parklawn 
Drive, Rockville, Md. 20852.
---------------------------------------------------------------------------

    (16) Suspension 16. The test organism is Streptococcus faecalis 
(ATCC 29212)1. Follow the procedure described for suspension 
14 in paragraph (b)(14) of this section.
    (17) Suspension 17. The test organism is Pseudomonas aeruginosa 
(ATCC 27853)1. Follow the procedure described for suspension 
14 in paragraph (b)(14) of this section, except under paragraph 
(b)(14)(ii) of this section use medium Q in place of medium P.
    (18) Suspension 18. The test organism is Staphyloccocus aureas (ATCC 
29247)1. Follow the procedure described for suspension 14 in 
paragraph (b)(14) of this section.
    (19) Suspension 19. The test organism is Enterobacter clocacae (ATCC 
29249)1. Follow the procedure described for suspension 14 in 
paragraph (b)(14) of this section.
    (20) Suspension 20. The test organism is Pseudomonas aeruginosa 
(ATCC 29248)1. Follow the procedure described for suspension 
14 in paragraph (b)(14) of this section, except under paragraph 
(b)(14)(ii) of this section use medium Q in place of medium P.
    (c) Preparation of plates--(1) Baselayer. Depending on the 
particular antibiotic in the discs to be tested, add 42 milliliters of 
the appropriate medium prescribed in paragraph (c)(3) of this section to 
each Petri dish (20 millimeters x 150 millimeters) and allow to harden 
on a flat, level surface and dry slightly by raising the tops on one 
side.
    (2) Seed layer. Add the appropriate amount of inoculum, as 
prescribed by paragraph (c)(3) of this section, to the seed agar which 
has been melted and cooled to 48 deg. C. Swirl the flasks to obtain a 
homogeneous suspension. Add 8 milliliters of the appropriate seed agar, 
as specified in paragraph (c)(3) of this section, to each plate, spread 
evenly over the hardened base layer, and allow to harden and dry on a 
flat level surface. For accurate results, it is necessary to obtain 
uniform distribution of the agar over the surface of the plates.

[[Page 1029]]

    (3) Inoculum and media to be used. Depending on the particular 
antibiotic in the disc to be tested, select from the following table the 
inoculum and media to be used:

                                                                                                                
----------------------------------------------------------------------------------------------------------------
                                                                   Volume of                      Medium        
                                                                  suspension             -----------------------
                                                                   added to                                     
                                                                   each 100   Suspension                        
                           Antibiotic                               ml. of      number                          
                                                                   seed agar              Base layer  Seed layer
                                                                   used for                                     
                                                                     test                                       
----------------------------------------------------------------------------------------------------------------
                                                                         Ml.                                    
Ampicillin......................................................         1.0           3           E           A
Bacitracin......................................................         1.0           3           E           A
Carbenicillin...................................................         3.0          12           F           G
Cefamandole (lithium)...........................................         1.0          10           E           A
Cefoxitin (sodium)..............................................         1.0          10           E           A
Cephaloglycin (dihydrate).......................................         1.0          10           E           A
Cephaloridine...................................................         1.0          10           E           A
Cephalothin.....................................................         1.0          10           E           A
Chloramphenicol.................................................         4.0           4           E           A
Clindamycin.....................................................         2.0           2           A           I
Colistin (sulfate)..............................................         1.0           7           F           G
Erythromycin....................................................         2.0          11           C           C
Gentamicin (sulfate)............................................         0.5           3           C           C
Kanamycin (sulfate).............................................         1.0           9           E           A
Methicillin.....................................................         1.0           3           E           A
Neomycin (sulfate)..............................................         2.5           6           C           C
Novobiocin (sodium).............................................         4.0           5           E           A
Oleandomycin (phosphate)........................................         2.0           3           E           A
Penicillin G....................................................         1.0           3           E           A
Polymyxin B (sulfate)...........................................         1.0           7           F           G
Rifampin........................................................         1.0           5           E           A
Rifampin discs for use in culture media.........................         0.5          13           A           L
Streptomycin (sulfate)..........................................         3.0           1           C           C
Tetracycline (hydrochloride)....................................         1.5           1           E           A
Tobramycin......................................................         0.5           3           C           C
Vancomycin......................................................         1.0           6           C           C
----------------------------------------------------------------------------------------------------------------

    (d) Preparation of control discs. Use round, blank discs having a 
diameter of \1/4\-inch made of clear-white paper weighing 30 milligrams 
plus-minus4 milligrams per square centimeter, and which will 
absorb 2.5 to 3.0 times its own weight of distilled water. The paper 
shall contain no material that either enhances or inhibits the activity 
of any antibacterial agent impregnated thereon. In addition, the paper 
shall contain no materials which will affect the pH of any solvent 
placed on it or buffer any solution placed on it. The following methods 
shall be used to determine the suitability in this regard of any paper 
proposed for this use: Weigh 2 grams of paper or paper discs into a 
clean, glass-stoppered, 250-milliliter flask. Add 30 milliliters of 
freshly boiled and cooled distilled water (the pH of which has been 
determined). Stopper and shake vigorously for 1 hour on a shaking 
machine. Filter through a medium-porosity sintered glass filter. 
Determine the pH of the filtrate. Take the two 10-milliliter aliquots. 
To one add 0.05 milliliter of 0.01N HCl. To the second aliquot add 0.05 
milliliter of 0.01 N NaOH. Determine the pH of each solution. The paper 
shall be satisfactory for use, if (1) the pH of the paper filtrate was 
not more than plus-minus0.3 pH units different from the pH of 
the distilled water used; (2) the pH of the acidified aliquot was 
lowered by at least 1.0 pH units; (3) the pH of the alkalinized aliquot 
was raised by at least 1.5 pH units. Place blank discs on aluminum or 
stainless steel wire mesh which is supported in a manner to allow 
circulation of air above and below the discs. Prepare the desired number 
of discs for each point on the standard curve by accurately adding 0.02-
milliliter-increments of the appropriate standard stock solution to each 
disc, using a suitable pipette. Dry discs in circulating air or under 
vacuum. Discs may be stored for 2 weeks in a

[[Page 1030]]

desiccator under refrigeration. Depending on the antibiotic contained in 
the sample to be tested, prepare the stock solutions for the standard 
discs by dissolving an accurately weighed quantity of the working 
standard in the solvent indicated to obtain stock solutions that will 
contain the following concentrations required for the standard discs:

                                                                        
------------------------------------------------------------------------
                                                       Standard curve   
                                                        (antibiotic     
          Antibiotic                 Solvent         concentration per  
                                                           disc)        
------------------------------------------------------------------------
Ampicillin....................  Water............  1.3.2.4, 4.4, 8.1,   
                                                    15.0g.     
Bacitracin....................  ......do.........  1.3, 2.4, 4.4, 8.1,  
                                                    15.0 units.         
Carbenicillin.................  Methyl alcohol...  25.0, 35.5, 50.0,    
                                                    70.7, 100g.
Cefamandole (lithium).........  50 percent methyl  5, 30, and 60 gg.               
Cefoxitin (sodium)............  50 percent methyl  5, 30, 60 g.
                                 alcohol.                               
Cephalothin...................  50 percent methyl  15.0, 21.2, 30.3,    
                                 alcohol.           42.4, 60.0g.                
Chloramphenicol...............  ......do.........  3.3, 6.3, 12.2, 23.4,
                                                    45.0g.     
Clindamycin...................  ......do.........  1.0, 1.41, 2.0, 2.82,
                                                    4.0g.      
Colistin (sulfate)............  Water............  1.3, 2.4, 4.4, 8.1,  
                                                    15.0g.     
Erythromycin..................  Methyl alcohol...  1.3, 2.7, 5.4, 11.0, 
                                                    2250g.     
Gentamicin (sulfate)..........  Water............  5, 7.1, 10, 14.1,    
                                                    20g.       
Kanamycin (sulfate)...........  ......do.........  3.3, 6.3, 12.2, 23.4,
                                                    45g.       
Methicillin...................  ......do.........  1.3, 2.4, 4.4, 8.1,  
                                                    15.0g.     
Neomycin (sulfate)............  ......do.........  3.3, 6.3, 12.2, 23.4,
                                                    45.0g.     
Novobiocin (sodium)...........  ......do.........  3.3, 6.3, 12.2, 23.4,
                                                    45.0g.     
Oleandomycin (phosphate)......  ......do.........  1.3, 2.7, 5.4, 11.0, 
                                                    22.5g.     
Penicillin G..................  ......do.........  1.3, 2.4, 4.4, 8.1,  
                                                    15.0 units.         
Polymyxin B (sulfate).........  ......do.........  33, 63, 122, 234, 450
                                                    units.              
Rifampin......................  Methyl alcohol...  3.0, 6.0, 12.0, 24.0,
                                                    48.0g.     
Rifampin discs for use in       ......do.........  12.5, 25, 50g.                
Streptomycin (sulfate)........  Water............  1.3, 2.4, 4.4, 8.1,  
                                                    15.0g.     
Tetracycline (hydrochloride)..  Methyl alcohol...  3.3, 6.3, 12.2, 23.4,
                                                    45.0g.     
Tobramycin....................  Water............  5, 10, and 20g.                
Vancomycin (hydrochloride)....  ......do.........  3.3, 6.3, 12.2, 23.4,
                                                    45.0g.     
------------------------------------------------------------------------

    (e) Assay--(1) Individual discs one-fourth inch in diameters--(i) 
Standard curves with five antibiotic concentrations. On each of five 
plates prepared as directed in paragraph (c) of this section, place the 
five control discs for the standard curve and two discs from each batch 
to be tested. The control discs for the standard curve and the sample 
discs are placed on the plates in a random arrangement, with no discs 
being closer than 24 millimeters (on centers) to another disc. Discs are 
placed on the plates with the aid of forceps within as short a period of 
time as possible (not to exceed 3 minutes per plate) and tapped gently 
to ensure an even seal. Incubate the plates overnight at 32 deg. C to 
35 deg. C, except if it is cephalothin, colistin, novobiocin, polymyxin, 
or viomycin, the incubation temperature is 37 deg. C. After incubation, 
measure the diameter of each circle of inhibition, using calipers or a 
measuring device of comparable accuracy. Average the three zone sizes 
for each of the five standard-curve concentrations and plot the mean 
sizes on the arithmetic scale of semilogarithmic graph paper with the 
antibiotic concentrations on the logarithmic scale. Use the following 
equation to calculate the best straight line:

                           L=(3a+2b+c-e)/(5),

                           H=(3e+2d+c-a)/(5),

where:
L=the calculated zone size of the low concentration;
H=the calculated zone size of the high concentration;
a, b, c, d, e=the observed average zone sizes for each respective 
concentration, a being that for the lowest concentration.


Plot the values obtained for L and H and connect these two points with a 
straight line. Average the six sample zone sizes and read the 
corresponding antibiotic concentration of this mean from the standard 
curve. This is the potency obtained for a single assay. Perform two or 
more replicate assays on each of 2 days. The average of all assays is 
the potency of the sample disc.
    (ii) Standard curves with three antibiotic concentrations. On each 
of three plates prepared as directed in paragraph (c) of this section, 
place the three control discs for the standard

[[Page 1031]]

curve and two discs from each batch to be tested. The control discs for 
the standard curve and the sample discs are placed on the plates in a 
random arrangement, with no discs being closer than 24 millimeters (on 
centers) to any other discs. Discs are placed on the plates with the aid 
of forceps within as short a period of time as possible (not to exceed 3 
minutes per plate) and tapped gently to ensure an even seal. Incubate 
the plates overnight at 32 deg. C to 35 deg. C, except if it is rifampin 
discs for use in culture media, the incubation temperature is 37 deg. C. 
After incubation, measure the diameter of each circle of inhibition, 
using calipers or a measuring device of comparable accuracy. Average the 
three zone sizes for each of the three standard curve concentrations and 
plot the mean sizes on the arithmetic scale of semilogarithmic graph 
paper with the antibiotic concentrations on the logarithmic scale. Using 
the following equation to calculate the best straight line:

                            L=(5a+2b-c)/(6),

                            H=(5c+2b-a)/(6),

where:
L=calculated zone diameter of the lowest concentration of the standard 
curve;
H=calculated zone diameter of the highest concentration of the standard 
curve;
a, b, c=observed average zone sizes for each respective concentration, a 
being that for the lowest concentration.


Plot the values obtained for L and H and connect these two points with a 
straight line. Average the six sample zone sizes and read the 
corresponding antibiotic concentration of this mean from the standard 
curve. This is the potency obtained for a single assay. Perform two or 
more replicate assays on each of 2 days. The average of all assays is 
the potency of the sample disc.
    (2) Discs one-fourth inch in diameter attached to rings, spokes, or 
other devices. Remove or cut the disc from the device, including a small 
portion of the device to which it is attached, before testing, and 
proceed as directed in paragraph (e)(1) of this section.
    (3) Individual discs with diameters larger than one-fourth inch but 
no larger than three-eighths inch for use in impregnating culture media. 
Proceed as directed in paragraph (e)(1) of this section, except instead 
of measuring the diameters of the zones of inhibition, measure the 
widths of the zones from any edge of the sample discs and the standard 
discs. The results obtained are multiplied by the factor 2 for 
determining whether the discs meet the requirements for uniformity 
prescribed by paragraph (f) of this section.
    (f) The potency is satisfactory if the result obtained is not less 
than 67 percent and not more than 150 percent of that represented. The 
batch has a uniform potency if on the first or second test of six discs 
each, the diameter of the largest zone of inhibition is not more than 
2.5 millimeters larger than the smallest zone, or if the number of zones 
that fall outside this range in three or more consecutive tests is not 
more than 10 percent of the total number of discs tested.
[39 FR 19181, May 30, 1974, as amended at 41 FR 7094, Feb. 17, 1976; 41 
FR 53476, Dec. 7, 1976; 43 FR 9792, Mar. 10, 1978; 43 FR 12858, Mar. 28, 
1978; 44 FR 10376, Feb. 20, 1979; 44 FR 20667, Apr. 6, 1979]



Sec. 460.11  Certification procedures for antibiotic elution susceptibility discs.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Antibiotic elution susceptibility discs 
are round flat discs that have a diameter of 6.35 millimeters (\1/4\ 
inch) and are made of absorbent paper containing antibiotic compounds. 
The identity of each disc is signified by means of an identifying sign. 
The absorbent paper and dye or ink used must not affect either bacterial 
growth or the antibiotic. Each disc shall have a potency that is 
equivalent to that contained in a standard disc prepared with the 
following quantities of antibiotic drugs:

    Ampicillin: 0.22 mcg.
    Ampicillin: 4.5 mcg.
    Bacitracin: 18.0 units.
    Carbenicillin: 120.0 mcg.
    Cephalothin: 15.0 mcg.
    Chloramphenicol: 4.0 mcg.
    Clindamycin: 2.0 mcg.
    Colistin: 13.0 mcg.
    Doxycycline: 0.5 mcg.
    Doxycycline: 1.6 mcg.
    Erythromycin: 2.5 mcg.
    Gentamicin: 9.0 mcg.

[[Page 1032]]

    Kanamycin: 22.0 mcg.
    Methicillin: 5.0 mcg.
    Neomycin: 24.0 mcg.
    Novobiocin: 2.5 mcg.
    Oleandomycin: 6.0 mcg.
    Penicillin: 0.2 unit.
    Streptomycin: 20.0 mcg.
    Tetracycline: 0.5 mcg.
    Tetracycline: 1.2 mcg.
    Tobramycin: 10.0 mcg.
    Vancomycin: 10.0 mcg.

    The standard discs used to determine the potency shall be made of 
paper as described in Sec. 460.6(d). Each antibiotic compound used to 
impregnate such standard discs shall be equilibrated in terms of the 
working standard designated by the Commissioner for use in determining 
the potency or purity of such antibiotic.
    (2) Packaging. The immediate container shall be a tight container as 
defined by the U.S.P. and shall be of such composition as will not cause 
any change in the strength, quality, or purity of the contents beyond 
any limit therefor in applicable standards, except that minor changes so 
caused that are normal and unavoidable in good packaging, storage, and 
distribution practice shall be disregarded. Each immediate container may 
contain a desiccant, and each may be packaged in combination with 
containers of suitable discs of drugs other than those described in 
paragraph (a)(1) of this section. Such other discs shall be suitable 
only if the manufacturer and packer have submitted to the Commissioner 
information of the kind described in Sec. 431.17 of this chapter, and 
such information has been accepted by the Commissioner.
    (3) Labeling. Each package of discs shall bear on its label or 
labeling, as hereinafter indicated, the following:
    (i) On the outside wrapper or container and the immediate container:
    (a) The batch mark.
    (b) The name and potency of each disc in the batch according to the 
following:

 Name of disc and content of antibiotic in micrograms or units per disc

    Ampicillin elution disc, 0.22 mcg.
    Ampicillin elution disc, 4.5 mcg.
    Bacitracin elution disc, 18.0 units.
    Carbenicillin elution disc, 120.0 mcg.
    Cephalothin elution disc, 15.0 mcg.
    Chloramphenicol elution disc, 4.0 mcg.
    Clindamycin elution disc, 2.0 mcg.
    Colistin elution disc, 13.0 mcg.
    Doxycycline elution disc, 0.5 mcg.
    Doxycycline elution disc, 1.6 mcg.
    Erythromycin elution disc, 2.5 mcg.
    Gentamicin elution disc, 9.0 mcg.
    Kanamycin elution disc, 22.0 mcg.
    Neomycin elution disc, 24.0 mcg.
    Novobiocin elution disc, 2.5 mcg.
    Oleandomycin elution disc, 6.0 mcg.
    Penicillin elution disc, 0.2 unit.
    Methicillin elution disc, 5.0 mcg.
    Streptomycin elution disc, 20.0 mcg.
    Tetracycline elution disc, 0.5 mcg.
    Tetracycline elution disc, 1.2 mcg.
    Tobramycin elution disc, 10.0 mcg.
    Vancomycin elution disc, 10.0 mcg.

    (c) The statement ``Expiration date ----------'', the blank being 
filled in with the date that is 6 months after the month during which 
the batch was certified, except that the blank may be filled in with a 
date that is 12, 18, 24, 30, 36, 42, 48, 54, or 60 months after the 
month during which the batch was certified if the person who requests 
certification has submitted to the Commissioner results of tests and 
assays showing that such drugs as prepared by that person are stable for 
such longer period of time. If it is a packaged combination of discs of 
two or more drugs, its outside wrapper shall bear only one expiration 
date, and that date shall be the date that is required for the shortest 
dated discs contained in the package.
    (d) The statement ``FOR IN VITRO DIAGNOSTIC USE''.
    (ii) On the circular or other labeling within or attached to the 
package, adequate directions for the use of such discs.
    (4) Request for certification; samples. (i) In addition to complying 
with the requirements of Sec. 431.1 of this chapter, a person who 
requests certification of a batch of antibiotic elution susceptibility 
discs shall submit with the request a statement showing the batch mark, 
the number of packages of each size in such batch, and the date on which 
the latest assay of the antibiotic used in making such batch was 
completed, the potency of each disc batch, the quantity of each 
ingredient used in making the batch, the date on which the latest assay 
of the drug constituting such batch was completed, and a statement that 
each ingredient used in

[[Page 1033]]

making the batch conforms to the requirements prescribed therefor by 
this section.
    (ii) In connection with the request, such person shall submit 
results of the tests and assays made by him or her on an accurately 
representative sample of the batch for potency.
    (iii) In connection with the request, such person shall submit an 
accurately representative sample of the batch, consisting of one disc 
for each 5,000 discs in the batch, but in no case collecting less than 
100 discs. Single discs will be taken at regular intervals throughout 
the entire time of packaging the batch.
    (b) Tests and methods of assay for potency of antibiotic elution 
susceptibility discs--(1) Preparation for assay. Use culture media as 
directed in Sec. 460.6(a).
    (2) Test organisms--(i) Culture of test organism suspensions. For 
each test organism listed in the following table, select the appropriate 
medium (as listed in Sec. 460.6(a)), incubation period of the Roux 
bottle, and suggested storage period under refrigeration for the 
particular test organism.

                                                                                                                
----------------------------------------------------------------------------------------------------------------
                                                                            Medium used for                     
                                                     -----------------------------------------------------------
                    Test organism                       Method                 Roux                             
                                                         used      Slants    bottles          Storage \2\       
----------------------------------------------------------------------------------------------------------------
Suspension 1--Staphylococcus aureus (ATCC 29737) \1\          1          A          A  2 weeks.                 
Suspension 2--Staphylococcus aureus (ATCC 13150) \1\          1          A          A  1 week.                  
Suspension 3--Pseudomonas aeruginosa (ATCC 25619)             6          J          J  1 week.                  
 \1\.                                                                                                           
Suspension 4--Klebsiella pneumoniae (ATCC 10031) \1\          1          A          A  2 weeks.                 
Suspension 5--Micrococcus luteus (ATCC 9341) \1\....          2          A          A  2 weeks.                 
Suspension 6--Bordetella bronchiseptica (ATCC 4617)           1          A          A  2 weeks.                 
 \1\.                                                                                                           
Suspension 7--Streptococcus faecalis (ATCC 14506)             3          E         --  3 days.                  
 \1\.                                                                                                           
Suspension 8--Staphylococcus epidermidis (ATCC                4          A         --  3 days.                  
 12228) \1\.                                                                                                    
Suspension 9--Bacillus subtilis (ATCC 6633) \1\.....          5          A          B  1 year.                  
----------------------------------------------------------------------------------------------------------------
\1\ Available from: American Type Culture Collection, 12301 Parklawn Dr., Rockville, MD 20852.                  
\2\ Storage period under refrigeration.                                                                         

    (ii) Methods of preparation of test organism suspensions--(a) Method 
1. Maintain organisms on agar slants containing 10 milliliters of the 
appropriate medium. Transfer organisms to fresh slants using an 
inoculating loop. Streak the fresh slants thoroughly. Incubate the 
slants for 24 hours at 37 deg. C. Remove resulting growth from the agar 
slant with sterile glass beads. Transfer the cells onto a large agar 
surface, such as a Roux bottle, containing 250 milliliters of the 
appropriate medium. Spread the cells over the entire surface of the Roux 
bottle. Incubate the Roux bottle for 24 hours at 37 deg. C. Wash the 
resulting growth from the agar surface with 50 milliliters of sterile 
U.S.P. saline test solution.
    (b) Method 2. Proceed as directed in paragraph (b)(2)(ii)(a) of this 
section, except wash the growth from the surface of the Roux bottle with 
20 milliliters sterile U.S.P. saline test solution.
    (c) Method 3. Using an inoculation loop, transfer a portion of the 
growth on the slant to a culture tube containing 10 milliliters of 
sterile medium of the following composition:

    Calf brains, infusion from, 200.0 gm.
    Beef heart, infusion from, 250.0 gm.
    Proteose peptone, 10.0 gm.
    Dextrose, 2.0 gm.
    Sodium cloride, 5.0 gm.
    Sodium phosphate dibasic, 2.5 gm.
    Distilled water, q.s. pH 7.4 after sterilization, 1,000.0 ml.
    Incubate for 24 hours at 37 deg. C.

    (d) Method 4. Proceed as directed in paragraph (b)(2)(ii)(c) of this 
section, except transfer growth from the slant to a culture tube 
containing 50 milliliters of Medium D.
    (e) Method 5. Proceed as directed in paragraph (b)(2)(ii)(a) of this 
section. Incubate the Roux bottle for 7 days at 37 deg. C. Centrifuge 
the suspension at 3,500 RPM for 30 minutes. Decant the supernatant 
liquid. Resuspend the sediment in 50 milliliters sterile U.S.P. saline 
test solution. Heat-shock the suspension by placing in a 70 deg. C. 
water bath for 30 minutes.
    (f) Method 6. Proceed as directed in paragraph (b)(2)(ii)(b) of this 
section, except use 20 milliliters of Medium K.

[[Page 1034]]

    (3)(i) Preparation of plates. Use volumes of appropriate media and 
plates as directed in Sec. 460.6(c)(1) and (2).
    (ii) Inoculum and media to be used. Depending on the particular 
antibiotic in the disc to be tested, select from the following table the 
inoculum and media to be used:

                                                                                                                
----------------------------------------------------------------------------------------------------------------
                                                                Medium                                          
                                                        ---------------------- Suspension                Incub. 
                       Antibiotic                           Base       Seed        No.     Volume \3\    temp.  
                                                           layer      layer                                     
----------------------------------------------------------------------------------------------------------------
                                                                                                   ml     deg. C
Ampicillin (.22 mcg)...................................          E          A           1         1.5      32-35
Ampicillin (4.5 mcg)...................................          A          L           2     \1\ 1.0      32-35
B acitracin............................................          A          L           2     \1\ 1.0      32-35
Carbenicillin..........................................          A          L           3         5.0         37
Cephalothin............................................          A          L           4         1.0         37
Chloramphenicol........................................          A          L           5         1.0      32-35
Clindamycin............................................          C          L           1         2.0      32-35
Colistin...............................................          G          L           6         0.3         37
Doxycycline............................................          E          A           5         0.1      32-35
Erythromycin...........................................          C          L           7         0.2         37
Gentamicin.............................................          C          L           2         0.1      32-35
Kanamycin..............................................          A          L           1     \1\ 1.0      32-35
Methicillin............................................          A          L           2         0.1      32-35
Neomycin...............................................          C          L           8         6.0      32-35
Novobiocin.............................................          C          L           9     \2\ 0.2         37
Oleandomycin...........................................          A          L           2     \2\ 1.0      32-35
Penicillin.............................................          E          A           1         1.5      32-35
Streptomycin...........................................          C          L           1         0.1      32-35
Tetracycline...........................................          E          A           5         0.1      32-35
Tobramycin.............................................          C          C           2         0.1      32-35
Vancomycin.............................................          C          L           3         2.0      32-35
----------------------------------------------------------------------------------------------------------------
\1\ Prepare a 1:100 dilution of the bulk suspension in sterile U.S.P. saline test solution. Use the indicated   
  quantity of the diluted suspension to inoculate the seed medium.                                              
\2\ Prepare a 1:10 dilution of the bulk suspension in sterile U.S.P. saline test solution. Use the indicated    
  quantity of the diluted suspension to inoculate the seed medium.                                              
\3\ Suggested volume of suspension to be added to each 100 ml of seed agar.                                     

    (4) Preparation of standard discs. Depending on the concentration of 
antibiotic contained in the disc to be tested, prepare a stock solution 
for the standard disc by dissolving an accurately weighed quantity of 
the working standard in the solvent indicated to obtain an appropriate 
stock solution. Make further dilutions as required in the solvent 
indicated to obtain the following concentrations required on the 
standard discs:

                                                                        
------------------------------------------------------------------------
                                                         Standard disc  
           Antibiotic                   Solvent         concentrations  
------------------------------------------------------------------------
Ampicillin (0.22 mcg.)..........  Water.............  0.1, 0.2, 0.4 mcg.
Ampicillin (4.5 mcg.)...........  ......do..........  1, 5, 15 mcg.     
Bacitracin......................  ......do..........  5, 15, 30 units.  
Carbenicillin...................  Methanol..........  24, 80, 240 mcg.  
Cephalothin.....................  50-percent          7.5, 20, 60 mcg.  
                                   methanol.                            
Chloramphenicol.................  ......do..........  2, 4, 8 mcg.      
Clindamycin.....................  ......do..........  1, 2, 4 mcg.      
Colistin........................  Water.............  5, 12.5, 25 mcg.  
Doxycycline.....................  Methanol..........  0.25, 1, 3 mcg.   
Erythromycin....................  ......do..........  1.25, 5, 22.5 mcg.
Gentamicin......................  Water.............  5, 10, 20 mcg.    
Kanamycin.......................  ......do..........  3, 15, 45 mcg.    
Methicillin.....................  ......do..........  2.5, 5, 10 mcg.   
Neomycin........................  ......do..........  3, 15, 45 mcg.    
Novobiocin......................  Methanol..........  1.25, 2.5, 5 mcg. 
Oleandomycin....................  Water \1\.........  1.25, 5, 22.5 mcg.
Penicillin......................  ......do..........  0.1, 0.2, 0.4     
                                                       unit.            
Streptomycin....................  ......do..........  6.25, 25, 100 mcg.
Tetracycline....................  Methanol..........  0.25, 1, 3 mcg.   
Tobramycin......................  Water.............  5, 10, 20 mcg.    
Vancomycin......................  ......do..........  3, 15, 45 mcg.    
------------------------------------------------------------------------
\1\ If the chloroform adduct of oleandomycin is used as the standard,   
  dissolve the weighing in absolute ethanol to a stock concentration of 
  10,000 micrograms per milliliter. Dilute this solution in water to    
  achieve the working concentrations.                                   

    Use round, blank discs that conform to Sec. 460.6(d). Place blank 
discs on aluminum or stanless steel wire mesh that is supported to allow 
circulation of air above and below the discs. Prepare the desired number 
of discs for each standard disc concentration by accurately adding 0.02-
milliliter aliquots of the appropriate concentration of standard 
solution to each disc. Dry the discs in circulating air. Store standard 
discs under refrigeration in the presence of desiccant for a period not 
to exceed 2 weeks. Determine the stability of stored standard discs by 
assaying them

[[Page 1035]]

at daily and weekly interveral using freshly prepared standard disc for 
comparison.
    (5) Assay--(i) Individual discs. On each of three plates prepared as 
directed in paragraph (b)(3) of this section, place standard disc and 
two or more discs from each batch to be tested. The standard disc and 
the sample discs are placed on the plates in a circular pattern with 
random arrangement, with no disc being closer than 24 millimeters (on 
centers) to any other disc. Discs are placed on the plates within as 
short a period of time as possible (not to exceed 3 minutes per plate) 
and tapped gently to ensure an even seal. After incubation as directed 
in paragraph (b)(3) of this section, measure the diameter of each circle 
of inhibition as accurately as possible. (In most cases, it is possible 
to estimate diameters to the nearest 0.1 millimeter).
    (ii) Estimation of potency. Determine the logarithm of each dose of 
standard (x values) and the mean zone diameter for each dose of standard 
(y values). Using the three values of x and the three corresponding 
values of y, calculate x, x2, (x)2, 
y, and xy. Calculate the regression coefficient 
(slope, b) and the Y-intercept (a) of the standard response line by 
using the following equations:
[GRAPHIC] [TIFF OMITTED] TR01JA93.348

[GRAPHIC] [TIFF OMITTED] TR01JA93.349


where n = the number of standard doses.
    Determine the zone diameter (Y) for each sample disc being tested. 
Using the regression equation
[GRAPHIC] [TIFF OMITTED] TR01JA93.350


calculate the concentration (X) for the mean response (Y) of the sample 
discs.
    (6) Potency. The potency of the batch is satisfactory if the mean 
result obtained for the batch is not less than 85 percent and not more 
than 150 percent of that represented.
[45 FR 20668, Apr. 6, 1979]



Sec. 460.15  Streptomycin sulfate discs for use in culture media.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Streptomycin sulfate discs for use in 
culture media are paper discs intended for impregnation of culture media 
in the sensitivity testing of mycobacteria. They conform to all 
requirements and to all procedures prescribed by Sec. 460.1(a) for 
antibiotic sensitivity discs, except that each disc shall contain 
streptomycin sulfate equivalent to 10, 25, 50, or 500 micrograms of 
streptomycin.
    (2) Packaging. It shall be packaged in accordance with the 
requirements of Sec. 460.1(b).
    (3) Labeling. In addition to complying with the requirements of 
Sec. 460.1(c) of this chapter, the labeling shall also bear information 
indicating that the discs are for use in culture media for the 
sensitivity testing of mycobacteria and not for use in ordinary 
sensitivity disc plate tests.
    (4) Requests for certification; samples. Requests for certification 
shall comply with Sec. 460.1(d).
    (b) Tests and methods of assay; potency. Proceed as directed in 
Sec. 460.6 for the assay of streptomycin sulfate discs, except that:
    (1) In the assay of streptomycin sulfate discs labeled to contain 
the equivalent of 10, 25, or 50 micrograms of streptomycin, the control 
discs shall be made to contain the equivalent of 6.25, 12.5, 25, 50, and 
100 micrograms of streptomycin per disc.
    (2) In the assay of streptomycin sulfate discs labeled to contain 
the equivalent of 500 micrograms of streptomycin:
    (i) To each 100 milliliters of seed agar used for the test add 2.0 
milliliters of suspension number 11.
    (ii) The control discs shall be made to contain the equivalent of 
50, 100, 200, 400, and 800 micrograms of streptomycin per disc.



Sec. 460.16  Rifampin discs for use in culture media.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Rifampin discs for use in culture media 
are paper discs intended for impregnation of culture media in

[[Page 1036]]

the susceptibility testing of mycobacteria. They conform to all 
requirements and to all procedures prescribed by Sec. 460.1(a) for 
antibiotic susceptibility discs, except that each disc shall contain 25 
micrograms of rifampin activity.
    (2) Packaging. It shall be packaged in accordance with the 
requirements of Sec. 460.1(b).
    (3) Labeling. In addition to complying with the requirements of 
Sec. 460.1(c), the labeling shall also bear information indicating that 
the discs are for use in culture media for the susceptibility testing of 
mycobacteria and not for use in susceptibility tests of other 
microorganisms as described in Sec. 460.1(c)(2).
    (4) Requests for certification; samples. Requests for certification 
shall comply with Sec. 460.1(d), except an accurately representative 
sample of the batch shall consist of one disc for each 5,000 in the 
batch, but in no case less than 100 discs collected by taking single 
discs at such intervals throughout the entire time of manufacturing the 
batch that the quantities manufactured during the intervals are 
approximately equal.
    (b) Tests and methods of assay; potency. Proceed as directed in 
Sec. 460.6.
[41 FR 53476, Dec. 7, 1976]



                    Subpart B--Susceptibility Powders



Sec. 460.25  Bacitracin diagnostic sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Bacitracin diagnostic sensitivity powder 
is bacitracin, with or without one or more suitable buffers and 
diluents, packaged in vials and intended for use in clinical 
laboratories for determining in vitro the sensitivity of microorganisms 
to bacitracin. Each vial contains 2,000 units of bacitracin. The potency 
of each immediate container is satisfactory if it contains not less than 
90 percent and not more than 115 percent of its labeled content. It is 
sterile. Its loss on drying is not more than 5.0 percent. When 
reconstituted as directed in the labeling, its pH is not less than 5.5 
and not more than 7.5. The bacitracin used conforms to the standards 
prescribed by Sec. 448.10a(a)(1) (i), (v), and (vi) of this chapter. 
Each other substance used, if its name is recognized in the U.S.P. or 
N.F., conforms to the standards prescribed therefor by such official 
compendium.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile diluent when preparing a stock 
solution for use in making further dilutions for microbial 
susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement ``For laboratory diagnostic use only.''
    (b) The statement ``Sterile.''
    (c) The batch mark.
    (d) The number of units of bacitracin in each immediate container.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The bacitracin used in making the batch for potency, moisture, 
and pH.
    (b) The batch for potency sterility, loss on drying, and pH.
    (ii) Samples required:
    (a) The bacitracin used in making the batch: 6 packages, each 
containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an aliquot with 1.0 
percent potassium phosphate buffer, pH 6.0 (solution 1), to the 
prescribed reference concentration.

[[Page 1037]]

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.



Sec. 460.28  Disodium carbenicillin diagnostic sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Disodium carbenicillin diagnostic 
sensitivity powder is disodium carbenicillin packaged in vials and 
intended for use in clinical laboratories for determining in vitro the 
sensitivity of microorganisms to carbenicillin. Each vial contains 
disodium carbenicillin equivalent to not more than 1.0 gram of 
carbenicillin. The potency of each immediate container is satisfactory 
if it contains not less than 90 percent and not more than 120 percent of 
its labeled content. It is sterile. Its moisture content is not more 
than 6 percent. When reconstituted as directed in the labeling, its pH 
is not less than 6.0 and not more than 8.0. The disodium carbenicillin 
used conforms to the standards prescribed by Sec. 440.13a(a)(1) (i), 
(v), (vi), and (vii) of this chapter.
    (2) Packaging. It shall conform to the packaging requirements of 
Sec. 432.1 of this chapter.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement ``For laboratory diagnostic use only.''
    (b) The statement ``Sterile.''
    (c) The batch mark.
    (d) The number of milligrams of carbenicillin in each immediate 
container.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The disodium carbenicillin used in making the batch for potency, 
moisture, pH, and identity.
    (b) The batch for potency, sterility, moisture, and pH.
    (ii) Samples required:
    (a) The disodium carbenicillin used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an aliquot with 1 
percent potassium phosphate buffer, pH 6.0 (solution 1), to the 
reference concentration of 20 micrograms of carbenicillin per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.



Sec. 460.33  Clindamycin hydrochloride hydrate sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clindamycin hydrochloride hydrate 
diagnostic sensitivity powder is clindamycin hydrochloride hydrate 
packaged in vials and intended for use in clinical laboratories for 
determining in vitro the sensitivity of microorganisms to clindamycin. 
Each vial contains clindamycin hydrochloride hydrate equivalent to 20 
milligrams of clindamycin. Its potency is satisfactory if it is not less 
than 90 percent and not more than 120 percent of the amount of 
clindamycin it is represented to contain. It is sterile. Its moisture 
content is not more than 6.0 percent. Its pH in a solution containing

[[Page 1038]]

20 milligrams of clindamycin per milliliter is not less than 3.0 and not 
more than 6.5. The clindamycin hydrochloride hydrate used conforms to 
the standards prescribed by Sec. 453.20(a)(1) (i), (ii), (iv), (v), 
(vi), and (vii) of this chapter.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile diluent when preparing a stock 
solution for use in making further dilutions for microbial 
susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement ``For laboratory diagnosis only.''
    (b) The statement ``Sterile''.
    (c) The batch mark.
    (d) The number of milligrams of clindamycin in each immediate 
container.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Request for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The clindamycin hydrochloride hydrate used in making the batch 
for clindamycin content, microbiological activity, moisture, pH, 
crystallinity, and identity.
    (b) The batch for potency, sterility, moisture, and pH.
    (ii) Samples required:
    (a) The clindamycin hydrochloride hydrate used in making the batch: 
10 packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an aliquot of this 
solution with 0.1M potassium phosphate buffer, pH 8.0 (solution 3), to 
the reference concentration of 1.0 microgram of clindamycin per 
milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution obtained when the vial is reconstituted with 1.0 milliliter 
of distilled water.



Sec. 460.38  Sodium colistimethate diagnostic sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sodium colistimethate diagnostic 
sensitivity powder is sodium colistimethate packaged in vials and 
intended for use in clinical laboratories for determining in vitro the 
sensitivity of microorganisms to sodium colistimethate. Each vial 
contains sodium colistimethate equivalent to 100 milligrams of colistin 
base activity. It is sterile. Its moisture content is not more than 9.0 
percent. When reconstituted as directed in the labeling, its pH is not 
less than 6.5 and not more than 9.0. It gives a positive identity test 
for sodium colistimethate. The sodium colistimethate used conforms to 
the standards prescribed by Sec. 448.20a(a)(1) (i), (vi), (vii), and 
(viii) of this chapter. Each other substance used, if its name is 
recognized in the U.S.P. or N.F., conforms to the standards prescribed 
therefor by such official compendium.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile diluent when preparing a stock 
solution for use in making serial dilutions for microbial susceptibility 
testing.

[[Page 1039]]

    (3) Labeling. Each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container.
    (a) The statements ``Not for therapeutic use'' and ``For laboratory 
diagnosis only''.
    (b) The statement ``Sterile''.
    (c) The batch mark.
    (d) The number of milligrams of colistin base activity in each 
immediate container.
    (e) The statements ``Stock solutions are stable for 14 days when 
refrigerated. For periods of storage up to 6 months, they should be 
frozen''.
    (f) Its expiration date which is 12 months, except that the date may 
be used that is 18, 24, 30, 36, 42, 48, 54, or 60 months after the month 
during which the batch was certified if the person who requests 
certification has submitted to the Commissioner results of tests and 
assays showing that such drug as prepared by him is stable for such 
period of time. If the manufacturer or repacker of the drug has been 
exempted from the certification requirements, such date shall be the 
number of months after the month during which the batch was last assayed 
and released by the manufacturer or repacker.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The sodium colistimethate used in making the batch for potency, 
moisture, pH, and identity.
    (b) The batch for potency, sterility, moisture, pH, and identity.
    (ii) Samples required:
    (a) The sodium colistimethate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 30 immediate 
containers.
    (2) For sterility testing: 20 immediate containers collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 448.20a(b)(1) of this chapter, except prepare the sample for assay 
as follows: Reconstitute as directed in the labeling and further dilute 
with 10 percent potassium phosphate buffer, pH 6.0, to the proper 
prescribed reference concentration. Its potency is satisfactory if it 
contains not less than 90 percent and not more than 115 percent of the 
number of milligrams of colistin base activity that it is represented to 
contain.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Moisture. Proceed as directed in Sec. 440.80a(b)(5)(i) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 440.80a(b)(5)(ii) of this 
chapter, using the drug reconstituted as directed in the labeling.
    (5) Identity. Proceed as directed in Sec. 448.20a(b)(7) of this 
chapter.



Sec. 460.42  Dihydrostreptomycin sulfate diagnostic sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Dihydrostreptomycin sulfate sensitivity 
powder is crystalline dihydrostreptomycin sulfate, with or without one 
or more suitable buffers and diluents, packaged in vials and intended 
for use in clinical laboratories for determining in vitro the 
sensitivity of microorganisms to dihydrostreptomycin. Each vial contains 
dihydrostreptomycin sulfate equivalent to 20 milligrams of 
dihydrostreptomycin. The potency of each immediate container is 
satisfactory if it contains not less than 90 percent and not more than 
115 percent of its labeled content. It is sterile. Its loss on drying is 
not more than 5.0 percent. When reconstituted as directed in the 
labeling, its pH is not less than 4.5 and not more than 7.0. The 
dihydrostreptomycin sulfate used conforms to the standards prescribed by 
Sec. 444.10a(a)(1) of this chapter, except the standards for sterility, 
pyrogens, and depressor substances. Each other substance used, if its 
name is recognized in the U.S.P. or N.F., conforms to the standards 
prescribed therefor by such official compendium.

[[Page 1040]]

    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile diluent when preparing a stock 
solution for use in making further dilutions for microbial 
susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement ``For laboratory diagnostic use only''.
    (b) The statement ``Sterile''.
    (c) The batch mark.
    (d) The number of milligrams of dihydrostreptomycin in each 
immediate container.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The dihydrostreptomycin sulfate used in making the batch for 
potency, moisture, pH, streptomycin content, and crystallinity.
    (b) The batch for potency, sterility, loss on drying, and pH.
    (ii) Samples required:
    (a) The dihydrostreptomycin sulfate used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an aliquot with sterile 
distilled water to the prescribed reference concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.
[39 FR 19181, May 30, 1974, as amended at 46 FR 60569, Dec. 11, 1981; 50 
FR 19921, May 13, 1985]



Sec. 460.47  Doxycycline hyclate diagnostic sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Doxycycline hyclate diagnostic 
sensitivity powder is crystalline doxycycline hyclate, with or without 
one or more suitable buffers and diluents, packaged in vials and 
intended for use in clinical laboratories for determining in vitro the 
sensitivity of microorganisms to doxycycline. Each vial contains 
doxycycline hyclate equivalent to 20 milligrams of doxycycline. The 
potency of each immediate container is satisfactory if it contains not 
less than 90 percent and not more than 115 percent of its labeled 
content. It is sterile. Its moisture content is not more than 4 percent. 
When reconstituted as directed in the labeling, its pH is not less than 
2.0 and not more than 3.5. The doxycycline hyclate used conforms to the 
standards prescribed by Sec. 446.20(a)(1) (i), (iii), (iv), (v), and 
(vi) of this chapter. Each other substance used, if its name is 
recognized in the U.S.P. or N.F., conforms to the standards prescribed 
therefor by such official compendium.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile diluent when preparing a stock 
solution for use in making further dilutions for microbial 
susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:

[[Page 1041]]

    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement ``For laboratory diagnostic use only''.
    (b) The statement ``Sterile''.
    (c) The batch mark.
    (d) The number of milligrams of doxycycline in each immediate 
container.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The doxycycline hyclate used in making the batch for potency, 
moisture, pH, doxycycline content, identity, and crystallinity.
    (b) The batch for potency, sterility moisture, and pH.
    (ii) Samples required:
    (a) The doxycycline hyclate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Transfer a 10-milliliter 
aliquot to a 100-milliliter volumetric flask and dilute to volume with 
0.1N hydrochloric acid. Further dilute an aliquot of this solution with 
0.1M potassium phosphate buffer, pH 4.5 (solution 4), to the reference 
concentration of 0.1 microgram of doxycycline per milliliter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.



Sec. 460.55  Lincomycin hydrochloride monohydrate diagnostic sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Lincomycin hydrochloride monohydrate 
diagnostic sensitivity powder is lincomycin hydrochloride monohydrate 
powder packaged in vials and intended for use in clinical laboratories 
for determining in vitro the sensitivity of microorganisms to 
lincomycin. Each vial contains 20 milligrams of lincomycin. Its potency 
is satisfactory if it is not less than 90 percent and not more than 120 
percent of the number of milligrams of lincomycin that it is represented 
to contain. It is sterile. Its moisture content is not more than 7 
percent. It gives a positive identity test for lincomycin hydrochloride 
monohydrate. The lincomycin hydrochloride monohydrate used conforms to 
the standards prescribed by Sec. 453.30(a)(1) (i), (iii), (iv), (v), and 
(ix) of this chapter.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass, and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile broth medium when preparing a 
stock solution for use in making serial dilutions for microbial 
susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statements ``Not for therapeutic use'' and ``For laboratory 
diagnosis only''.
    (b) The statement ``Sterile.''
    (c) The batch mark.
    (d) The number of milligrams of lincomycin in each immediate 
container.
    (e) The statements ``Store in a refrigerator'' and ``Reconstituted 
solutions should be refrigerated.''
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.

[[Page 1042]]

    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The lincomycin hydrochloride monohydrate used in making the 
batch for potency, moisture, pH, crystallinity, and specific rotation.
    (b) The batch for potency, sterility, moisture, and identity.
    (ii) Samples required:
    (a) The lincomycin hydrochloride monohydrate used in making the 
batch: 10 packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 30 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an aliquot with 0.1M 
potassium phosphate buffer, pH 8.0 (solution 3), to the reference 
concentration of 2.0 micrograms of lincomycin per milliliter 
(estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) Identity. In a 100-milliliter beaker, dissolve sufficient sample 
to yield a concentration of at least 80 milligrams per milliliter, using 
no more than 2.0 milliliters of water. Add acetone until precipitation 
begins and then add an additional 20 milliliters of acetone. Filter the 
solution through filter paper and wash with two 10-milliliter portions 
of acetone. Expose the residue at room temperature until it is dry 
enough to be reduced to moderately fine particles. Dry the material for 
4 hours in a 60 deg. C. vacuum oven. After drying the material, care 
must be taken to avoid extended exposure to the atmosphere. The infrared 
spectrum of a mineral oil dispersion of the residue thus obtained 
exhibits maxima at the same wavelengths as that of the lincomycin 
working standards, similarly treated.



Sec. 460.58  Methacycline hydrochloride diagnostic sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Methacycline hydrochloride diagnostic 
sensitivity powder is the crystalline methacycline hydrochloride, with 
or without one or more suitable buffers and diluents, packaged in vials 
and intended for use in clinical laboratories for determining in vitro 
the sensitivity of microorganisms to methacycline. Each vial contains 
methacycline hydrochloride equivalent to 20 milligrams of methacycline. 
The potency of each immediate container is satisfactory if it contains 
not less than 90 percent and not more than 115 percent of its labeled 
content. It is sterile. Its moisture content is not more than 4.0 
percent. When reconstituted as directed in the labeling, its pH is not 
less than 2.0 and not more than 3.5. The methacycline hydrochloride used 
conforms to the standards prescribed by Sec. 446.50(a)(1) (i), (iii), 
(v), and (vi) of this chapter.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile diluent when preparing a stock 
solution for use in making further dilutions for microbial 
susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement ``For laboratory diagnostic use only''.
    (b) The statement ``Sterile.''
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:

[[Page 1043]]

    (i) Results of tests and assays on:
    (a) The methacycline hydrochloride used in making the batch for 
potency, moisture, absorptivity, identity, and crystallinity.
    (b) The batch for potency, sterility, moisture, and pH.
    (ii) Samples required:
    (a) The methacycline hydrochloride used in making the batch: 10 
packages, each containing 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample as follows: 
Reconstitute as directed in the labeling. Dilute an aliquot of this 
solution with 0.1M potassium phosphate buffer, pH 4.5 (solution 4), to 
the reference concentration of 0.06 microgram of methacycline per 
milliliter.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.



Sec. 460.64  Minocycline hydrochloride powder for microbial susceptibility testing.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Minocycline hydrochloride powder for 
microbial susceptibility testing is minocycline hydrochloride with or 
without one or more suitable buffers and diluents, packaged in vials and 
intended for use in clinical laboratories for determining in vitro the 
susceptibility of microorganisms to minocycline. Each vial contains 
minocycline hydrochloride equivalent to 20 milligrams of minocycline. 
The potency of each immediate container is satisfactory if it contains 
not less than 90 percent and not more than 115 percent of its labeled 
content. It is sterile. Its moisture content is not more than 5.0 
percent. When reconstituted as directed in the labeling, its pH is not 
less than 2.0 and not more than 4.0. The minocycline hydrochloride used 
conforms to the standards prescribed by Sec. 446.60(a)(1) (i), (iii), 
(iv), (v), (vi), and (vii) of this chapter.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass, and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile diluent when preparing a stock 
solution for use in making further dilutions for microbial 
susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement ``For laboratory use only.''
    (b) The statement ``Sterile.''
    (c) The batch mark.
    (d) The number of milligrams of minocycline in each immediate 
container.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The minocycline hydrochloride used in making the batch for 
potency, moisture, pH, minocycline content, identity, and crystallinity.
    (b) The batch for potency, sterility, moisture, and pH.
    (ii) Samples required:
    (a) The minocycline hydrochloride used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for

[[Page 1044]]

assay as follows: Reconstitute as directed in the labeling. Dilute an 
aliquot with 0.1M potassium phosphate buffer, pH 4.5 (solution 4), to 
the reference concentration of 0.100 microgram of minocycline per 
milliliter (estimated).
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.



Sec. 460.66  Oleandomycin phosphate diagnostic sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Oleandomycin phosphate diagnostic 
sensitivity powder is oleandomycin phosphate, with or without one or 
more suitable buffers and dilutents, packaged in vials and intended for 
use in clinical laboratories, for determining in vitro the sensitivity 
of microorganisms to oleandomycin. Each vial contains oleandomycin 
phosphate equivalent to 20 milligrams of oleandomycin. The potency of 
each immediate container is satisfactory if it contains not less than 90 
percent and not more than 115 percent of its labeled content. It is 
sterile. Its moisture content is not more than 5.0 percent. When 
reconstituted as directed in the labeling, its pH is not less than 4.0 
and not more than 7.0. Each other substance used, if its name is 
recognized in the U.S.P. or N.F., conforms to the standards prescribed 
therefor by such official compendium.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile diluent when preparing a stock 
solution for use in making further dilutions for microbial 
susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement ``For laboratory diagnostic use only.''
    (b) The statement ``Sterile.''
    (c) The batch mark.
    (d) The number of milligrams of oleandomycin in each immediate 
container.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oleandomycin phosphate used in making the batch for potency, 
moisture, pH, and identity.
    (b) The batch for potency, sterility, moisture, and pH.
    (ii) Samples required:
    (a) The oleandomycin phosphate used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an aliquot with 0.1M 
potassium phosphate buffer, pH 8.0 (solution 3), to the prescribed 
reference concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.



Sec. 460.70  Oxytetracycline hydrochloride diagnostic sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality,

[[Page 1045]]

and purity. Oxytetracycline hydrochloride diagnostic sensitivity powder 
is crystalline oxytetracycline hydrochloride, with or without one or 
more suitable buffers and diluents, packaged in vials and intended for 
use in clinical laboratories for determining in vitro the sensitivity of 
microorganisms to oxytetracycline. Each vial contains oxytetracycline 
hydrochloride equivalent to 20 milligrams of oxytetracycline. The 
potency of each immediate container is satisfactory if it contains not 
less than 90 percent and not more than 115 percent of its labeled 
content. It is sterile. Its loss on drying is not more than 2.0 percent. 
When reconstituted as directed in the labeling, its pH is not less than 
2.0 and not more than 3.5. The oxytetracycline hydrochloride used 
conforms to the standards prescribed by Sec. 446.67a(a)(1) (i), (vi), 
(vii), (viii), and (ix) of this chapter.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile diluent when preparing a stock 
solution for use in making further dilutions for microbial 
susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement ``For laboratory diagnostic use only''.
    (b) The statement ``Sterile''.
    (c) The batch mark.
    (d) The number of milligrams of oxytetracycline in each immediate 
container.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The oxytetracycline hydrochloride used in making the batch for 
potency, moisture, pH, crystallinity, absorptivity, and identity.
    (b) The batch for potency, sterility, loss on drying, and pH.
    (ii) Samples required:
    (a) The oxytetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 30 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an aliquot with 0.1M 
potassium phosphate buffer, pH 4.5 (solution 4), to the prescribed 
reference concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e) (1) of that section.
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.



Sec. 460.75  Potassium penicillin G diagnostic sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Potassium penicillin G diagnostic 
sensitivity powder is crystalline potassium penicillin G, with or 
without one or more suitable buffers and diluents, packaged in vials and 
intended for use in clinical laboratories for determining in vitro the 
sensitivity of microorganisms to penicillin G. Each vial contains 20,000 
units of penicillin G. The potency of each immediate container is 
satisfactory if it contains not less than 90 percent and not more than 
115 percent of its labeled content. It is sterile. Its loss on drying is 
not more than 1.5 percent. When reconstituted as directed in the 
labeling, its pH is not less than 5.0 and not more than 7.5. The 
potassium penicillin G used conforms to the standards prescribed by 
Sec. 440.80a(a)(1) (i), (v), and (vi) of this chapter.

[[Page 1046]]

    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile diluent when preparing a stock 
solution for use in making further dilutions for microbial 
susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement ``For laboratory diagnostic use only''.
    (b) The statement ``Sterile''.
    (c) The batch mark.
    (d) The number of units of penicillin G in each immediate container.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The penicillin G used in making the batch for potency, moisture, 
pH, and crystallinity.
    (b) The batch for potency, sterility, loss on drying, and pH.
    (ii) Samples required:
    (a) The potassium penicillin G used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an aliquot with 1 
percent potassium phosphate buffer, pH 6.0 (solution 1), to the 
prescribed reference concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e) (1) or (2) of that section, 
except if using the method in paragraph (e)(2), use medium B in lieu of 
medium A.
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.



Sec. 460.79  Polymyxin B sulfate diagnostic sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Polymyxin B sulfate diagnostic 
sensitivity powder is polymyxin B sulfate, with or without one or more 
suitable buffers and diluents, packaged in vials and intended for use in 
clinical laboratories for determining in vitro the sensitivity of 
microorganisms to polymyxin B. Each vial contains the equivalent of 
20,000 units of polymyxin B. The potency of each immediate container is 
satisfactory if it contains not less than 90 percent and not more than 
115 percent of its labeled content. It is sterile. Its loss on drying is 
not more than 7.0 percent. When reconstituted as directed in the 
labeling, its pH is not less than 5.0 and not more than 7.5. The 
polymyxin B sulfate used conforms to the standards prescribed by 
Sec. 448.30a(a)(1) (i), (v), (vi), and (ix) of this chapter. Each other 
substance used, if its name is recognized in the U.S.P. or N.F., 
conforms to the standards prescribed therefor by such official 
compendium.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile diluent when preparing a stock 
solution for use in making further dilutions for microbial 
susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement ``For laboratory diagnostic use only''.

[[Page 1047]]

    (b) The statement ``Sterile''.
    (c) The batch mark.
    (d) The number of units of polymyxin B in each immediate container.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The polymyxin B sulfate used in making the batch for potency, 
moisture, pH, and identity.
    (b) The batch for potency, sterility, loss on drying, and pH.
    (ii) Samples required:
    (a) The polymyxin B sulfate used in making the batch: 10 packages, 
each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.105 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an aliquot with 10 
percent potassium phosphate buffer, pH 6.0 (solution 6), to the 
prescribed reference concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.



Sec. 460.86  Spectinomycin hydrochloride powder for microbial susceptibility testing.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Spectinomycin hydrochloride powder for 
microbial susceptibility testing is spectinomycin dihydrochloride 
pentahydrate with or without one or more suitable buffers and diluents, 
packaged in vials and intended for use in clinical laboratories for 
determining in vitro the susceptibility of microorganisms to 
spectinomycin. Each vial contains spectinomycin hydrochloride equivalent 
to 100 milligrams of spectinomycin. The potency of each immediate 
container is satisfactory if it contains not less than 90 percent and 
not more than 115 percent of its labeled content. It is sterile. Its 
moisture content is not more than 8 percent. When reconstituted as 
directed in the labeling, its pH is not less than 3.8 nor more than 5.6. 
The spectinomycin hydrochloride used conforms to the standards 
prescribed by Sec. 455.80a(a)(1) (i), (ii), (vii), (viii), (ix), (x), 
and (xi) of this chapter.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of the amount of sterile diluent prescribed in the labeling 
when preparing a stock solution for use in making further dilutions for 
microbial susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement, ``For laboratory use only''.
    (b) The statement, ``Sterile''.
    (c) The batch mark.
    (d) The number of milligrams of spectinomycin in each immediate 
container.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The spectinomycin hydrochloride used in making the batch for 
spectinomycin content, microbiological activity, moisture, pH, identity, 
residue on ignition, and crystallinity.

[[Page 1048]]

    (b) The batch for potency, sterility, moisture, and pH.
    (ii) Samples required:
    (a) The spectinomycin hydrochloride used in making the batch: 10 
packages, each containing approximately 300 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an aliquot with sterile 
distilled water to the prescribed reference concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Moisture. Proceed as directed in Sec. 436.201 of this chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.



Sec. 460.89  Streptomycin sulfate diagnostic sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Streptomycin sulfate diagnostic 
sensitivity powder is streptomycin sulfate, with or without one or more 
suitable buffers and diluents, packaged in vials and intended for use in 
clinical laboratories for determining in vitro the sensitivity of 
microorganisms to streptomycin. Each vial contains streptomycin sulfate 
equivalent to 20 milligrams of streptomycin. The potency of each 
immediate container is satisfactory if it contains not less than 90 
percent and not more than 115 percent of its labeled content. It is 
sterile. Its loss on drying is not more than 5.0 percent. When 
reconstituted as directed in the labeling, its pH is not less than 4.5 
and not more than 7.0. The streptomycin sulfate used conforms to the 
standards prescribed by Sec. 444.70a(a)(1) (i), (vi), and (vii) of this 
chapter. Each other substance used, if its name is recognized in the 
U.S.P. or N.F., conforms to the standards prescribed therefor by such 
official compendium.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile diluent when preparing a stock 
solution for use in making further dilutions for microbial 
susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement ``For laboratory diagnostic use only''.
    (b) The statement ``Sterile''.
    (c) The batch mark.
    (d) The number of milligrams of streptomycin in each immediate 
container.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The streptomycin sulfate used in making the batch for potency, 
loss on drying, and pH.
    (b) The batch for potency, sterility, loss on drying, and pH.
    (ii) Samples required:
    (a) The streptomycin sulfate used in making the batch: 10 packages, 
each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an aliquot with sterile 
distilled water to the prescribed reference concentration.

[[Page 1049]]

    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the drug reconstituted as directed in the labeling.



Sec. 460.93  Tetracycline hydrochloride diagnostic sensitivity powder.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline hydrochloride diagnostic 
sensitivity powder is crystalline tetracycline hydrochloride, with or 
without one or more suitable buffers and diluents, packaged in vials and 
intended for use in clinical laboratories for determining in vitro the 
sensitivity of microorganisms to tetracycline. Each vial contains 20 
milligrams of tetracycline hydrochloride. The potency of each immediate 
container is satisfactory if it contains not less than 90 percent and 
not more than 115 percent of its labeled content. It is sterile. Its 
loss on drying is not more than 2.0 percent. When reconstituted as 
directed in the labeling, its pH is not less than 1.8 and not more than 
3.0. The tetracycline hydrochloride used conforms to the standards 
prescribed by Sec. 446.81a(a)(1) (i), (vi), and (vii), and (viii) of 
this chapter. Each other substance used, if its name is recognized in 
the U.S.P. or N.F., conforms to the standards prescribed therefor by 
such official compendium.
    (2) Packaging. The immediate container shall be of colorless, 
transparent glass and it shall be a tight container as defined by the 
U.S.P. It shall be so sealed that the contents cannot be used without 
destroying such seal. It shall be of appropriate size to permit the 
addition of 20 milliliters of sterile diluent when preparing a stock 
solution for use in making further dilutions for microbial 
susceptibility testing.
    (3) Labeling. In addition to the requirements of Sec. 432.5(a)(3) of 
this chapter, each package shall bear on its label or labeling, as 
hereinafter indicated, the following:
    (i) On its outside wrapper or container and on the immediate 
container:
    (a) The statement ``For laboratory diagnostic use only.''
    (b) The statement ``Sterile.''
    (c) The batch mark.
    (d) The number of milligrams of tetracycline hydrochloride in each 
immediate container.
    (ii) On the circular or other labeling within or attached to the 
package, adequate information for use of the drug in the clinical 
laboratory.
    (4) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The tetracycline hydrochloride used in making the batch for 
potency, moisture, pH, crystallinity, and absorptivity.
    (b) The batch for potency, sterility, loss on drying, and pH.
    (ii) Samples required:
    (a) The tetracycline hydrochloride used in making the batch: 10 
packages, each containing approximately 500 milligrams.
    (b) The batch:
    (1) For all tests except sterility: A minimum of 20 immediate 
containers.
    (2) For sterility testing: 20 immediate containers, collected at 
regular intervals throughout each filling operation.
    (b) Tests and methods of assay--(1) Potency. Proceed as directed in 
Sec. 436.106 of this chapter, preparing the sample for assay as follows: 
Reconstitute as directed in the labeling. Dilute an aliquot with 0.1M 
potassium phosphate buffer, pH 4.5 (solution 4), to the prescribed 
reference concentration.
    (2) Sterility. Proceed as directed in Sec. 436.20 of this chapter, 
using the method described in paragraph (e)(1) of that section.
    (3) Loss on drying. Proceed as directed in Sec. 436.200(b) of this 
chapter.
    (4) pH. Proceed as directed in Sec. 436.202 of this chapter using 
the drug reconstituted as directed in the labeling.



                  Subpart C--Susceptibility Test Panels

    Source:  43 FR 9793, Mar. 10, 1978, unless otherwise noted.

[[Page 1050]]



Sec. 460.100  Antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Antimicrobial susceptibility test panels 
are polystyrene trays molded with separate wells which contain frozen 
aliquots of antibiotic and non-antibiotic antimicrobial solutions in 
Mueller-Hinton broth. The trays are used in clinical laboratories for 
determining susceptibility of microorganisms to antimicrobial drugs. The 
broth antimicrobial solutions are prepared from serially diluted 
antimicrobial stock solutions. The concentrated aqueous antimicrobial 
stock solutions must conform to the requirements for certification 
prescribed by this section.
    (2) Labeling. In addition to the requirements of Secs. 432.5 and 
809.10 of this chapter, each test panel shall bear on its label or 
labeling, as hereinafter indicated, the following:
    (i) On the outside wrapper or immediate container of trays:
    (a) The batch mark.
    (b) The name and potency of each solution in the batch according to 
the following:

                                                                        
------------------------------------------------------------------------
                                             Content of antimicrobic in 
               Name of drug                   micrograms per milliliter 
------------------------------------------------------------------------
Ampicillin (gram-positive panel)..........  8, 4, 2, 1, 0.5, 0.25, 0.12.
Ampicillin (gram-negative panel)..........  16, 8, 4, 2, 1, 0.5, 0.25.  
Carbenicillin.............................  512, 256, 128, 64, 32, 16,  
                                             8.                         
Cephalothin...............................  64, 32, 16, 8, 4, 2, 1.     
Chloramphenicol...........................  32, 16, 8, 4, 2, 1, 0.5.    
Clindamycin...............................  16, 8, 4, 2, 1, 0.5, 0.25.  
Colistin..................................  4.                          
Erythromycin..............................  16, 8, 4, 2, 1, 0.5, 0.25.  
Gentamicin................................  16, 8, 4, 2, 1, 0.5, 0.25.  
Kanamycin.................................  64, 32, 16, 8, 4, 2, 1.     
Methicillin...............................  16, 8, 4, 2, 1, 0.5, 0.25.  
Penicillin G..............................  4, 2, 1, 0.5, 0.25, 0.12,   
                                             0.06.                      
Tetracycline..............................  16, 8, 4, 2, 1, 0.5, 0.25.  
Tobramycin................................  16, 8, 4, 2, 1, 0.5, 0.25.  
Nitrofurantoin............................  64.                         
Trimethoprim (Gram-positive and gram-       32, 16, 8, 4, 2, 1, 0.5.    
 negative panel).                                                       
Trimethoprim (Combination identification    2.                          
 panel).                                                                
Sulfamethoxazole (Gram-positive and gram-   608, 304, 152, 76, 38, 19,  
 negative panel).                            9.5.                       
Sulfamethoxazole (Combination               38.                         
 identification panel).                                                 
------------------------------------------------------------------------

    (c) The statement ``For in vitro diagnostic use''.
    (ii) On each tray: The name of the panel, the expiration date, and 
the batch mark, including filling operation identification.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The concentrated antimicrobial stock solutions used in making 
the batch for potency and pH.
    (b) The antimicrobial susceptibility test panels from the batch for 
performance and identity.
    (ii) Samples required: (a) The concentrated antimicrobial stock 
solutions used in making the batch as directed in each individual 
monograph.
    (b) The batch: A minimum of 25 panels selected at such intervals 
throughout the entire time of the filling operation so that the 
quantities of panels filled during the intervals are approximately 
equal.
    (b) Tests and methods of assay--(1) Performance--(i) Procedure. Test 
randomly selected panels with each of the four test organisms as 
follows: Use the test organism suspensions prepared as described in 
Sec. 460.6(b) (14), (15), (16), and (17). Transfer 0.005 milliliter of 
the appropriate test organism suspension into all wells of a panel. For 
the purpose of this section, wells are identified 1-10 from left to 
right and A-H from front to rear. Incubate inoculated panels in groups 
of three or less, each with a clean cover, at 35 deg. C for 16 to 18 
hours. For each control organism, the lowest concentration showing 
complete inhibition of growth is the minimal inhibitory concentration 
for that particular antimicrobial agent (referred to hereafter as its 
end point). For the purpose of this section, an on-scale and point means 
an end point which has been established by growth in the next lower 
concentration well. No growth in any well in row G does not indicate an 
on-scale end point unless the next lower concentration can be shown to 
produce growth. Establishment of an end point for a no-growth result in 
a well in row G requires additional testing in which 0.1 milliliter of 
sterile medium N is added to well G in some panels prior to inoculation 
with the test organism. An on-scale end point in row

[[Page 1051]]

G is established by no growth in the undiluted well G and growth in the 
two-fold diluted well G.
    (ii) Evaluation. Susceptibility test panels of each filling 
operation pass the performance test if the on-scale end point for each 
antimicrobial agent and each test organism meet the limits specified in 
the following table (allowable off-scale end points are identified by 
asterisks):

                                     Performance End Point Acceptance Limits                                    
----------------------------------------------------------------------------------------------------------------
                                                     end points (micrograms per milliliter)                     
                               ---------------------------------------------------------------------------------
         Antimicrobic             E. coli (ATCC    S. faecalis (ATCC   S. aureus (ATCC     P. aeruginosa (ATCC  
                                      25922)             29212)             29213)                27853)        
----------------------------------------------------------------------------------------------------------------
Ampicillin....................  1-4..............  0.5-2............  0.25-1...........  Greater than 8 (gram-  
                                                                                          positive panel).      
                                                                                          Greater than 16 (gram-
                                                                                          negative panel).      
Carbenicillin.................  4*-16............  16-64............  No growth........  16-64.                 
Cephalothin...................  4-16.............  16-64............  ......do.........  64-greater than 64.    
Chloramphenicol...............  2-8..............  4-16.............  4-16.............  Greater than 32.       
Clindamycin...................  16-greater than    4-16.............  No growth........  Greater than 16.       
                                 16.                                                                            
Colistin......................  No growth........  Growth...........  Growth...........  No growth.             
Erythromycin..................  16-greater than    1-4..............  0.125*-0.5.......  Greater than 16.       
                                 16.                                                                            
Gentamicin....................  0.125*-0.5.......  4-16.............  0.125*-0.5.......  0.125*-0.5             
Kanamycin.....................  1-4..............  16-64............  0.5*-2...........  Greater than 64.       
Methicillin...................  Greater than 16..  Greater than 16..  0.5-2............  Greater than 16.       
Nitrofurantoin................  No growth........  No growth........  No growth........  Growth.                
Penicillin G..................  Greater than 4...  1-4..............  0.125-0.5........  Greater than 4.        
Tetracycline..................  0.25-1...........  8-greater than 16  0.125*-0.5.......  4-16.                  
Trimethoprim-sulfa-methoxazole  No growth........  No growth........  No growth........  16/304-greater than 32/
                                                                                          608 (gram-positive and
                                                                                          gram-negative panel). 
                                ......do.........  ......do.........  ......do.........  Growth (combination    
                                                                                          identification panel).
Tobramycin....................  0.25-1...........  8-greater than 16  0.125*-0.5.......  No growth.             
----------------------------------------------------------------------------------------------------------------
* An additional two-fold dilution below well G.                                                                 


                                                                                                                
----------------------------------------------------------------------------------------------------------------
                    Test organism                                          Identity test data                   
----------------------------------------------------------------------------------------------------------------
                                                                            Type of panel and                   
                                                                               well to be                       
                                           Suspension     Antibiotics          inoculated                       
             Name                ATCC No.      No.           tested      ----------------------     Response    
                                                                            Gram-      Gram-                    
                                                                           positive   negative                  
----------------------------------------------------------------------------------------------------------------
S. aureus.....................      29247          18  Methicillin......         3A  .........  No growth.      
                                                       Penicillin.......         4A  .........  Growth.         
                                                       Erythromycin.....         2E  .........  Do.             
                                                       Cephalothin......         6D         2D  No growth.      
                                                       Clindamycin......         1E         4D  Do.             
                                                       Tetracycline.....         8D  .........  Growth.         
----------------------------------------------------------------------------------------------------------------
Ent. cloacae..................      29249          19  Cephalothin......  .........         2B  Do.             
                                                       Kanamycin........  .........         7B  No growth.      
----------------------------------------------------------------------------------------------------------------
Ps. aeruginosa................      29248          20  Gentamicin.......  .........         3D  Growth.         
                                                       Tobramycin.......  .........         8D  No growth.      
----------------------------------------------------------------------------------------------------------------
(Rows are numbered 1-10 from left to right; A-H from front to rear.)                                            


Transfer 0.005 milliliter of the test organism suspension into 
designated wells of the panel. Incubate the panels at 35 deg. C for 16 
to 18 hours in covered stacks of three or fewer panels. Read the 
designated wells for growth or no growth.

[[Page 1052]]

    (ii) Evaluation. Each susceptibility test panel passes the identity 
test if the determinations of growth in the designated wells agree with 
those shown in the table in paragraph (b)(2)(i) of this section.
[43 FR 9793, Mar. 10, 1978; 43 FR 12859, Mar. 28, 1978]



Sec. 460.110  Ampicillin concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Ampicillin concentrated stock solutions 
for use in preparing antimicrobial susceptibility test panels are frozen 
aqueous ampicillin trihydrate stock solutions serially diluted with 
distilled water to contain approximately 3,200, 1,600, 800, 400, 200, 
100, and 50 micrograms ampicillin per milliliter. The potency of each 
solution is satisfactory if it is not less than 100 percent and not more 
than 150 percent of the number of micrograms of ampicillin that it is 
represented to contain. The pH of the solution containing 3,200 
micrograms of ampicillin per milliliter is not less than 4.0 and not 
more than 7.0. The ampicillin trihydrate used conforms to the 
requirements of Sec. 440.7(a)(1) (i), (iii), (iv), (v), (vi), (vii), and 
(viii) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing at least 
2.5 milliliters.
    (b) Tests and methods of assay. The sample solutions must be thawed 
and brought to room temperature before testing.
    (1) Potency. Proceed as directed in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Dilute an accurately measured 
representative portion of the sample with 0.1M potassium phosphate 
buffer, pH 8.0 (solution 3), to the reference concentration of 0.1 
microgram of ampicillin per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution containing 3,200 micrograms of ampicillin per milliliter.



Sec. 460.113  Carbenicillin concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Carbenicillin concentrated stock 
solutions for use in preparing antimicrobial susceptibility test panels 
are frozen aqueous carbenicillin disodium stock solutions serially 
diluted with distilled water to contain approximately the following 
concentrations: 20,480, 10,240, 5,120, 2,560, 1,280, 640, and 320 
micrograms of carbenicillin per milliliter. The potency of each solution 
is satisfactory if it is not less than 100 percent and not more than 150 
percent of the number of micrograms of carbenicillin that it is 
represented to contain. The pH of the solution containing 20,480 
micrograms of carbenicillin per milliliter is not less than 6.0 and not 
more than 8.0. The carbenicillin disodium used conforms to the 
requirements of Sec. 440.13a (a)(1) (i), (v), (vi), and (vii) of this 
chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing at least 5 
milliliters.
    (b) Tests and methods of assay. The sample solutions must be thawed 
and brought to room temperature before testing.
    (1) Potency. Proceed as directed in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Dilute an accurately measured 
representative portion of the sample with 1.0 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to the reference concentration of 
20 micrograms of carbenicillin per milliliter (estimated).

[[Page 1053]]

    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution containing 20,480 micrograms of carbenicillin per 
milliliter.



Sec. 460.116  Cephalothin concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Cephalothin concentrated stock solutions 
for use in preparing susceptibility test panels are frozen cephalothin 
sodium aqueous stock solutions serially diluted with distilled water to 
contain approximately the following concentrations: 2,560, 1,280, 640, 
320, 160, 80, and 40 micrograms of cephalothin per milliliter. The 
potency of each diluted solution is satisfactory if it is not less than 
90 percent and not more than 140 percent of the number of micrograms of 
cephalothin that it is represented to contain. The pH of the solution 
containing 2,560 micrograms of cephalothin per milliliter is not less 
than 4.2 and not more than 7.0. The cephalothin used conforms to the 
standards prescribed by Sec. 442.25a(a)(1) (i), (v), (vi), (vii), 
(viii), and (ix) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing at least 5 
milliliters.
    (b) Tests and methods of assay. The sample solutions used for 
testing must be thawed and brought to room temperature before testing.
    (1) Potency. Proceed as directed in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Dilute an accurately measured 
representative portion of the sample with 1.0 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to the reference concentration of 
1.0 microgram of cephalothin per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution containing 2,560 micrograms of cephalothin per milliliter.



Sec. 460.119  Chloramphenicol concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Chloramphenicol concentrated stock 
solutions for use in preparing susceptibility test panels are frozen 
aqueous chloramphenicol stock solutions serially diluted with distilled 
water to contain approximately the following concentrations: 1,280, 640, 
320, 160, 80, 40, and 20 micrograms of chloramphenicol per milliliter. 
The potency of each diluted solution is satisfactory if it is not less 
than 90 percent and not more than 140 percent of the number of 
micrograms of chloramphenicol that it is represented to contain. The pH 
of the solution containing 1,280 micrograms of chloramphenicol per 
milliliter is not less than 4.5 and not more than 7.5. The 
chloramphenicol used conforms to the standards prescribed by 
Sec. 455.10(a)(1) (i), (iii), (iv), (v), (vi), and (vii) of this 
chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing at least 5 
milliliters.
    (b) Tests and methods of assay. The sample solution must be thawed 
and brought to room temperature before further testing.
    (1) Potency. Proceed as directed in Sec. 436.106 of this chapter, 
preparing the sample for assay as follows: Dilute an accurately measured 
representative portion of the sample with distilled water to the 
reference concentration of 2.5 micrograms of chloramphenicol per 
milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter using 
the solution containing 1,280 micrograms of chloramphenicol per 
milliliter.

[[Page 1054]]



Sec. 460.122  Clindamycin concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Clindamycin concentrated stock solutions 
for use in preparing susceptibility test panels are frozen aqueous 
clindamycin hydrochloride stock solutions serially diluted with 
distilled water to contain approximately the following concentrations: 
640, 320, 160, 80, 40, 20, and 10 micrograms of clindamycin per 
milliliter. The potency of each diluted solution is satisfactory if it 
is not less than 90 percent and not more than 140 percent of the number 
of micrograms of clindamycin that it is represented to contain. The pH 
of the solution containing 640 micrograms of clindamycin per milliliter 
is not less than 4.5 and not more than 7.0. The clindamycin used 
conforms to the standards prescribed by Sec. 453.20(a)(1) (i), (ii), 
(iv), (v), (vi) and (vii) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing at least 5 
milliliters.
    (b) Tests and methods of assay. The sample solutions must be thawed 
and brought to room temperature before testing.
    (1) Potency. Proceed as directed in Sec. 436.105 of this chapter. 
Prepare the sample for assay by diluting an accurately measured 
representative portion of the sample with 0.1M potassium phosphate 
buffer, pH 8.0 (solution 3), to the reference concentration of 1.0 
microgram of clindamycin per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution containing 640 micrograms of clindamycin per milliliter.



Sec. 460.125  Colistin concentrated stock solution for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Colistin concentrated stock solutions for 
use in preparing antimicrobial susceptibility test panels are frozen 
aqueous colistin sulfate stock solutions serially diluted with distilled 
water to contain an approximate concentration of 160 micrograms of 
colistin per milliliter. Its potency is satisfactory if it is not less 
than 90 percent and not more than 140 percent of the number of 
micrograms of colistin that it is represented to contain. Its pH is not 
less than 5.0 and not more than 8.0. The colistin used conforms to the 
requirements of Sec. 448.21(a)(1) (i), (iii), (iv), and (v) of this 
chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: Five frozen aliquots of the concentrated 
stock solution containing at least 5 milliliters.
    (b) Tests and methods of assay. The sample solution must be thawed 
and brought to room temperature before testing.
    (1) Potency. Proceed as directed in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Dilute an accurately measured 
representative portion of the sample with 10 percent potassium phosphate 
buffer (solution 6), to the reference concentration of 1.0 microgram of 
colistin per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution without further dilution.



Sec. 460.128  Erythromycin concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Erythromycin concentrated stock solutions 
for use in preparing antimicrobial susceptibility test panels are frozen 
aqueous erythromycin stock

[[Page 1055]]

solutions serially diluted with 0.1 M potassium phosphate buffer, pH 8.0 
(solution 3) to contain approximately the following concentrations: 
3,200, 1,600, 800, 400, 200, 100, and 50 micrograms of erythromycin per 
milliliter. The potency of each solution is satisfactory if it is not 
less than 90 percent and not more than 140 percent of the number of 
micrograms of erythromycin that it is represented to contain. The pH of 
the solution containing 3,200 micrograms of erythromycin per milliliter 
is not less than 7.5 and not more than 10.0. The erythromycin used 
conforms to the requirements of Sec. 452.10(a)(1) (i), (iii), (iv), 
(vii), and (viii) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing at least 5 
milliliters.
    (b) Tests and methods of assay. The sample solutions must be thawed 
and brought to room temperature before testing.
    (1) Potency. Proceed as directed in Sec. 436.105 of this chapter. 
Prepare the sample for assay by diluting an accurately measured 
representative portion of the sample with 0.1 M potassium phosphate 
buffer, pH 8.0 (solution 3), to the reference concentration of 1.0 
microgram of erythromycin per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution containing 3,200 micrograms of erythromycin per milliliter.



Sec. 460.131  Gentamicin concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Gentamicin concentrated stock solutions 
for use in preparing susceptibility test panels are frozen aqueous 
gentamicin sulfate stock solutions serially diluted with distilled water 
to contain approximately the following concentrations: 640, 320, 160, 
80, 40, 20, and 10 micrograms of gentamicin per milliliter. The potency 
of each diluted solution is satisfactory if it is not less than 90 
percent and not more than 140 percent of the number of micrograms of 
gentamicin that it is represented to contain. The pH of the solution 
containing 640 micrograms of gentamicin per milliliter is not less than 
4.5 and not more than 7.0. The gentamicin used conforms to the standards 
prescribed by Sec. 444.20(a)(1) (iii), (iv), (v), and (vii) of this 
chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain.
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing at least 5 
milliliters.
    (b) Tests and methods of assay. The sample solutions must be thawed 
and brought to room temperature before testing.
    (1) Potency. Proceed as directed in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Dilute an accurately measured 
representative portion of the sample with 0.1M potassium phosphate 
buffer, pH 8.0 (solution 3), to the reference concentration of 0.1 
microgram of gentamicin per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution containing 640 micrograms of gentamicin per milliliter.



Sec. 460.134  Kanamycin concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Kanamycin concentrated stock solutions 
for use in preparing susceptibility test panels are frozen aqueous 
kanamycin sulfate stock solutions serially diluted with distilled water 
to contain approximately the following concentrations: 2,560, 1,280, 
640, 320, 160, 80, and 40 micrograms of

[[Page 1056]]

kanamycin per milliliter. The potency of each diluted solution is 
satisfactory if it is not less than 90 percent and not more than 140 
percent of the number of micrograms of kanamycin that it is represented 
to contain. The pH of the solution containing 2,560 micrograms of 
kanamycin per milliliter is not less than 6.5 and not more than 8.5. The 
kanamycin used conforms to the standards prescribed by Sec. 444.30(a)(1) 
(i), (iii), (iv), (vi), (vii), and (viii) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing at least 5 
milliliters.
    (b) Tests and methods of assay. The sample solutions must be thawed 
and brought to room temperature before testing.
    (1) Performance. Proceed as directed in Sec. 436.106 of this 
chapter, preparing the sample for assay as follows: Dilute an accurately 
measured representative portion with distilled water to the reference 
concentration of 10 micrograms of kanamycin per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution containing 2,560 micrograms of kanamycin per milliliter.



Sec. 460.137  Methicillin concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Methicillin concentrated stock solutions 
for use in preparing susceptibility test panels are frozen aqueous 
methicillin sodium stock solutions serially diluted with distilled water 
to contain approximately 6,400, 3,200, 1,600, 800, 400, 200, and 100 
micrograms of methicillin per milliliter. The potency of each diluted 
solution is satisfactory if it is not less than 100 percent and not more 
than 150 percent for the number of micrograms of methicillin that it is 
represented to contain. The pH of the solution containing 6,400 
micrograms of methicillin per milliliter is not less than 5.0 and not 
more than 7.5. The methicillin used conforms to the standards prescribed 
by Sec. 440.36a(a)(1) (i), (v), (vi), (vii), (viii), and (ix) of this 
chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Request for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing at least 
2.5 milliliters.
    (b) Tests and methods of assay. The sample solutions must be thawed 
and brought to room temperature before testing.
    (1) Potency. Proceed as directed in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Dilute an accurately measured 
representative portion of the sample with 1.0 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to the reference concentration of 
10 micrograms of methicillin per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution containing 6,400 micrograms of methicillin per milliliter.



Sec. 460.140  Penicillin G concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Penicillin G concentrated stock solutions 
for use in preparing antimicrobial susceptibility test panels are frozen 
aqueous penicillin G potassium solutions serially diluted with distilled 
water to contain approximately 1,600, 800, 400, 200, 100, 50, and 25 
micrograms of penicillin G per milliliter. The potency of each diluted 
solution is satisfactory if it is not less than 100 percent and not more 
than 150 percent of the number of micrograms of penicillin G that it is 
represented to

[[Page 1057]]

contain. The pH of the solution containing 1,600 micrograms of 
penicillin G per milliliter is not less than 5.0 and not more than 7.5. 
The penicillin G potassium used conforms to the standards prescribed by 
Sec. 440.80a(a)(1) (i), (v) and (vi) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing at least 
2.5 milliliters.
    (b) Tests and methods of assay. The sample solutions must be thawed 
and brought to room temperature before testing.
    (1) Potency. Proceed as directed in Sec. 436.105 of this chapter, 
preparing the sample for assay as follows: Dilute an accurately measured 
representative portion of the sample with 1.0 percent potassium 
phosphate buffer, pH 6.0 (solution 1), to the reference concentration of 
1.0 unit (0.600 microgram) of penicillin G per milliliter (estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution containing 1,600 micrograms of penicillin G per milliliter.



Sec. 460.146  Tetracycline concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tetracycline concentrated stock solutions 
for use in preparing antimicrobial susceptibility test panels are frozen 
aqueous tetracycline hydrochloride stock solutions serially diluted with 
distilled water to contain approximately the following concentrations: 
640, 320, 160, 80, 40, 20, and 10 micrograms of tetracycline per 
milliliter. The potency of each diluted solution is satisfactory if it 
is not less than 90 percent and not more than 140 percent of the number 
of micrograms of tetracycline that it is represented to contain. The pH 
of the solution containing 640 micrograms of tetracycline per milliliter 
is not less than 3.0 and not more than 7.0. The tetracycline 
hydrochloride used conforms to the standards prescribed by 
Sec. 446.81a(a)(1) (i), (vi), (vii), and (viii) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing at least 5 
milliliters.
    (b) Tests and methods of assay. The sample solutions must be thawed 
and brought to room temperature before testing.
    (1) Potency. Proceed as directed in Sec. 436.106 of this chapter, 
preparing the sample for assay as follows: Dilute an accurately measured 
representative portion of the sample with distilled water to the 
reference concentration of 0.24 microgram of tetracycline per milliliter 
(estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution containing 640 micrograms of tetracycline per milliliter.



Sec. 460.149  Tobramycin concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Tobramycin concentrated stock solutions 
for use in preparing antimicrobial susceptibility test panels are frozen 
aqueous tobramycin sulfate stock solutions serially diluted with 
distilled water to contain approximately the following concentrations: 
1,280, 640, 220, 160, 80, 40, and 20 micrograms of tobramycin per 
milliliter. The potency of each diluted solution is satisfactory if it 
is not less than 90 percent and not more than 140 percent of the number 
of micrograms of tobramycin that it is represented to contain. The pH of 
the solution containing 1,280 micrograms of tobramycin per milliliter is 
not less than 8.5 and not more than 10.5. The tobramycin

[[Page 1058]]

used conforms to the standards prescribed by Sec. 444.80(a)(1)(i), 
(iii), (iv), (v), and (vi) of this chapter.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing 
approximately 5.0 milliliters.
    (b) Tests and methods of assay. The sample solutions must be thawed 
and brought to room temperature before testing.
    (1) Potency. Proceed as directed in Sec. 436.106 of this chapter, 
preparing the sample for assay as follows: Dilute an accurately measured 
representative portion of the sample with distilled water to the 
reference concentration of 2.5 micrograms of tobramycin per milliliter 
(estimated).
    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution containing 1,280 micrograms of tobramycin per milliliter.



Sec. 460.152  Trimethoprim concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Trimethoprim concentrated stock solutions 
for use in antimicrobial susceptibility test panels are frozen aqueous 
acidified trimethoprim lactate stock solutions serially diluted with 
distilled water to contain approximately the following concentrations: 
6,400, 3,200, 1,600, 800, 400, 200, and 100 micrograms of trimethoprim 
per milliliter, or to a single concentration of 400 micrograms of 
trimethoprim per milliliter. The potency of each diluted solution is 
satisfactory if it is not less than 90 percent and not more than 140 
percent of the number of micrograms of trimethoprim that it is 
represented to contain. The pH of the solution, containing 400 
micrograms of trimethoprim per milliliter, is not less than 2.5 and not 
more than 6.0. The trimethoprim lactate used is a white, odorless, 
crystalline powder. Its potency is not less than 74 percent nor more 
than 78 percent trimethoprim. Its melting range is between 183 deg. C 
and 187 deg. C. Its loss on drying is not more than 1.0 percent. It 
passes the identity test. It conforms to the standards prescribed by 
this section.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on:
    (a) The batch for potency and pH.
    (b) The trimethoprim lactate used in preparing the solutions for 
potency, melting range, loss on drying, and identity.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing at least 5 
milliliters.
    (b) Tests and methods of assay--(1) Trimethoprim stock solution. The 
sample solutions must be thawed and brought to room temperature before 
testing.
    (i) Potency--(a) Working standard. Accurately weigh approximately 52 
milligrams of the trimethoprim working standard. Dissolve and dilute the 
working standard in 100 milliliters of 0.1N hydrochloric acid to make a 
stock solution, containing approximately 400 micrograms of trimethoprim 
per milliliter. Further dilute the working standard twentyfold in 
distilled water to approximately 20 micrograms of trimethoprim per 
milliliter.
    (b) Preparation of sample. The sample solution must be thawed and 
brought to room temperature. Further dilute with distilled water to an 
estimated concentration of 20 micrograms of trimethoprim per milliliter.
    (c) Procedure. Using a suitable spectrophotometer equipped with 1.0 
centimeter cells, determine the absorbance of the sample and working 
standard solutions at a wavelength of 270 nanometers.
    (d) Calculations. Calculate the potency of the trimethoprim 
solutions as follows:

    Micrograms of trimethoprim per milliliter in trimethoprim 
lactate=sample absorbance

[[Page 1059]]

times weight of standard (mg) times 0.763 times f times 1,000 times 
purity of standard in percent, divided by standard absorbance times 100 
times 20 times 100

where:

    f=dilution factor of each sample solution.

    (ii) pH. [Reserved]
    (2) Trimethoprim--(i) Potency. Proceed as directed in Sec. 436.213 
of this chapter using the method described in paragraph (e)(2) of that 
section except, to prepare the sample for assay, dissolve approximately 
50 milligrams of sample accurately weighed in 60 milliliters of glacial 
acetic acid. Calculate the trimethoprim content as follows:

Percent trimethoprim=(V1-V2) (N) (290.3) (100) 
          (100) divided by (100-M) times W

where:
V1=Volume perchloric acid used to titrate sample;
V2=Volume perchloric acid used to titrate blank;
W=Sample weight in milligrams;
N=Normality of perchloric acid reagent;
M=Percent moisture in the sample.

    (ii) Melting range. Proceed as directed in Sec. 436.209 of this 
chapter.
    (iii) Loss on drying. Proceed as directed in Sec. 436.200(e) of this 
chapter.
    (iv) Identity. Proceed as directed in Sec. 436.211 of this chapter, 
using a 0.25 percent potassium bromide disc prepared as described in 
paragraph (b)(1) of that section.
[43 FR 9793, Mar. 10, 1978; 43 FR 12859, Mar. 28, 1978]



Sec. 460.153  Sulfamethoxazole concentrated stock solutions for use in antimicrobial susceptibility test panels.

    (a) Requirements for certification--(1) Standards of identity, 
strength, quality, and purity. Sulfamethoxazole concentrated stock 
solutions for use in antimicrobial susceptibility test panels are frozen 
aqueous alkaline sulfamethoxazole stock solutions serially diluted with 
distilled water containing approximately the following concentrations: 
12,160, 6,080, 3,040, 1,520, 760, 380, and 190 micrograms of 
sulfamethoxazole per milliliter, or to a single concentration of 760 
micrograms of sulfamethoxazole per milliliter. The potency of each 
diluted solution is satisfactory if it is not less than 90 percent and 
not more than 140 percent of the number of micrograms of 
sulfamethoxazole that it is represented to contain. The pH of the 
solution containing 760 micrograms of sulfamethoxazole per milliliter is 
not less than 9.0 and not more than 12.5. The sulfamethoxazole used 
conforms to the standards prescribed by the National Formulary.
    (2) Labeling. It shall be labeled in accordance with the 
requirements of Sec. 432.5 of this chapter.
    (3) Requests for certification; samples. In addition to complying 
with the requirements of Sec. 431.1 of this chapter, each such request 
shall contain:
    (i) Results of tests and assays on the batch for potency and pH.
    (ii) Samples required: A minimum of five frozen aliquots of each 
dilution of the concentrated stock solutions, each containing at least 
10 milliliters.
    (b) Tests and methods of assay. The sample solutions must be thawed 
and brought to room temperature before testing.
    (1) Potency. Dilute aliquots of each sample in sufficient distilled 
water to make solutions containing 10 micrograms of sulfamethoxazole per 
milliliter. Place approximately 100 milligrams of the standard, 
accurately weighed, into a 100-milliliter volumetric flask and make to 
volume with 0.1N sodium hydroxide. Pipet 1.0 milliliter of this solution 
into a 100-milliliter volumetric flask and make to volume with distilled 
water. Using a suitable spectrophotometer equipped with 1.0 centimeter 
cells, and distilled water as the blank, determine the absorbance of 
sample and standard solutions at 257 nanometers. Calculate the potency 
of the sulfamethoxazole as follows:

    Micrograms of sulfamethoxazole per milliliter=sample absorbance 
times weight of standard (in mcg) times f times purity of standard in 
percent divided by standard absorbance times 100 times 100


where:

    f=Dilution factor of each sample solution.


[[Page 1060]]


    (2) pH. Proceed as directed in Sec. 436.202 of this chapter, using 
the solution containing 760 micrograms of sulfamethoxazole per 
milliliter.
[43 FR 9793, Mar. 10, 1978; 43 FR 12859, Mar. 28, 1978]



PARTS 461-499  [RESERVED]



[[Page 1061]]


                              FINDING AIDS




  --------------------------------------------------------------------

  A list of CFR titles, subtitles, chapters, subchapters and parts and 
an alphabetical list of agencies publishing in the CFR are included in 
the CFR Index and Finding Aids volume to the Code of Federal Regulations 
which is published separately and revised annually.
  Material Approved for Incorporation by Reference
  Table of CFR Titles and Chapters
  Alphabetical List of Agencies Appearing in the CFR
  List of CFR Sections Affected

[[Page 1063]]

            Material Approved for Incorporation by Reference

                      (Revised as of April 1, 1998)

  The Director of the Federal Register has approved under 5 U.S.C. 
552(a) and 1 CFR part 51 the incorporation by reference of the following 
publications. This list contains only those incorporations by reference 
effective as of the revision date of this volume. Incorporations by 
reference found within a regulation are effective upon the effective 
date of that regulation. For more information on incorporation by 
reference, see the preliminary pages of this volume.


21 CFR (PARTS 300 TO 499)

FOOD AND DRUG ADMINISTRATION, DEPARTMENT OF HEALTH AND HUMAN SERVICES
                                                                  21 CFR


American Pharmaceutical Association

  2215 Constitution Ave., NW, Washington, DC 20037
``Outline of Details for Official Microbiological             436.102(b)
  Assays of Antibiotics,'' A. Kirshbaum and B. 
  Arret, ``Journal of Pharmaceutical Sciences,'' 
  Vol 56, No. 4, April 1967, p. 512.


American Statistical Association

  806 15th St. NW., Washington, DC 20005
Cornfield and Mantel's Modification of Karber's        450.240(b)(4)(ii)
  Method published in the ``Journal of the 
  American Statistical Association,'' Vol. 45, pp. 
  194-210 (1950).


``Biometrics'' Managing Editor,

  P.O. Box 5962, Raleigh, NC 27607
Carrol S. Weil Method published in ``Biometrics''     450.20(b)(4)(ii); 
  Vol. 8 pp. 249-263 (1952) pp 619-621.                450.24(b)(5)(ii).


Medical Encyclopedia, Inc.

  30 E. 60th St., N.Y., NY
``Assay Methods of Antibiotics,'' D.C. Grove and            436.101(a); 
  W.A. Randall, Medical Encyclopedia, Inc., New              436.102(b).
  York, NY (1955) p. 222, p. 220.


AOAC International (Association of Official Analytical Chemists)

  481 N. Frederick Ave., Suite 500, Gaithersburg, 
  MD 20877-2417 Telephone: (301) 924-7077
Official Methods of Analysis of the Association of                331.22
  Official Analytical Chemists, 11th Ed., 1970.



[[Page 1065]]



                    Table of CFR Titles and Chapters




                     (Revised as of March 31, 1998)

                      Title 1--General Provisions

         I  Administrative Committee of the Federal Register 
                (Parts 1--49)
        II  Office of the Federal Register (Parts 50--299)
        IV  Miscellaneous Agencies (Parts 400--500)

                          Title 2--[Reserved]

                        Title 3--The President

         I  Executive Office of the President (Parts 100--199)

                           Title 4--Accounts

         I  General Accounting Office (Parts 1--99)
        II  Federal Claims Collection Standards (General 
                Accounting Office--Department of Justice) (Parts 
                100--299)

                   Title 5--Administrative Personnel

         I  Office of Personnel Management (Parts 1--1199)
        II  Merit Systems Protection Board (Parts 1200--1299)
       III  Office of Management and Budget (Parts 1300--1399)
        IV  Advisory Committee on Federal Pay (Parts 1400--1499)
         V  The International Organizations Employees Loyalty 
                Board (Parts 1500--1599)
        VI  Federal Retirement Thrift Investment Board (Parts 
                1600--1699)
       VII  Advisory Commission on Intergovernmental Relations 
                (Parts 1700--1799)
      VIII  Office of Special Counsel (Parts 1800--1899)
        IX  Appalachian Regional Commission (Parts 1900--1999)
        XI  Armed Forces Retirement Home (Part 2100)
       XIV  Federal Labor Relations Authority, General Counsel of 
                the Federal Labor Relations Authority and Federal 
                Service Impasses Panel (Parts 2400--2499)
        XV  Office of Administration, Executive Office of the 
                President (Parts 2500--2599)
       XVI  Office of Government Ethics (Parts 2600--2699)
       XXI  Department of the Treasury (Parts 3100--3199)
      XXII  Federal Deposit Insurance Corporation (Part 3201)
     XXIII  Department of Energy (Part 3301)

[[Page 1066]]

      XXIV  Federal Energy Regulatory Commission (Part 3401)
       XXV  Department of the Interior (Part 3501)
      XXVI  Department of Defense (Part 3601)
    XXVIII  Department of Justice (Part 3801)
      XXIX  Federal Communications Commission (Parts 3900--3999)
       XXX  Farm Credit System Insurance Corporation (Parts 4000--
                4099)
      XXXI  Farm Credit Administration (Parts 4100--4199)
    XXXIII  Overseas Private Investment Corporation (Part 4301)
      XXXV  Office of Personnel Management (Part 4501)
        XL  Interstate Commerce Commission (Part 5001)
       XLI  Commodity Futures Trading Commission (Part 5101)
      XLII  Department of Labor (Part 5201)
     XLIII  National Science Foundation (Part 5301)
       XLV  Department of Health and Human Services (Part 5501)
      XLVI  Postal Rate Commission (Part 5601)
     XLVII  Federal Trade Commission (Part 5701)
    XLVIII  Nuclear Regulatory Commission (Part 5801)
         L  Department of Transportation (Part 6001)
       LII  Export-Import Bank of the United States (Part 6201)
      LIII  Department of Education (Parts 6300--6399)
       LIV  Environmental Protection Agency (Part 6401)
      LVII  General Services Administration (Part 6701)
     LVIII  Board of Governors of the Federal Reserve System (Part 
                6801)
       LIX  National Aeronautics and Space Administration (Part 
                6901)
        LX  United States Postal Service (Part 7001)
       LXI  National Labor Relations Board (Part 7101)
      LXII  Equal Employment Opportunity Commission (Part 7201)
     LXIII  Inter-American Foundation (Part 7301)
       LXV  Department of Housing and Urban Development (Part 
                7501)
      LXVI  National Archives and Records Administration (Part 
                7601)
      LXIX  Tennessee Valley Authority (Part 7901)
      LXXI  Consumer Product Safety Commission (Part 8101)
     LXXIV  Federal Mine Safety and Health Review Commission (Part 
                8401)
     LXXVI  Federal Retirement Thrift Investment Board (Part 8601)
    LXXVII  Office of Management and Budget (Part 8701)

                          Title 6--[Reserved]

                         Title 7--Agriculture

            Subtitle A--Office of the Secretary of Agriculture 
                (Parts 0--26)
            Subtitle B--Regulations of the Department of 
                Agriculture
         I  Agricultural Marketing Service (Standards, 
                Inspections, Marketing Practices), Department of 
                Agriculture (Parts 27--209)

[[Page 1067]]

        II  Food and Nutrition Service, Department of Agriculture 
                (Parts 210--299)
       III  Animal and Plant Health Inspection Service, Department 
                of Agriculture (Parts 300--399)
        IV  Federal Crop Insurance Corporation, Department of 
                Agriculture (Parts 400--499)
         V  Agricultural Research Service, Department of 
                Agriculture (Parts 500--599)
        VI  Natural Resources Conservation Service, Department of 
                Agriculture (Parts 600--699)
       VII  Farm Service Agency, Department of Agriculture (Parts 
                700--799)
      VIII  Grain Inspection, Packers and Stockyards 
                Administration (Federal Grain Inspection Service), 
                Department of Agriculture (Parts 800--899)
        IX  Agricultural Marketing Service (Marketing Agreements 
                and Orders; Fruits, Vegetables, Nuts), Department 
                of Agriculture (Parts 900--999)
         X  Agricultural Marketing Service (Marketing Agreements 
                and Orders; Milk), Department of Agriculture 
                (Parts 1000--1199)
        XI  Agricultural Marketing Service (Marketing Agreements 
                and Orders; Miscellaneous Commodities), Department 
                of Agriculture (Parts 1200--1299)
      XIII  Northeast Dairy Compact Commission (Parts 1300--1399)
       XIV  Commodity Credit Corporation, Department of 
                Agriculture (Parts 1400--1499)
        XV  Foreign Agricultural Service, Department of 
                Agriculture (Parts 1500--1599)
       XVI  Rural Telephone Bank, Department of Agriculture (Parts 
                1600--1699)
      XVII  Rural Utilities Service, Department of Agriculture 
                (Parts 1700--1799)
     XVIII  Rural Housing Service, Rural Business-Cooperative 
                Service, Rural Utilities Service, and Farm Service 
                Agency, Department of Agriculture (Parts 1800--
                2099)
      XXVI  Office of Inspector General, Department of Agriculture 
                (Parts 2600--2699)
     XXVII  Office of Information Resources Management, Department 
                of Agriculture (Parts 2700--2799)
    XXVIII  Office of Operations, Department of Agriculture (Parts 
                2800--2899)
      XXIX  Office of Energy, Department of Agriculture (Parts 
                2900--2999)
       XXX  Office of the Chief Financial Officer, Department of 
                Agriculture (Parts 3000--3099)
      XXXI  Office of Environmental Quality, Department of 
                Agriculture (Parts 3100--3199)
     XXXII  [Reserved]
    XXXIII  Office of Transportation, Department of Agriculture 
                (Parts 3300--3399)
     XXXIV  Cooperative State Research, Education, and Extension 
                Service, Department of Agriculture (Parts 3400--
                3499)

[[Page 1068]]

      XXXV  Rural Housing Service, Department of Agriculture 
                (Parts 3500--3599)
     XXXVI  National Agricultural Statistics Service, Department 
                of Agriculture (Parts 3600--3699)
    XXXVII  Economic Research Service, Department of Agriculture 
                (Parts 3700--3799)
   XXXVIII  World Agricultural Outlook Board, Department of 
                Agriculture (Parts 3800--3899)
       XLI  [Reserved]
      XLII  Rural Business-Cooperative Service and Rural Utilities 
                Service, Department of Agriculture (Parts 4200--
                4299)

                    Title 8--Aliens and Nationality

         I  Immigration and Naturalization Service, Department of 
                Justice (Parts 1--499)

                 Title 9--Animals and Animal Products

         I  Animal and Plant Health Inspection Service, Department 
                of Agriculture (Parts 1--199)
        II  Grain Inspection, Packers and Stockyards 
                Administration (Packers and Stockyards Programs), 
                Department of Agriculture (Parts 200--299)
       III  Food Safety and Inspection Service, Meat and Poultry 
                Inspection, Department of Agriculture (Parts 300--
                599)

                           Title 10--Energy

         I  Nuclear Regulatory Commission (Parts 0--199)
        II  Department of Energy (Parts 200--699)
       III  Department of Energy (Parts 700--999)
         X  Department of Energy (General Provisions) (Parts 
                1000--1099)
        XI  United States Enrichment Corporation (Parts 1100--
                1199)
      XVII  Defense Nuclear Facilities Safety Board (Parts 1700--
                1799)

                      Title 11--Federal Elections

         I  Federal Election Commission (Parts 1--9099)

                      Title 12--Banks and Banking

         I  Comptroller of the Currency, Department of the 
                Treasury (Parts 1--199)
        II  Federal Reserve System (Parts 200--299)
       III  Federal Deposit Insurance Corporation (Parts 300--399)
        IV  Export-Import Bank of the United States (Parts 400--
                499)
         V  Office of Thrift Supervision, Department of the 
                Treasury (Parts 500--599)
        VI  Farm Credit Administration (Parts 600--699)
       VII  National Credit Union Administration (Parts 700--799)

[[Page 1069]]

      VIII  Federal Financing Bank (Parts 800--899)
        IX  Federal Housing Finance Board (Parts 900--999)
        XI  Federal Financial Institutions Examination Council 
                (Parts 1100--1199)
       XIV  Farm Credit System Insurance Corporation (Parts 1400--
                1499)
        XV  Thrift Depositor Protection Oversight Board (Parts 
                1500--1599)
      XVII  Office of Federal Housing Enterprise Oversight, 
                Department of Housing and Urban Development (Parts 
                1700-1799)
     XVIII  Community Development Financial Institutions Fund, 
                Department of the Treasury (Parts 1800--1899)

               Title 13--Business Credit and Assistance

         I  Small Business Administration (Parts 1--199)
       III  Economic Development Administration, Department of 
                Commerce (Parts 300--399)

                    Title 14--Aeronautics and Space

         I  Federal Aviation Administration, Department of 
                Transportation (Parts 1--199)
        II  Office of the Secretary, Department of Transportation 
                (Aviation Proceedings) (Parts 200--399)
       III  Commercial Space Transportation, Federal Aviation 
                Administration, Department of Transportation 
                (Parts 400--499)
         V  National Aeronautics and Space Administration (Parts 
                1200--1299)

                 Title 15--Commerce and Foreign Trade

            Subtitle A--Office of the Secretary of Commerce (Parts 
                0--29)
            Subtitle B--Regulations Relating to Commerce and 
                Foreign Trade
         I  Bureau of the Census, Department of Commerce (Parts 
                30--199)
        II  National Institute of Standards and Technology, 
                Department of Commerce (Parts 200--299)
       III  International Trade Administration, Department of 
                Commerce (Parts 300--399)
        IV  Foreign-Trade Zones Board, Department of Commerce 
                (Parts 400--499)
       VII  Bureau of Export Administration, Department of 
                Commerce (Parts 700--799)
      VIII  Bureau of Economic Analysis, Department of Commerce 
                (Parts 800--899)
        IX  National Oceanic and Atmospheric Administration, 
                Department of Commerce (Parts 900--999)
        XI  Technology Administration, Department of Commerce 
                (Parts 1100--1199)
      XIII  East-West Foreign Trade Board (Parts 1300--1399)
       XIV  Minority Business Development Agency (Parts 1400--
                1499)

[[Page 1070]]

            Subtitle C--Regulations Relating to Foreign Trade 
                Agreements
        XX  Office of the United States Trade Representative 
                (Parts 2000--2099)
            Subtitle D--Regulations Relating to Telecommunications 
                and Information
     XXIII  National Telecommunications and Information 
                Administration, Department of Commerce (Parts 
                2300--2399)

                    Title 16--Commercial Practices

         I  Federal Trade Commission (Parts 0--999)
        II  Consumer Product Safety Commission (Parts 1000--1799)

             Title 17--Commodity and Securities Exchanges

         I  Commodity Futures Trading Commission (Parts 1--199)
        II  Securities and Exchange Commission (Parts 200--399)
        IV  Department of the Treasury (Parts 400--499)

          Title 18--Conservation of Power and Water Resources

         I  Federal Energy Regulatory Commission, Department of 
                Energy (Parts 1--399)
       III  Delaware River Basin Commission (Parts 400--499)
        VI  Water Resources Council (Parts 700--799)
      VIII  Susquehanna River Basin Commission (Parts 800--899)
      XIII  Tennessee Valley Authority (Parts 1300--1399)

                       Title 19--Customs Duties

         I  United States Customs Service, Department of the 
                Treasury (Parts 1--199)
        II  United States International Trade Commission (Parts 
                200--299)
       III  International Trade Administration, Department of 
                Commerce (Parts 300--399)

                     Title 20--Employees' Benefits

         I  Office of Workers' Compensation Programs, Department 
                of Labor (Parts 1--199)
        II  Railroad Retirement Board (Parts 200--399)
       III  Social Security Administration (Parts 400--499)
        IV  Employees' Compensation Appeals Board, Department of 
                Labor (Parts 500--599)
         V  Employment and Training Administration, Department of 
                Labor (Parts 600--699)
        VI  Employment Standards Administration, Department of 
                Labor (Parts 700--799)
       VII  Benefits Review Board, Department of Labor (Parts 
                800--899)
      VIII  Joint Board for the Enrollment of Actuaries (Parts 
                900--999)

[[Page 1071]]

        IX  Office of the Assistant Secretary for Veterans' 
                Employment and Training, Department of Labor 
                (Parts 1000--1099)

                       Title 21--Food and Drugs

         I  Food and Drug Administration, Department of Health and 
                Human Services (Parts 1--1299)
        II  Drug Enforcement Administration, Department of Justice 
                (Parts 1300--1399)
       III  Office of National Drug Control Policy (Parts 1400--
                1499)

                      Title 22--Foreign Relations

         I  Department of State (Parts 1--199)
        II  Agency for International Development, International 
                Development Cooperation Agency (Parts 200--299)
       III  Peace Corps (Parts 300--399)
        IV  International Joint Commission, United States and 
                Canada (Parts 400--499)
         V  United States Information Agency (Parts 500--599)
        VI  United States Arms Control and Disarmament Agency 
                (Parts 600--699)
       VII  Overseas Private Investment Corporation, International 
                Development Cooperation Agency (Parts 700--799)
        IX  Foreign Service Grievance Board Regulations (Parts 
                900--999)
         X  Inter-American Foundation (Parts 1000--1099)
        XI  International Boundary and Water Commission, United 
                States and Mexico, United States Section (Parts 
                1100--1199)
       XII  United States International Development Cooperation 
                Agency (Parts 1200--1299)
      XIII  Board for International Broadcasting (Parts 1300--
                1399)
       XIV  Foreign Service Labor Relations Board; Federal Labor 
                Relations Authority; General Counsel of the 
                Federal Labor Relations Authority; and the Foreign 
                Service Impasse Disputes Panel (Parts 1400--1499)
        XV  African Development Foundation (Parts 1500--1599)
       XVI  Japan-United States Friendship Commission (Parts 
                1600--1699)
      XVII  United States Institute of Peace (Parts 1700--1799)

                          Title 23--Highways

         I  Federal Highway Administration, Department of 
                Transportation (Parts 1--999)
        II  National Highway Traffic Safety Administration and 
                Federal Highway Administration, Department of 
                Transportation (Parts 1200--1299)
       III  National Highway Traffic Safety Administration, 
                Department of Transportation (Parts 1300--1399)

[[Page 1072]]

                Title 24--Housing and Urban Development

            Subtitle A--Office of the Secretary, Department of 
                Housing and Urban Development (Parts 0--99)
            Subtitle B--Regulations Relating to Housing and Urban 
                Development
         I  Office of Assistant Secretary for Equal Opportunity, 
                Department of Housing and Urban Development (Parts 
                100--199)
        II  Office of Assistant Secretary for Housing-Federal 
                Housing Commissioner, Department of Housing and 
                Urban Development (Parts 200--299)
       III  Government National Mortgage Association, Department 
                of Housing and Urban Development (Parts 300--399)
         V  Office of Assistant Secretary for Community Planning 
                and Development, Department of Housing and Urban 
                Development (Parts 500--599)
        VI  Office of Assistant Secretary for Community Planning 
                and Development, Department of Housing and Urban 
                Development (Parts 600--699) [Reserved]
       VII  Office of the Secretary, Department of Housing and 
                Urban Development (Housing Assistance Programs and 
                Public and Indian Housing Programs) (Parts 700--
                799)
      VIII  Office of the Assistant Secretary for Housing--Federal 
                Housing Commissioner, Department of Housing and 
                Urban Development (Section 8 Housing Assistance 
                Programs and Section 202 Direct Loan Program) 
                (Parts 800--899)
        IX  Office of Assistant Secretary for Public and Indian 
                Housing, Department of Housing and Urban 
                Development (Parts 900--999)
         X  Office of Assistant Secretary for Housing--Federal 
                Housing Commissioner, Department of Housing and 
                Urban Development (Interstate Land Sales 
                Registration Program) (Parts 1700--1799)
       XII  Office of Inspector General, Department of Housing and 
                Urban Development (Parts 2000--2099)
        XX  Office of Assistant Secretary for Housing--Federal 
                Housing Commissioner, Department of Housing and 
                Urban Development (Parts 3200--3899)
       XXV  Neighborhood Reinvestment Corporation (Parts 4100--
                4199)

                           Title 25--Indians

         I  Bureau of Indian Affairs, Department of the Interior 
                (Parts 1--299)
        II  Indian Arts and Crafts Board, Department of the 
                Interior (Parts 300--399)
       III  National Indian Gaming Commission, Department of the 
                Interior (Parts 500--599)
        IV  Office of Navajo and Hopi Indian Relocation (Parts 
                700--799)
         V  Bureau of Indian Affairs, Department of the Interior, 
                and Indian Health Service, Department of Health 
                and Human Services (Part 900)
        VI  Office of the Assistant Secretary-Indian Affairs, 
                Department of the Interior (Part 1001)

[[Page 1073]]

       VII  Office of the Special Trustee for American Indians, 
                Department of the Interior (Part 1200--1299)

                      Title 26--Internal Revenue

         I  Internal Revenue Service, Department of the Treasury 
                (Parts 1--799)

           Title 27--Alcohol, Tobacco Products and Firearms

         I  Bureau of Alcohol, Tobacco and Firearms, Department of 
                the Treasury (Parts 1--299)

                   Title 28--Judicial Administration

         I  Department of Justice (Parts 0--199)
       III  Federal Prison Industries, Inc., Department of Justice 
                (Parts 300--399)
         V  Bureau of Prisons, Department of Justice (Parts 500--
                599)
        VI  Offices of Independent Counsel, Department of Justice 
                (Parts 600--699)
       VII  Office of Independent Counsel (Parts 700--799)

                            Title 29--Labor

            Subtitle A--Office of the Secretary of Labor (Parts 
                0--99)
            Subtitle B--Regulations Relating to Labor
         I  National Labor Relations Board (Parts 100--199)
        II  Office of Labor-Management Standards, Department of 
                Labor (Parts 200--299)
       III  National Railroad Adjustment Board (Parts 300--399)
        IV  Office of Labor-Management Standards, Department of 
                Labor (Parts 400--499)
         V  Wage and Hour Division, Department of Labor (Parts 
                500--899)
        IX  Construction Industry Collective Bargaining Commission 
                (Parts 900--999)
         X  National Mediation Board (Parts 1200--1299)
       XII  Federal Mediation and Conciliation Service (Parts 
                1400--1499)
       XIV  Equal Employment Opportunity Commission (Parts 1600--
                1699)
      XVII  Occupational Safety and Health Administration, 
                Department of Labor (Parts 1900--1999)
        XX  Occupational Safety and Health Review Commission 
                (Parts 2200--2499)
       XXV  Pension and Welfare Benefits Administration, 
                Department of Labor (Parts 2500--2599)
     XXVII  Federal Mine Safety and Health Review Commission 
                (Parts 2700--2799)
        XL  Pension Benefit Guaranty Corporation (Parts 4000--
                4999)

[[Page 1074]]

                      Title 30--Mineral Resources

         I  Mine Safety and Health Administration, Department of 
                Labor (Parts 1--199)
        II  Minerals Management Service, Department of the 
                Interior (Parts 200--299)
       III  Board of Surface Mining and Reclamation Appeals, 
                Department of the Interior (Parts 300--399)
        IV  Geological Survey, Department of the Interior (Parts 
                400--499)
        VI  Bureau of Mines, Department of the Interior (Parts 
                600--699)
       VII  Office of Surface Mining Reclamation and Enforcement, 
                Department of the Interior (Parts 700--999)

                 Title 31--Money and Finance: Treasury

            Subtitle A--Office of the Secretary of the Treasury 
                (Parts 0--50)
            Subtitle B--Regulations Relating to Money and Finance
         I  Monetary Offices, Department of the Treasury (Parts 
                51--199)
        II  Fiscal Service, Department of the Treasury (Parts 
                200--399)
        IV  Secret Service, Department of the Treasury (Parts 
                400--499)
         V  Office of Foreign Assets Control, Department of the 
                Treasury (Parts 500--599)
        VI  Bureau of Engraving and Printing, Department of the 
                Treasury (Parts 600--699)
       VII  Federal Law Enforcement Training Center, Department of 
                the Treasury (Parts 700--799)
      VIII  Office of International Investment, Department of the 
                Treasury (Parts 800--899)

                      Title 32--National Defense

            Subtitle A--Department of Defense
         I  Office of the Secretary of Defense (Parts 1--399)
         V  Department of the Army (Parts 400--699)
        VI  Department of the Navy (Parts 700--799)
       VII  Department of the Air Force (Parts 800--1099)
            Subtitle B--Other Regulations Relating to National 
                Defense
       XII  Defense Logistics Agency (Parts 1200--1299)
       XVI  Selective Service System (Parts 1600--1699)
       XIX  Central Intelligence Agency (Parts 1900--1999)
        XX  Information Security Oversight Office, National 
                Archives and Records Administration (Parts 2000--
                2099)
       XXI  National Security Council (Parts 2100--2199)
      XXIV  Office of Science and Technology Policy (Parts 2400--
                2499)
     XXVII  Office for Micronesian Status Negotiations (Parts 
                2700--2799)
    XXVIII  Office of the Vice President of the United States 
                (Parts 2800--2899)
      XXIX  Presidential Commission on the Assignment of Women in 
                the Armed Forces (Part 2900)

[[Page 1075]]

               Title 33--Navigation and Navigable Waters

         I  Coast Guard, Department of Transportation (Parts 1--
                199)
        II  Corps of Engineers, Department of the Army (Parts 
                200--399)
        IV  Saint Lawrence Seaway Development Corporation, 
                Department of Transportation (Parts 400--499)

                          Title 34--Education

            Subtitle A--Office of the Secretary, Department of 
                Education (Parts 1--99)
            Subtitle B--Regulations of the Offices of the 
                Department of Education
         I  Office for Civil Rights, Department of Education 
                (Parts 100--199)
        II  Office of Elementary and Secondary Education, 
                Department of Education (Parts 200--299)
       III  Office of Special Education and Rehabilitative 
                Services, Department of Education (Parts 300--399)
        IV  Office of Vocational and Adult Education, Department 
                of Education (Parts 400--499)
         V  Office of Bilingual Education and Minority Languages 
                Affairs, Department of Education (Parts 500--599)
        VI  Office of Postsecondary Education, Department of 
                Education (Parts 600--699)
       VII  Office of Educational Research and Improvement, 
                Department of Education (Parts 700--799)
        XI  National Institute for Literacy (Parts 1100-1199)
            Subtitle C--Regulations Relating to Education
       XII  National Council on Disability (Parts 1200--1299)

                        Title 35--Panama Canal

         I  Panama Canal Regulations (Parts 1--299)

             Title 36--Parks, Forests, and Public Property

         I  National Park Service, Department of the Interior 
                (Parts 1--199)
        II  Forest Service, Department of Agriculture (Parts 200--
                299)
       III  Corps of Engineers, Department of the Army (Parts 
                300--399)
        IV  American Battle Monuments Commission (Parts 400--499)
         V  Smithsonian Institution (Parts 500--599)
       VII  Library of Congress (Parts 700--799)
      VIII  Advisory Council on Historic Preservation (Parts 800--
                899)
        IX  Pennsylvania Avenue Development Corporation (Parts 
                900--999)
        XI  Architectural and Transportation Barriers Compliance 
                Board (Parts 1100--1199)
       XII  National Archives and Records Administration (Parts 
                1200--1299)
       XIV  Assassination Records Review Board (Parts 1400-1499)

[[Page 1076]]

             Title 37--Patents, Trademarks, and Copyrights

         I  Patent and Trademark Office, Department of Commerce 
                (Parts 1--199)
        II  Copyright Office, Library of Congress (Parts 200--299)
        IV  Assistant Secretary for Technology Policy, Department 
                of Commerce (Parts 400--499)
         V  Under Secretary for Technology, Department of Commerce 
                (Parts 500--599)

           Title 38--Pensions, Bonuses, and Veterans' Relief

         I  Department of Veterans Affairs (Parts 0--99)

                       Title 39--Postal Service

         I  United States Postal Service (Parts 1--999)
       III  Postal Rate Commission (Parts 3000--3099)

                  Title 40--Protection of Environment

         I  Environmental Protection Agency (Parts 1--799)
         V  Council on Environmental Quality (Parts 1500--1599)

          Title 41--Public Contracts and Property Management

            Subtitle B--Other Provisions Relating to Public 
                Contracts
        50  Public Contracts, Department of Labor (Parts 50-1--50-
                999)
        51  Committee for Purchase From People Who Are Blind or 
                Severely Disabled (Parts 51-1--51-99)
        60  Office of Federal Contract Compliance Programs, Equal 
                Employment Opportunity, Department of Labor (Parts 
                60-1--60-999)
        61  Office of the Assistant Secretary for Veterans 
                Employment and Training, Department of Labor 
                (Parts 61-1--61-999)
            Subtitle C--Federal Property Management Regulations 
                System
       101  Federal Property Management Regulations (Parts 101-1--
                101-99)
       105  General Services Administration (Parts 105-1--105-999)
       109  Department of Energy Property Management Regulations 
                (Parts 109-1--109-99)
       114  Department of the Interior (Parts 114-1--114-99)
       115  Environmental Protection Agency (Parts 115-1--115-99)
       128  Department of Justice (Parts 128-1--128-99)
            Subtitle D--Other Provisions Relating to Property 
                Management [Reserved]
            Subtitle E--Federal Information Resources Management 
                Regulations System
       201  Federal Information Resources Management Regulation 
                (Parts 201-1--201-99) [Reserved]
            Subtitle F--Federal Travel Regulation System
       301  Travel Allowances (Parts 301-1--301-99)
       302  Relocation Allowances (Parts 302-1--302-99)

[[Page 1077]]

       303  Payment of Expenses Connected with the Death of 
                Certain Employees (Parts 303-1--303-2)
       304  Payment from a Non-Federal Source for Travel Expenses 
                (Parts 304-1--304-99)

                        Title 42--Public Health

         I  Public Health Service, Department of Health and Human 
                Services (Parts 1--199)
        IV  Health Care Financing Administration, Department of 
                Health and Human Services (Parts 400--499)
         V  Office of Inspector General-Health Care, Department of 
                Health and Human Services (Parts 1000--1999)

                   Title 43--Public Lands: Interior

            Subtitle A--Office of the Secretary of the Interior 
                (Parts 1--199)
            Subtitle B--Regulations Relating to Public Lands
         I  Bureau of Reclamation, Department of the Interior 
                (Parts 200--499)
        II  Bureau of Land Management, Department of the Interior 
                (Parts 1000--9999)
       III  Utah Reclamation Mitigation and Conservation 
                Commission (Parts 10000--10005)

             Title 44--Emergency Management and Assistance

         I  Federal Emergency Management Agency (Parts 0--399)
        IV  Department of Commerce and Department of 
                Transportation (Parts 400--499)

                       Title 45--Public Welfare

            Subtitle A--Department of Health and Human Services 
                (Parts 1--199)
            Subtitle B--Regulations Relating to Public Welfare
        II  Office of Family Assistance (Assistance Programs), 
                Administration for Children and Families, 
                Department of Health and Human Services (Parts 
                200--299)
       III  Office of Child Support Enforcement (Child Support 
                Enforcement Program), Administration for Children 
                and Families, Department of Health and Human 
                Services (Parts 300--399)
        IV  Office of Refugee Resettlement, Administration for 
                Children and Families Department of Health and 
                Human Services (Parts 400--499)
         V  Foreign Claims Settlement Commission of the United 
                States, Department of Justice (Parts 500--599)
        VI  National Science Foundation (Parts 600--699)
       VII  Commission on Civil Rights (Parts 700--799)
      VIII  Office of Personnel Management (Parts 800--899)

[[Page 1078]]

         X  Office of Community Services, Administration for 
                Children and Families, Department of Health and 
                Human Services (Parts 1000--1099)
        XI  National Foundation on the Arts and the Humanities 
                (Parts 1100--1199)
       XII  Corporation for National and Community Service (Parts 
                1200--1299)
      XIII  Office of Human Development Services, Department of 
                Health and Human Services (Parts 1300--1399)
       XVI  Legal Services Corporation (Parts 1600--1699)
      XVII  National Commission on Libraries and Information 
                Science (Parts 1700--1799)
     XVIII  Harry S. Truman Scholarship Foundation (Parts 1800--
                1899)
       XXI  Commission on Fine Arts (Parts 2100--2199)
      XXII  Christopher Columbus Quincentenary Jubilee Commission 
                (Parts 2200--2299)
     XXIII  Arctic Research Commission (Part 2301)
      XXIV  James Madison Memorial Fellowship Foundation (Parts 
                2400--2499)
       XXV  Corporation for National and Community Service (Parts 
                2500--2599)

                          Title 46--Shipping

         I  Coast Guard, Department of Transportation (Parts 1--
                199)
        II  Maritime Administration, Department of Transportation 
                (Parts 200--399)
        IV  Federal Maritime Commission (Parts 500--599)

                      Title 47--Telecommunication

         I  Federal Communications Commission (Parts 0--199)
        II  Office of Science and Technology Policy and National 
                Security Council (Parts 200--299)
       III  National Telecommunications and Information 
                Administration, Department of Commerce (Parts 
                300--399)

           Title 48--Federal Acquisition Regulations System

         1  Federal Acquisition Regulation (Parts 1--99)
         2  Department of Defense (Parts 200--299)
         3  Department of Health and Human Services (Parts 300--
                399)
         4  Department of Agriculture (Parts 400--499)
         5  General Services Administration (Parts 500--599)
         6  Department of State (Parts 600--699)
         7  United States Agency for International Development 
                (Parts 700--799)
         8  Department of Veterans Affairs (Parts 800--899)
         9  Department of Energy (Parts 900--999)
        10  Department of the Treasury (Parts 1000--1099)

[[Page 1079]]

        12  Department of Transportation (Parts 1200--1299)
        13  Department of Commerce (Parts 1300--1399)
        14  Department of the Interior (Parts 1400--1499)
        15  Environmental Protection Agency (Parts 1500--1599)
        16  Office of Personnel Management Federal Employees 
                Health Benefits Acquisition Regulation (Parts 
                1600--1699)
        17  Office of Personnel Management (Parts 1700--1799)
        18  National Aeronautics and Space Administration (Parts 
                1800--1899)
        19  United States Information Agency (Parts 1900--1999)
        20  Nuclear Regulatory Commission (Parts 2000--2099)
        21  Office of Personnel Management, Federal Employees 
                Group Life Insurance Federal Acquisition 
                Regulation (Parts 2100--2199)
        23  Social Security Administration (Parts 2300--2399)
        24  Department of Housing and Urban Development (Parts 
                2400--2499)
        25  National Science Foundation (Parts 2500--2599)
        28  Department of Justice (Parts 2800--2899)
        29  Department of Labor (Parts 2900--2999)
        34  Department of Education Acquisition Regulation (Parts 
                3400--3499)
        35  Panama Canal Commission (Parts 3500--3599)
        44  Federal Emergency Management Agency (Parts 4400--4499)
        51  Department of the Army Acquisition Regulations (Parts 
                5100--5199)
        52  Department of the Navy Acquisition Regulations (Parts 
                5200--5299)
        53  Department of the Air Force Federal Acquisition 
                Regulation Supplement (Parts 5300--5399)
        54  Defense Logistics Agency, Department of Defense (Part 
                5452)
        57  African Development Foundation (Parts 5700--5799)
        61  General Services Administration Board of Contract 
                Appeals (Parts 6100--6199)
        63  Department of Transportation Board of Contract Appeals 
                (Parts 6300--6399)
        99  Cost Accounting Standards Board, Office of Federal 
                Procurement Policy, Office of Management and 
                Budget (Parts 9900--9999)

                       Title 49--Transportation

            Subtitle A--Office of the Secretary of Transportation 
                (Parts 1--99)
            Subtitle B--Other Regulations Relating to 
                Transportation
         I  Research and Special Programs Administration, 
                Department of Transportation (Parts 100--199)
        II  Federal Railroad Administration, Department of 
                Transportation (Parts 200--299)

[[Page 1080]]

       III  Federal Highway Administration, Department of 
                Transportation (Parts 300--399)
        IV  Coast Guard, Department of Transportation (Parts 400--
                499)
         V  National Highway Traffic Safety Administration, 
                Department of Transportation (Parts 500--599)
        VI  Federal Transit Administration, Department of 
                Transportation (Parts 600--699)
       VII  National Railroad Passenger Corporation (AMTRAK) 
                (Parts 700--799)
      VIII  National Transportation Safety Board (Parts 800--999)
         X  Surface Transportation Board, Department of 
                Transportation (Parts 1000--1399)

                   Title 50--Wildlife and Fisheries

         I  United States Fish and Wildlife Service, Department of 
                the Interior (Parts 1--199)
        II  National Marine Fisheries Service, National Oceanic 
                and Atmospheric Administration, Department of 
                Commerce (Parts 200--299)
       III  International Fishing and Related Activities (Parts 
                300--399)
        IV  Joint Regulations (United States Fish and Wildlife 
                Service, Department of the Interior and National 
                Marine Fisheries Service, National Oceanic and 
                Atmospheric Administration, Department of 
                Commerce); Endangered Species Committee 
                Regulations (Parts 400--499)
         V  Marine Mammal Commission (Parts 500--599)
        VI  Fishery Conservation and Management, National Oceanic 
                and Atmospheric Administration, Department of 
                Commerce (Parts 600--699)

                      CFR Index and Finding Aids

            Subject/Agency Index
            List of Agency Prepared Indexes
            Parallel Tables of Statutory Authorities and Rules
            List of CFR Titles, Chapters, Subchapters, and Parts
            Alphabetical List of Agencies Appearing in the CFR



[[Page 1081]]





           Alphabetical List of Agencies Appearing in the CFR




                     (Revised as of March 31, 1998)

                                                  CFR Title, Subtitle or 
                     Agency                               Chapter

ACTION                                            45, XII
Administrative Committee of the Federal Register  1, I
Advanced Research Projects Agency                 32, I
Advisory Commission on Intergovernmental          5, VII
     Relations
Advisory Committee on Federal Pay                 5, IV
Advisory Council on Historic Preservation         36, VIII
African Development Foundation                    22, XV
  Federal Acquisition Regulation                  48, 57
Agency for International Development, United      22, II
     States
  Federal Acquisition Regulation                  48, 7
Agricultural Marketing Service                    7, I, IX, X, XI
Agricultural Research Service                     7, V
Agriculture Department
  Agricultural Marketing Service                  7, I, IX, X, XI
  Agricultural Research Service                   7, V
  Animal and Plant Health Inspection Service      7, III; 9, I
  Chief Financial Officer, Office of              7, XXX
  Commodity Credit Corporation                    7, XIV
  Cooperative State Research, Education, and      7, XXXIV
       Extension Service
  Economic Research Service                       7, XXXVII
  Energy, Office of                               7, XXIX
  Environmental Quality, Office of                7, XXXI
  Farm Service Agency                             7, VII, XVIII
  Federal Acquisition Regulation                  48, 4
  Federal Crop Insurance Corporation              7, IV
  Food and Consumer Service                       7, II
  Food Safety and Inspection Service              9, III
  Foreign Agricultural Service                    7, XV
  Forest Service                                  36, II
  Grain Inspection, Packers and Stockyards        7, VIII; 9, II
       Administration
  Information Resources Management, Office of     7, XXVII
  Inspector General, Office of                    7, XXVI
  National Agricultural Library                   7, XLI
  National Agricultural Statistics Service        7, XXXVI
  Natural Resources Conservation Service          7, VI
  Operations, Office of                           7, XXVIII
  Rural Business-Cooperative Service              7, XVIII, XLII
  Rural Development Administration                7, XLII
  Rural Housing Service                           7, XVIII, XXXV
  Rural Telephone Bank                            7, XVI
  Rural Utilities Service                         7, XVII, XVIII, XLII
  Secretary of Agriculture, Office of             7, Subtitle A
  Transportation, Office of                       7, XXXIII
  World Agricultural Outlook Board                7, XXXVIII
Air Force Department                              32, VII
  Federal Acquisition Regulation Supplement       48, 53
Alaska Natural Gas Transportation System, Office  10, XV
     of the Federal Inspector
Alcohol, Tobacco and Firearms, Bureau of          27, I
AMTRAK                                            49, VII
American Battle Monuments Commission              36, IV
American Indians, Office of the Special Trustee   25, VII

[[Page 1082]]

Animal and Plant Health Inspection Service        7, III; 9, I
Appalachian Regional Commission                   5, IX
Architectural and Transportation Barriers         36, XI
     Compliance Board
Arctic Research Commission                        45, XXIII
Armed Forces Retirement Home                      5, XI
Arms Control and Disarmament Agency, United       22, VI
     States
Army Department                                   32, V
  Engineers, Corps of                             33, II; 36, III
  Federal Acquisition Regulation                  48, 51
Assassination Records Review Board                36, XIV
Benefits Review Board                             20, VII
Bilingual Education and Minority Languages        34, V
     Affairs, Office of
Blind or Severely Disabled, Committee for         41, 51
     Purchase From People Who Are
Board for International Broadcasting              22, XIII
Census Bureau                                     15, I
Central Intelligence Agency                       32, XIX
Chief Financial Officer, Office of                7, XXX
Child Support Enforcement, Office of              45, III
Children and Families, Administration for         45, II, III, IV, X
Christopher Columbus Quincentenary Jubilee        45, XXII
     Commission
Civil Rights, Commission on                       45, VII
Civil Rights, Office for                          34, I
Coast Guard                                       33, I; 46, I; 49, IV
Commerce Department                               44, IV
  Census Bureau                                   15, I`
  Economic Affairs, Under Secretary               37, V
  Economic Analysis, Bureau of                    15, VIII
  Economic Development Administration             13, III
  Emergency Management and Assistance             44, IV
  Export Administration, Bureau of                15, VII
  Federal Acquisition Regulation                  48, 13
  Fishery Conservation and Management             50, VI
  Foreign-Trade Zones Board                       15, IV
  International Trade Administration              15, III; 19, III
  National Institute of Standards and Technology  15, II
  National Marine Fisheries Service               50, II, IV
  National Oceanic and Atmospheric                15, IX; 50, II, III, IV, 
       Administration                             VI
  National Telecommunications and Information     15, XXIII; 47, III
       Administration
  National Weather Service                        15, IX
  Patent and Trademark Office                     37, I
  Productivity, Technology and Innovation,        37, IV
       Assistant Secretary for
  Secretary of Commerce, Office of                15, Subtitle A
  Technology, Under Secretary for                 37, V
  Technology Administration                       15, XI
  Technology Policy, Assistant Secretary for      37, IV
Commercial Space Transportation                   14, III
Commodity Credit Corporation                      7, XIV
Commodity Futures Trading Commission              5, XLI; 17, I
Community Planning and Development, Office of     24, V, VI
     Assistant Secretary for
Community Services, Office of                     45, X
Comptroller of the Currency                       12, I
Construction Industry Collective Bargaining       29, IX
     Commission
Consumer Product Safety Commission                5, LXXI; 16, II
Cooperative State Research, Education, and        7, XXXIV
     Extension Service
Copyright Office                                  37, II
Cost Accounting Standards Board                   48, 99
Council on Environmental Quality                  40, V
Customs Service, United States                    19, I
Defense Contract Audit Agency                     32, I
Defense Department                                5, XXVI; 32, Subtitle A
  Advanced Research Projects Agency               32, I
  Air Force Department                            32, VII

[[Page 1083]]

  Army Department                                 32, V; 33, II; 36, III, 
                                                  48, 51
  Defense Intelligence Agency                     32, I
  Defense Logistics Agency                        32, I, XII; 48, 54
  Engineers, Corps of                             33, II; 36, III
  Federal Acquisition Regulation                  48, 2
  National Imagery and Mapping Agency             32, I
  Navy Department                                 32, VI; 48, 52
  Secretary of Defense, Office of                 32, I
Defense Contract Audit Agency                     32, I
Defense Intelligence Agency                       32, I
Defense Logistics Agency                          32, XII; 48, 54
Defense Nuclear Facilities Safety Board           10, XVII
Delaware River Basin Commission                   18, III
Drug Enforcement Administration                   21, II
East-West Foreign Trade Board                     15, XIII
Economic Affairs, Under Secretary                 37, V
Economic Analysis, Bureau of                      15, VIII
Economic Development Administration               13, III
Economic Research Service                         7, XXXVII
Education, Department of                          5, LIII
  Bilingual Education and Minority Languages      34, V
       Affairs, Office of
  Civil Rights, Office for                        34, I
  Educational Research and Improvement, Office    34, VII
       of
  Elementary and Secondary Education, Office of   34, II
  Federal Acquisition Regulation                  48, 34
  Postsecondary Education, Office of              34, VI
  Secretary of Education, Office of               34, Subtitle A
  Special Education and Rehabilitative Services,  34, III
       Office of
  Vocational and Adult Education, Office of       34, IV
Educational Research and Improvement, Office of   34, VII
Elementary and Secondary Education, Office of     34, II
Employees' Compensation Appeals Board             20, IV
Employees Loyalty Board                           5, V
Employment and Training Administration            20, V
Employment Standards Administration               20, VI
Endangered Species Committee                      50, IV
Energy, Department of                             5, XXIII; 10, II, III, X
  Federal Acquisition Regulation                  48, 9
  Federal Energy Regulatory Commission            5, XXIV; 18, I
  Property Management Regulations                 41, 109
Energy, Office of                                 7, XXIX
Engineers, Corps of                               33, II; 36, III
Engraving and Printing, Bureau of                 31, VI
Enrichment Corporation, United States             10, XI
Environmental Protection Agency                   5, LIV; 40, I
  Federal Acquisition Regulation                  48, 15
  Property Management Regulations                 41, 115
Environmental Quality, Office of                  7, XXXI
Equal Employment Opportunity Commission           5, LXII; 29, XIV
Equal Opportunity, Office of Assistant Secretary  24, I
     for
Executive Office of the President                 3, I
  Administration, Office of                       5, XV
  Environmental Quality, Council on               40, V
  Management and Budget, Office of                25, III, LXXVII; 48, 99
  National Drug Control Policy, Office of         21, III
  National Security Council                       32, XXI; 47, 2
  Presidential Documents                          3
  Science and Technology Policy, Office of        32, XXIV; 47, II
  Trade Representative, Office of the United      15, XX
       States
Export Administration, Bureau of                  15, VII
Export-Import Bank of the United States           5, LII; 12, IV
Family Assistance, Office of                      45, II
Farm Credit Administration                        5, XXXI; 12, VI
Farm Credit System Insurance Corporation          5, XXX; 12, XIV
Farm Service Agency                               7, VII, XVIII

[[Page 1084]]

Federal Acquisition Regulation                    48, 1
Federal Aviation Administration                   14, I
  Commercial Space Transportation                 14, III
Federal Claims Collection Standards               4, II
Federal Communications Commission                 5, XXIX; 47, I
Federal Contract Compliance Programs, Office of   41, 60
Federal Crop Insurance Corporation                7, IV
Federal Deposit Insurance Corporation             5, XXII; 12, III
Federal Election Commission                       11, I
Federal Emergency Management Agency               44, I
  Federal Acquisition Regulation                  48, 44
Federal Employees Group Life Insurance Federal    48, 21
     Acquisition Regulation
Federal Employees Health Benefits Acquisition     48, 16
     Regulation
Federal Energy Regulatory Commission              5, XXIV; 18, I
Federal Financial Institutions Examination        12, XI
     Council
Federal Financing Bank                            12, VIII
Federal Highway Administration                    23, I, II; 49, III
Federal Home Loan Mortgage Corporation            1, IV
Federal Housing Enterprise Oversight Office       12, XVII
Federal Housing Finance Board                     12, IX
Federal Inspector for the Alaska Natural Gas      10, XV
     Transportation System, Office of
Federal Labor Relations Authority, and General    5, XIV; 22, XIV
     Counsel of the Federal Labor Relations 
     Authority
Federal Law Enforcement Training Center           31, VII
Federal Maritime Commission                       46, IV
Federal Mediation and Conciliation Service        29, XII
Federal Mine Safety and Health Review Commission  5, LXXIV; 29, XXVII
Federal Pay, Advisory Committee on                5, IV
Federal Prison Industries, Inc.                   28, III
Federal Procurement Policy Office                 48, 99
Federal Property Management Regulations           41, 101
Federal Property Management Regulations System    41, Subtitle C
Federal Railroad Administration                   49, II
Federal Register, Administrative Committee of     1, I
Federal Register, Office of                       1, II
Federal Reserve System                            12, II
  Board of Governors                              5, LVIII
Federal Retirement Thrift Investment Board        5, VI, LXXVI
Federal Service Impasses Panel                    5, XIV
Federal Trade Commission                          5, XLVII; 16, I
Federal Transit Administration                    49, VI
Federal Travel Regulation System                  41, Subtitle F
Fine Arts, Commission on                          45, XXI
Fiscal Service                                    31, II
Fish and Wildlife Service, United States          50, I, IV
Fishery Conservation and Management               50, VI
Food and Drug Administration                      21, I
Food and Consumer Service                         7, II
Food Safety and Inspection Service                9, III
Foreign Agricultural Service                      7, XV
Foreign Assets Control, Office of                 31, V
Foreign Claims Settlement Commission of the       45, V
     United States
Foreign Service Grievance Board                   22, IX
Foreign Service Impasse Disputes Panel            22, XIV
Foreign Service Labor Relations Board             22, XIV
Foreign-Trade Zones Board                         15, IV
Forest Service                                    36, II
General Accounting Office                         4, I, II
General Services Administration                   5, LVII
  Contract Appeals, Board of                      48, 61
  Federal Acquisition Regulation                  48, 5
  Federal Property Management Regulations System  41, 101, 105
  Federal Travel Regulation System                41, Subtitle F
  Payment From a Non-Federal Source for Travel    41, 304
       Expenses
  Payment of Expenses Connected With the Death    41, 303
     of Certain Employees
[[Page 1085]]

  Relocation Allowances                           41, 302
  Travel Allowances                               41, 301
Geological Survey                                 30, IV
Government Ethics, Office of                      5, XVI
Government National Mortgage Association          24, III
Grain Inspection, Packers and Stockyards          7, VIII; 9, II
     Administration
Great Lakes Pilotage                              46, III
Harry S. Truman Scholarship Foundation            45, XVIII
Health and Human Services, Department of          5, XLV; 45, Subtitle A
  Child Support Enforcement, Office of            45, III
  Children and Families, Administration for       45, II, III, IV, X
  Community Services, Office of                   45, X
  Family Assistance, Office of                    45, II
  Federal Acquisition Regulation                  48, 3
  Food and Drug Administration                    21, I
  Health Care Financing Administration            42, IV
  Human Development Services, Office of           45, XIII
  Indian Health Service                           25, V
  Inspector General (Health Care), Office of      42, V
  Public Health Service                           42, I
  Refugee Resettlement, Office of                 45, IV
Health Care Financing Administration              42, IV
Housing and Urban Development, Department of      5, LXV; 24, Subtitle B
  Community Planning and Development, Office of   24, V, VI
       Assistant Secretary for
  Equal Opportunity, Office of Assistant          24, I
       Secretary for
  Federal Acquisition Regulation                  48, 24
  Federal Housing Enterprise Oversight, Office    12, XVII
       of
  Government National Mortgage Association        24, III
  Housing--Federal Housing Commissioner, Office   24, II, VIII, X, XX
       of Assistant Secretary for
  Inspector General, Office of                    24, XII
  Public and Indian Housing, Office of Assistant  24, IX
       Secretary for
  Secretary, Office of                            24, Subtitle A, VII
Housing--Federal Housing Commissioner, Office of  24, II, VIII, X, XX
     Assistant Secretary for
Human Development Services, Office of             45, XIII
Immigration and Naturalization Service            8, I
Independent Counsel, Office of                    28, VII
Indian Affairs, Bureau of                         25, I, V
Indian Affairs, Office of the Assistant           25, VI
     Secretary
Indian Arts and Crafts Board                      25, II
Indian Health Service                             25, V
Information Agency, United States                 22, V
  Federal Acquisition Regulation                  48, 19
Information Resources Management, Office of       7, XXVII
Information Security Oversight Office, National   32, XX
     Archives and Records Administration
Inspector General
  Agriculture Department                          7, XXVI
  Health and Human Services Department            42, V
  Housing and Urban Development Department        24, XII
Institute of Peace, United States                 22, XVII
Inter-American Foundation                         5, LXIII; 22, X
Intergovernmental Relations, Advisory Commission  5, VII
     on
Interior Department
  American Indians, Office of the Special         25, VII
       Trustee
  Endangered Species Committee                    50, IV
  Federal Acquisition Regulation                  48, 14
  Federal Property Management Regulations System  41, 114
  Fish and Wildlife Service, United States        50, I, IV
  Geological Survey                               30, IV
  Indian Affairs, Bureau of                       25, I, V
  Indian Affairs, Office of the Assistant         25, VI
       Secretary
  Indian Arts and Crafts Board                    25, II
  Land Management, Bureau of                      43, II
  Minerals Management Service                     30, II

[[Page 1086]]

  Mines, Bureau of                                30, VI
  National Indian Gaming Commission               25, III
  National Park Service                           36, I
  Reclamation, Bureau of                          43, I
  Secretary of the Interior, Office of            43, Subtitle A
  Surface Mining and Reclamation Appeals, Board   30, III
       of
  Surface Mining Reclamation and Enforcement,     30, VII
       Office of
Internal Revenue Service                          26, I
International Boundary and Water Commission,      22, XI
     United States and Mexico, United States 
     Section
International Development, United States Agency   22, II
     for
  Federal Acquisition Regulation                  48, 7
International Development Cooperation Agency,     22, XII
     United States
  International Development, United States        22, II; 48, 7
       Agency for
  Overseas Private Investment Corporation         5, XXXIII; 22, VII
International Fishing and Related Activities      50, III
International Investment, Office of               31, VIII
International Joint Commission, United States     22, IV
     and Canada
International Organizations Employees Loyalty     5, V
     Board
International Trade Administration                15, III; 19, III
International Trade Commission, United States     19, II
Interstate Commerce Commission                    5, XL
James Madison Memorial Fellowship Foundation      45, XXIV
Japan-United States Friendship Commission         22, XVI
Joint Board for the Enrollment of Actuaries       20, VIII
Justice Department                                5, XXVIII; 28, I
  Drug Enforcement Administration                 21, II
  Federal Acquisition Regulation                  48, 28
  Federal Claims Collection Standards             4, II
  Federal Prison Industries, Inc.                 28, III
  Foreign Claims Settlement Commission of the     45, V
       United States
  Immigration and Naturalization Service          8, I
  Offices of Independent Counsel                  28, VI
  Prisons, Bureau of                              28, V
  Property Management Regulations                 41, 128
Labor Department                                  5, XLII
  Benefits Review Board                           20, VII
  Employees' Compensation Appeals Board           20, IV
  Employment and Training Administration          20, V
  Employment Standards Administration             20, VI
  Federal Acquisition Regulation                  48, 29
  Federal Contract Compliance Programs, Office    41, 60
       of
  Federal Procurement Regulations System          41, 50
  Labor-Management Standards, Office of           29, II, IV
  Mine Safety and Health Administration           30, I
  Occupational Safety and Health Administration   29, XVII
  Pension and Welfare Benefits Administration     29, XXV
  Public Contracts                                41, 50
  Secretary of Labor, Office of                   29, Subtitle A
  Veterans' Employment and Training, Office of    41, 61; 20, IX
       the Assistant Secretary for
  Wage and Hour Division                          29, V
  Workers' Compensation Programs, Office of       20, I
Labor-Management Standards, Office of             29, II, IV
Land Management, Bureau of                        43, II
Legal Services Corporation                        45, XVI
Library of Congress                               36, VII
  Copyright Office                                37, II
Management and Budget, Office of                  5, III, LXXVII; 48, 99
Marine Mammal Commission                          50, V
Maritime Administration                           46, II
Merit Systems Protection Board                    5, II
Micronesian Status Negotiations, Office for       32, XXVII
Mine Safety and Health Administration             30, I
Minerals Management Service                       30, II

[[Page 1087]]

Mines, Bureau of                                  30, VI
Minority Business Development Agency              15, XIV
Miscellaneous Agencies                            1, IV
Monetary Offices                                  31, I
National Aeronautics and Space Administration     5, LIX; 14, V
  Federal Acquisition Regulation                  48, 18
National Agricultural Library                     7, XLI
National Agricultural Statistics Service          7, XXXVI
National Archives and Records Administration      5, LXVI; 36, XII
  Information Security Oversight Office           32, XX
National Bureau of Standards                      15, II
National Capital Planning Commission              1, IV
National Commission for Employment Policy         1, IV
National Commission on Libraries and Information  45, XVII
     Science
National and Community Service, Corporation for   45, XXV
National Council on Disability                    34, XII
National Credit Union Administration              12, VII
National Drug Control Policy, Office of           21, III
National Foundation on the Arts and the           45, XI
     Humanities
National Highway Traffic Safety Administration    23, II, III; 49, V
National Imagery and Mapping Agency               32, I
National Indian Gaming Commission                 25, III
National Institute for Literacy                   34, XI
National Institute of Standards and Technology    15, II
National Labor Relations Board                    5, LXI; 29, I
National Marine Fisheries Service                 50, II, IV
National Mediation Board                          29, X
National Oceanic and Atmospheric Administration   15, IX; 50, II, III, IV, 
                                                  VI
National Park Service                             36, I
National Railroad Adjustment Board                29, III
National Railroad Passenger Corporation (AMTRAK)  49, VII
National Science Foundation                       5, XLIII; 45, VI
  Federal Acquisition Regulation                  48, 25
National Security Council                         32, XXI
National Security Council and Office of Science   47, II
     and Technology Policy
National Telecommunications and Information       15, XXIII; 47, III
     Administration
National Transportation Safety Board              49, VIII
National Weather Service                          15, IX
Natural Resources Conservation Service            7, VI
Navajo and Hopi Indian Relocation, Office of      25, IV
Navy Department                                   32, VI
  Federal Acquisition Regulation                  48, 52
Neighborhood Reinvestment Corporation             24, XXV
Northeast Dairy Compact Commission                7, XIII
Nuclear Regulatory Commission                     5, XLVIII; 10, I
  Federal Acquisition Regulation                  48, 20
Occupational Safety and Health Administration     29, XVII
Occupational Safety and Health Review Commission  29, XX
Offices of Independent Counsel                    28, VI
Operations Office                                 7, XXVIII
Overseas Private Investment Corporation           5, XXXIII; 22, VII
Panama Canal Commission                           48, 35
Panama Canal Regulations                          35, I
Patent and Trademark Office                       37, I
Payment From a Non-Federal Source for Travel      41, 304
     Expenses
Payment of Expenses Connected With the Death of   41, 303
     Certain Employees
Peace Corps                                       22, III
Pennsylvania Avenue Development Corporation       36, IX
Pension and Welfare Benefits Administration       29, XXV
Pension Benefit Guaranty Corporation              29, XL
Personnel Management, Office of                   5, I, XXXV; 45, VIII
  Federal Acquisition Regulation                  48, 17
  Federal Employees Group Life Insurance Federal  48, 21
     Acquisition Regulation
[[Page 1088]]

  Federal Employees Health Benefits Acquisition   48, 16
       Regulation
Postal Rate Commission                            5, XLVI; 39, III
Postal Service, United States                     5, LX; 39, I
Postsecondary Education, Office of                34, VI
President's Commission on White House             1, IV
     Fellowships
Presidential Commission on the Assignment of      32, XXIX
     Women in the Armed Forces
Presidential Documents                            3
Prisons, Bureau of                                28, V
Productivity, Technology and Innovation,          37, IV
     Assistant Secretary
Public Contracts, Department of Labor             41, 50
Public and Indian Housing, Office of Assistant    24, IX
     Secretary for
Public Health Service                             42, I
Railroad Retirement Board                         20, II
Reclamation, Bureau of                            43, I
Refugee Resettlement, Office of                   45, IV
Regional Action Planning Commissions              13, V
Relocation Allowances                             41, 302
Research and Special Programs Administration      49, I
Rural Business-Cooperative Service                7, XVIII, XLII
Rural Development Administration                  7, XLII
Rural Housing Service                             7, XVIII, XXXV
Rural Telephone Bank                              7, XVI
Rural Utilities Service                           7, XVII, XVIII, XLII
Saint Lawrence Seaway Development Corporation     33, IV
Science and Technology Policy, Office of          32, XXIV
Science and Technology Policy, Office of, and     47, II
     National Security Council
Secret Service                                    31, IV
Securities and Exchange Commission                17, II
Selective Service System                          32, XVI
Small Business Administration                     13, I
Smithsonian Institution                           36, V
Social Security Administration                    20, III; 48, 23
Soldiers' and Airmen's Home, United States        5, XI
Special Counsel, Office of                        5, VIII
Special Education and Rehabilitative Services,    34, III
     Office of
State Department                                  22, I
  Federal Acquisition Regulation                  48, 6
Surface Mining and Reclamation Appeals, Board of  30, III
Surface Mining Reclamation and Enforcement,       30, VII
     Office of
Surface Transportation Board                      49, X
Susquehanna River Basin Commission                18, VIII
Technology Administration                         15, XI
Technology Policy, Assistant Secretary for        37, IV
Technology, Under Secretary for                   37, V
Tennessee Valley Authority                        5, LXIX; 18, XIII
Thrift Depositor Protection Oversight Board       12, XV
Thrift Supervision Office, Department of the      12, V
     Treasury
Trade Representative, United States, Office of    15, XX
Transportation, Department of                     5, L
  Coast Guard                                     33, I; 46, I; 49, IV
  Commercial Space Transportation                 14, III
  Contract Appeals, Board of                      48, 63
  Emergency Management and Assistance             44, IV
  Federal Acquisition Regulation                  48, 12
  Federal Aviation Administration                 14, I
  Federal Highway Administration                  23, I, II; 49, III
  Federal Railroad Administration                 49, II
  Federal Transit Administration                  49, VI
  Maritime Administration                         46, II
  National Highway Traffic Safety Administration  23, II, III; 49, V
  Research and Special Programs Administration    49, I
  Saint Lawrence Seaway Development Corporation   33, IV
  Secretary of Transportation, Office of          14, II; 49, Subtitle A
  Surface Transportation Board                    49, X

[[Page 1089]]

Transportation, Office of                         7, XXXIII
Travel Allowances                                 41, 301
Treasury Department                               5, XXI; 17, IV
  Alcohol, Tobacco and Firearms, Bureau of        27, I
  Community Development Financial Institutions    12, XVIII
       Fund
  Comptroller of the Currency                     12, I
  Customs Service, United States                  19, I
  Engraving and Printing, Bureau of               31, VI
  Federal Acquisition Regulation                  48, 10
  Federal Law Enforcement Training Center         31, VII
  Fiscal Service                                  31, II
  Foreign Assets Control, Office of               31, V
  Internal Revenue Service                        26, I
  International Investment, Office of             31, VIII
  Monetary Offices                                31, I
  Secret Service                                  31, IV
  Secretary of the Treasury, Office of            31, Subtitle A
  Thrift Supervision, Office of                   12, V
Truman, Harry S. Scholarship Foundation           45, XVIII
United States and Canada, International Joint     22, IV
     Commission
United States and Mexico, International Boundary  22, XI
     and Water Commission, United States Section
United States Enrichment Corporation              10, XI
Utah Reclamation Mitigation and Conservation      43, III
     Commission
Veterans Affairs Department                       38, I
  Federal Acquisition Regulation                  48, 8
Veterans' Employment and Training, Office of the  41, 61; 20, IX
     Assistant Secretary for
Vice President of the United States, Office of    32, XXVIII
Vocational and Adult Education, Office of         34, IV
Wage and Hour Division                            29, V
Water Resources Council                           18, VI
Workers' Compensation Programs, Office of         20, I
World Agricultural Outlook Board                  7, XXXVIII

[[Page 1091]]



List of CFR Sections Affected



All changes in this volume of the Code of Federal Regulations which were 
made by documents published in the Federal Register since January 1, 
1986, are enumerated in the following list. Entries indicate the nature 
of the changes effected. Page numbers refer to Federal Register pages. 
The user should consult the entries for chapters and parts as well as 
sections for revisions.
For the period before January 1, 1986, see the ``List of Sections 
Affected, 1949-1963, 1964-1972, and 1973-1985,'' published in seven 
separate volumes.

                                  1986

21 CFR
                                                                   51 FR
                                                                    Page
Chapter I
Mandatory compliance date 1-1-89...................................34085
310  Authority citation revised....................................24479
310.305  Added.....................................................24479
    Technical correction...........................................28810
310.534  Added; eff. 7-18-87.......................................26114
314.80  (a) amended................................................24481
    Technical correction...........................................28810
330  Authority citation revised....................................16265
330.1  (c) redesignated as (c)(1); (c)(2) added....................16266
    Technical correction...........................................18580
330.2  Authority citation removed..................................16265
330.10  Authority citation removed.................................16265
330.13  Authority citation removed.................................16265
331  Authority citation revised....................................16266
331.11  (k)(1) amended.............................................27763
    Technical correction...........................................32212
331.30  (b) revised................................................16266
    Technical correction....................................18580, 32212
    (g) added......................................................27763
332  Authority citation revised....................................16266
332.30  (a) revised................................................16266
    Technical correction....................................18580, 32212
    (c) added......................................................27763
341  Added; eff. 10-2-87...........................................35339
344  Added; eff. 8-10-87...........................................28660
357  Authority citation revised....................................16267
357.101--357.180 (Subpart B)  Added (effective date pending in 
        part)......................................................27759
357.250  (b) revised...............................................16267
    Technical correction...........................................18580
369  Authority citation revised....................................27759
    Correctly designated...........................................31763
369.20  Amended....................................................27760
    Amended; eff. 10-2-87..........................................35340
430.4  (a)(55) added...............................................11571
430.5  (a)(87) and (b)(89) added...................................11571
    (a)(88) and (b)(90) added......................................20263
430.6  (b)(89) added...............................................11572
    (b)(90) added..................................................20263
433  Authority citation revised....................................25524
433.1  Revised; authority citation removed.........................25524
    (a) corrected..................................................30478
    Technical correction...........................................33897
433.2  (b) revised; authority citation removed.....................25524
    Technical correction...........................................33897
436.200  (i) added.................................................11572
436.201  (c)(3)(iii) redesignated as (c)(3)(iv) and revised; 
        (b)(3)(iii) and new (c)(3)(iii) added......................22071
    (b)(1) amended.................................................27532
436.215  (a), (b), and (d) revised; (c)(7) added...................22072

[[Page 1092]]

    (b) table amended; (c)(8) added................................37721
436.216  Added.....................................................11572
436.360  (c)(3) corrected...........................................1367
436.545  Added.....................................................37721
440  Authority citation revised....................................27532
440.13a  (a)(1)(vi) amended........................................27532
441  Added.........................................................11573
441.20a  (a)(3)(i) and (b)(1) introductory text, (i)(a), (d), (ii) 
        heading and (a) corrected; (b) (2) through (6) correctly 
        added......................................................16517
441.220  (b)(1)(ii)(a) corrected...................................22275
442  Authority citation revised; section authority citations 
        removed....................................................20263
442.12  Added......................................................36688
442.14a  (b)(1) revised............................................27532
442.16a  (b)(4) corrected...........................................2478
442.53a  Added.....................................................20263
442.212  Redesignated as 442.212a; new 442.212 added...............36688
442.212a  Redesignated from 442.212................................36688
442.212b  Added....................................................36688
442.216  (a)(3)(i)(b) and (b)(1)(ii)(a) corrected...................2478
442.253  Added.....................................................20264
448.510f  Added....................................................35212
449.150d  Added.....................................................4617
452.32a  Added.....................................................35215
452.110d  Added....................................................37723
452.232  Redesignated as 452.232a; new 452.232 added...............35216
452.232a  Redesignated from 452.232................................35216
452.232b  Added....................................................35216
452.910 (Subpart J)  Added.........................................35213
455.185  Redesignated as 455.185a; new 455.185 added...............22072
455.185a  Redesignated from 455.185................................22072
455.185b  Added....................................................22072

                                  1987

21 CFR
                                                                   52 FR
                                                                    Page
Chapter I
310  Authority citation revised.............................30055, 37936
310.201  (a)(6) removed; eff. 5-2-88...............................15892
    (a)(14) removed; eff. 8-12-88..................................30055
    Technical correction...........................................34047
310.305  (b)(4) revised; (c)(1) redesignated as (c)(1)(i); 
        (c)(1)(ii) added...........................................37936
312  Revised; eff. 6-17-87..........................................8831
312.7  Heading and (d) revised; OMB number.........................19476
    Technical correction...........................................23628
312.10  OMB number.................................................23031
312.23  OMB number.................................................23031
312.30  OMB number.................................................23031
312.31  OMB number.................................................23031
312.32  OMB number.................................................23031
312.33  OMB number.................................................23031
312.34  Text added.................................................19476
    Technical correction...........................................23628
312.35  Added......................................................19477
    Technical correction...........................................23628
312.36  OMB number.................................................23031
312.38  OMB number.................................................23031
312.41  OMB number.................................................23031
312.42  (b)(3) added...............................................19477
    Technical correction...........................................23628
312.44  OMB number.................................................23031
312.45  OMB number.................................................23031
312.47  OMB number.................................................23031
312.53  OMB number.................................................23031
312.55  OMB number.................................................23031
312.56  OMB number.................................................23031
312.57  OMB number.................................................23031
312.59  OMB number.................................................23031
312.62  OMB number.................................................23031
312.64  OMB number.................................................23031
312.66  OMB number.................................................23031
312.70  OMB number.................................................23031
312.110  OMB number................................................23031
312.120  OMB number................................................23031
312.140  OMB number................................................23031
312.160  OMB number................................................23031
314.50  (d)(5) (x) and (xi) added; eff. 6-17-87.....................8847
314.80  (a), (c) introductory text, (1), and (2)(ii)(b), (d)(2), 
        and (f) (1), (3), and (4) amended; (c)(2)(iii) and (e) 
        revised....................................................37936
331  Petition denied...............................................33576
331.30  (b) corrected...............................................7830
332.30  (a) corrected...............................................7830
333  Added; eff. 12-12-88..........................................47322
333.120  (a)(11) corrected.........................................48792
336  Added; eff. 5-2-88............................................15892
341  Authority citation revised....................................30055
    Technical correction...........................................34047
341.3  (b) and (c) added; eff. 8-12-88.............................30055

[[Page 1093]]

341.14  Added; eff. 8-12-88........................................30055
341.74  Added; eff. 8-12-88........................................30055
    (c)(2) corrected...............................................35610
341.76  OMB number; eff. 10-2-87....................................7126
    OMB number......................................................7126
    (b) corrected...................................................7830
    Technical correction...........................................12521
341.90  (b) and (c) added; eff. 8-12-88............................30057
344.50  (b) corrected...............................................7830
357.150  (b) corrected..............................................7831
357.152  OMB number.................................................2515
357.250  (b) corrected..............................................7830
369  Authority citation revised.............................30057, 47324
369.6  Removed; eff. 12-12-88......................................47324
369.20  Amended; eff. 5-2-88.......................................15893
    Amended; eff. 8-12-88..........................................30057
    Technical correction...........................................34047
    Amended; eff. 12-12-88.........................................47324
369.21  Amended; eff. 5-2-88.......................................15893
    Amended; eff. 8-12-88..........................................30057
    Technical correction...........................................34047
    Amended; eff. 12-12-88.........................................47324
430.4  (a)(56) added................................................4611
    (a)(57) added..................................................42287
430.5  (a)(89) and (91) added.......................................4611
    (a)(90) and (b)(92) added......................................42287
    (a)(91) and (b)(93) added......................................42431
430.6  (b)(91) added................................................4611
    (b)(92) added..................................................42288
    (b)(93) added..................................................42431
436.20  (d)(9) added................................................4611
436.105  (b) table amended..........................................4617
436.212  (e)(4) added; (f) revised..................................4617
436.215  (b) table amended; (c)(9) added...........................42432
    Correctly designated...........................................43966
    (c)(9)(i) corrected............................................45528
436.217  Added.....................................................42432
436.361  Added......................................................4611
    436.361  (c)(4) corrected.......................................8550
436.542  (c) amended...............................................35912
440.9a  (b)(1)(i) revised; (b)(1)(ii)(d) added.....................42288
    (b)(1)(ii)(d)(4) formula corrected.............................45281
440.209  Redesignated as 440.209a; new 440.209 added...............42288
440.209a  Redesignated from 440.209................................42288
440.209b  Added....................................................42288
442.19  Added......................................................42432
    Heading corrected..............................................43966
    (b)(1)(i) (A) and (D) and (iv)(A) formula corrected............45528
442.27  (a)(1) amended.............................................35912
442.53a  (b)(1)(iv)(a) revised.....................................35912
442.55  Added......................................................44860
442.119  Added.....................................................42433
    (b)(1)(iii) formula corrected..................................45528
442.127a  (a)(1) amended...........................................20710
442.255  Redesignated as 442.255a; new 442.255 added...............44860
442.255a  Redesignated from 442.255................................44860
442.255b  Added....................................................44860
444.542i  Removed..................................................35912
449.150d  (a)(3)(i)(a) corrected....................................7741
455.4a  Added.......................................................4614
455.82a  Added.....................................................42290
    (b)(1)(iii)(D) and (iv) corrected..............................45281
455.204  Added......................................................4615
    (b)(1) and (b)(1)(ii)(c) corrected..............................8550

                                  1988

21 CFR
                                                                   53 FR
                                                                    Page
Chapter I
Uniform compliance date 1-1-91.....................................44861
310.540  Added.....................................................31271
312.23  (e) added...................................................1918
312.30  (d) introductory text amended...............................1918
312.31  (b) introductory text amended...............................1918
312.80--312.88 (Subpart E)  Added; interim.........................41523
    Technical correction...........................................44144
312.110--312.145 (Subpart E)  Redesignated as Subpart F; interim 
                                                                   41523
    (Subpart F)  Redesignated from Subpart E; interim..............41523
312.160 (Subpart F)  Redesignated as Subpart G; interim............41523
    (Subpart G)  Redesignated from Subpart F; interim..............41523
314.125  (c) added; interim........................................41524
    Technical correction...........................................44144
314.420  (c) amended...............................................33122
333.110  (e) redesignated as (f); new (e) added....................18838
333.120  (a)(10) revised...........................................18838

[[Page 1094]]

336.50  (d) (1), (2), (3), and (4) revised.........................35809
340  Added; eff. 3-1-89.............................................6105
    Authority citation corrected...................................11731
341.74  (d)(1) (i), (ii), and (iii) revised........................35809
341.76  (d)(1), (2)(i)(a), and (ii) revised........................35810
349  Added..........................................................7090
    Addition effective date corrected to 3-6-89....................13217
357  Authority citation revised....................................35810
357.150  (d)(1) revised............................................35810
369.20  Amended.....................................................7093
    Amendment effective date corrected to 3-6-89...................13217
430.4  (a)(58) added...............................................13400
    (a)(59) added..................................................24257
    (a)(59) corrected..............................................26712
430.5  (a)(92) and (b)(94) added...................................13400
    (a) (93) and (95) added........................................24257
430.6  (b)(94) added...............................................13401
    (b)(95) added..................................................24257
436.20  (d)(10) added..............................................13401
436.31  (b)(16) added..............................................13401
436.32  (j) added..................................................13401
436.106  (a) table and (b) table amended...........................32607
    Correctly designated...........................................39839
436.215  (b) table amended; (c)(10) added..........................24257
    (c)(10) correctly designated; (c)(10)(iii) corrected...........26712
436.362  Added......................................................1919
436.363  Added.....................................................13401
    (c)(3) corrected...............................................19368
436.364  Added.....................................................13401
442.15  Added......................................................24257
    (a)(3)(i) and (b)(1) introductory text corrected...............26712
442.22a  Added.....................................................13402
    (b)(4)(i) corrected............................................19368
442.115  Added.....................................................24259
442.115a  Added....................................................24259
442.115b  Added....................................................24259
442.222  Added.....................................................13403
    (b)(1)(iv)(A) corrected........................................19369
444  Correctly designated..........................................16615
444.42a  (a)(2) removed; (a) (3) and (4) redesignated as (a) (2) 
        and (3) and revised; (b)(1)(i)(d) and (ii) revised; 
        (b)(1)(ii) undesignated text removed.......................12660
444.320c  Added....................................................40725
444.542b  Heading, (a)(1) introductory text, and (2) revised.......18838
444.942a  Heading, (a)(1) introductory text, (3) and (4)(i) 
        revised....................................................12658
    (a)(3) correctly revised................................12658, 31837
    Technical correction...........................................36391
446.60  (a)(1)(i) and (v), (3)(i) and (b) (1) and (5) revised; 
        (a)(1) (viii) and (ix) and (b) (8) and (9) added...........32607
    (b)(1) introductory text and (i)(b) heading corrected..........39839
446.160a  (a)(3)(i)(a) and (b)(1) revised..........................32609
446.160b  (a)(3)(i)(a) and (b)(1) revised..........................32609
446.160c  (a)(3)(i)(a) and (b)(1) revised..........................32609
446.260  (a)(3)(i)(a) and (b)(1) revised...........................32609
450.24  (a)(1) (iii) through (vi) and (b) (3) through (6) 
        redesignated as (a)(1) (iv) through (vii) and (b) (4) 
        through (7); new (a)(1)(iii) and (b)(3) added; (a)(3)(i) 
        revised....................................................37292
450.224  Redesignated as 450.224a; new 450.224 added...............37292
450.224a  Redesignated from 450.224................................37292
450.224b  Added....................................................37292
452.15  (a)(1)(ii) and (b)(2) added; (a)(3)(i) and (b) (4) and (6) 
        revised.....................................................1920
452.510e  Added....................................................12415
    (a)(1) and (b) corrected.......................................16837

                                  1989

21 CFR
                                                                   54 FR
                                                                    Page
Chapter I
300  Authority citation added; sectional authority citations 
        removed....................................................39636
310  Authority citation revised; sectional authority citations 
        removed....................................................39636
310.501  Revised...................................................22587
310.501a  Removed..................................................22588
310.516  (f) and (g) revised........................................1163
310.527  Added.....................................................28777
    Technical correction...........................................31405
    Clarification..................................................50364
310.528  Added.....................................................28786

[[Page 1095]]

    Clarification..................................................50364
312  Authority citation revised....................................39636
314  Authority citation revised....................................39636
320  Authority citation revised; sectional and subpart authority 
        citations removed..........................................39636
329  Authority citation revised....................................39636
329.1  Authority citation removed..................................39636
330  Authority citation revised....................................39636
331  Authority citation revised....................................39636
332  Authority citation revised....................................39636
333  Authority citation revised....................................39636
336  Authority citation revised....................................39636
338  Added..........................................................6826
    Authority citation revised.....................................39636
340  Authority citation revised....................................39636
341  Policy statement..............................................36762
    Authority citation revised.....................................39636
341.3  (d) added....................................................8509
    Technical correction...........................................11866
341.18  Added.......................................................8509
    Technical correction...........................................11866
341.78  Added.......................................................8509
    Technical correction...........................................11866
341.90  (d) added...................................................8509
    Technical correction...........................................11866
344  Authority citation revised....................................39636
349  Authority citation revised....................................39636
357  Authority citation revised....................................39636
357.210  Revised....................................................8321
357.250  (d)(2) and (3) revised.....................................8321
357.280  Revised....................................................8321
361  Authority citation revised....................................39636
369  Authority citation revised....................................39636
429  Authority citation revised....................................39636
430  Authority citation revised; sectional authority citations 
        removed....................................................39637
430.4  (a)(60) added...............................................20784
430.5  (a)(94) and (b)(96) added...................................20784
    (a)(95) and (b)(97) added......................................38224
430.6  (b)(96) added...............................................20784
    (b)(97) added..................................................38224
    (b)(34) amended................................................47204
431  Authority citation revised; sectional authority citations 
        removed....................................................39637
432  Authority citation revised....................................39637
432.1  Authority citation removed..................................39637
433  Authority citation revised....................................39637
436  Authority citation revised; sectional authority citations 
        removed....................................................39637
436.215  (b) table footnote and (c)(10)(iii) amended...............47204
    (b) table amended; (c)(11) added...............................48859
436.216  (c) (1) and (2) revised; (c)(5) added.....................47351
436.357  Heading revised; (c)(2) amended...........................20785
436.365  Added.....................................................38375
    (e) corrected..................................................42886
436.366  Added.....................................................20383
    (g)(3) corrected...............................................25849
436.367  Added.....................................................48860
    (c) corrected..................................................51816
436.542  (b)(2) amended............................................41824
440  Authority citation revised; sectional authority citations 
        removed....................................................39637
440.9a  (a)(1) (vii) and (viii) revised............................47204
440.103b  (b)(4) revised...........................................47351
440.209b  (b)(1)(iii) (B) and (C) amended..........................47205
441  Authority citation revised....................................39637
442  Authority citation revised....................................39637
442.15  (a)(1), (b)(1)(ii) (A), (C) and (iii)(E) amended...........47205
442.16  Added......................................................40652
442.18  Added......................................................40654
    Heading corrected..............................................50686
442.19  (b)(1)(iv) revised.........................................47351
442.20a  (a)(1)(vi) amended........................................41824
    Corrected......................................................43384
442.28  Added......................................................48860
442.58a  Added.....................................................20785
442.104b  (a)(1) amended...........................................41824

[[Page 1096]]

442.106b  (a)(1) amended...........................................47352
442.119  (b)(1)(iii) revised.......................................47352
    (b)(1)(iii) corrected..........................................50472
442.128  Added.....................................................48860
442.212b  (a)(1) amended...........................................47352
442.216  Redesignated as 442.216a; new 442.216.....................40652
442.216a  Redesignated from 442.216................................40652
442.216b  Added....................................................40652
442.218  Redesignated as 442.218a; new 442.218 added...............40654
442.218a  Redesignated from 442.218................................40654
442.218b  Added....................................................40654
442.258  Added.....................................................20786
444  Authority citation revised; sectional authority citations 
        removed....................................................39637
444.380c  Added....................................................13879
446  Authority citation revised; sectional authority citations 
        removed....................................................39637
446.60  (b)(1)(iii)(e) and (iv) amended............................47205
446.115c  Removed..................................................47352
446.116b  Removed..................................................47352
446.116d  Removed..................................................47352
446.165c  Removed..................................................47352
446.165e  Removed..................................................47352
446.180a  Removed..................................................47352
446.180b  Removed..................................................47352
446.181a  Removed..................................................47352
446.181b  Removed..................................................47352
446.260  (b)(1)(ii)(a) amended.....................................47205
448  Authority citation revised; sectional authority citations 
        removed....................................................39637
448.330  Added.....................................................38376
449  Authority citation revised; sectional authorities removed.....39637
450  Authority citation revised; sectional authorities removed.....39637
452  Authority citation revised; sectional authority citations 
        removed....................................................39637
452.510b  (a)(2) revised...........................................47352
    (a)(2) corrected...............................................50472
453  Authority citation revised; sectional authority citations 
        removed....................................................39637
453.120  (a)(1) amended............................................41824
    (a)(1) corrected...............................................43384
453.222  (a)(1) and (b)(1) revised.................................43289
453.522  Redesignated as 453.522a; new 453.522 added...............38224
453.522a  Redesignated from 453.522................................38224
453.522b  Added....................................................38224
453.522c  Added....................................................40655
455  Authority citation revised; sectional authority citations 
        removed....................................................39637
455.4  Added.......................................................40385
455.82a  (b)(1)(iii)(B) amended....................................47205
455.204  Redesignated as 455.204a; new 455.204 added...............40385
    (a)(1) and (b)(1)(iv)(a)(3) amended; (a)(3)(i)(a) revised......41824
455.204a  Redesignated from 455.204................................40385
455.204b  Added....................................................40385
455.270  Added.....................................................38375
455.285  Added.....................................................20384
455.285b  Added....................................................20384
    (a)(3)(ii)(B)(1) corrected.....................................22838
460  Authority citation revised....................................39637
460.42  Authority citation removed.................................39637

                                  1990

21 CFR
                                                                   55 FR
                                                                    Page
Chapter I
300  Authority citation correctly added............................14968
310.6  (e) amended.................................................11578
310.103  (b) revised...............................................11578
310.201  (a)(23)(v)(b) removed; eff. 8-5-91........................31779
310.305  (c)(2), (d)(3)(ii), and (4) amended.......................11578
310.500  (a)(1) introductory text and (vii) introductory text, 
        (d), (f), (h) introductory text, (h)(3)(iv) amended........11578
310.502  (a)(2) and (b)(4) amended.................................11578
310.503  (a), (d)(3), (e), (f)(3), (5)(i) and (ii) amended.........11578
310.504  (f) amended...............................................11578
310.506  (c) revised...............................................11578
310.507  (c) revised...............................................11578
310.508  (c) revised...............................................11578
310.509  (a), (b), (e), and (i) amended............................11578

[[Page 1097]]

310.510  (c) revised...............................................11579
310.513  (c)(3), (d), and (e) amended..............................11579
310.515  (g) amended...............................................11579
    Revised........................................................18723
310.519  (c) revised...............................................11579
310.525  (c) revised...............................................11579
310.526  (c) revised...............................................11579
310.529  (c) revised...............................................11579
310.532  Added; eff. 8-27-90........................................6930
310.533  (c) revised...............................................11579
310.534  (c) revised...............................................11579
310.541  Added.....................................................19858
310.542  Added.....................................................19858
310.545  Added; eff. 5-7-91........................................46919
    (a)(7) corrected...............................................49973
312  Technical correction...................................50279, 51799
312.32  (c)(1) redesignated as (c)(1)(i); new (c)(1)(i), (2), and 
        (3) amended................................................11579
312.36  Amended....................................................11579
312.44  (d) amended................................................11579
312.47  (b)(1)(iv) and (v) amended.................................11580
312.48  (b) and (c)(1) amended.....................................11580
312.57  (c) added; interim.........................................47038
312.140  (a), (b), and (c) amended.................................11580
312.145  (b) revised...............................................11580
314  Technical correction..........................................37322
                                                            50279, 51799
314.50  (f)(1) revised.............................................11580
314.80  (f)(3) and (4) amended.....................................11580
314.81  (a) amended................................................11580
314.106  (a) amended...............................................11580
314.110  (c) amended...............................................11580
314.120  (c) amended...............................................11580
314.125  (b)(17) added; interim....................................47038
314.126  (c) amended...............................................11580
314.150  (b)(1) amended............................................11580
    (b)(9) added...................................................47038
314.200  (a) introductory text, (3), (f), (g)(2), and (3) amended 
                                                                   11580
314.300  (b)(7), (8)(ii) and (iii) amended.........................11580
314.420  (a)(1) through (5) revised................................28380
314.430  (b), (e)(2)(ii)(a) and (b), (f)(5) and (6) amended........11580
314.440  (a) introductory text, (1), (2), (3) amended; (b) 
        introductory text revised..................................11581
314.445  (b) revised...............................................11581
320  Technical correction...................................50279, 51799
320.30  (c) amended................................................11581
320.31  Heading revised; (a) introductory text, (c), (d), and (f) 
        amended....................................................11581
    (a) introductory text, (c) and (d) revised; (e) and (f) 
removed; interim...................................................47038
320.32  Added; interim.............................................47038
    (c) corrected..................................................50279
    (a) and (c) corrected; interim.................................52991
320.63  Added; interim.............................................47038
329.20  (d) amended................................................11581
330.1  (f) amended.................................................11581
330.10  (a)(4)(ii) amended.........................................11581
330.11  Amended....................................................11581
330.13  (d)(2)(i) amended..........................................11581
331.11  (a)(1) and (4) revised; eff. 5-13-91.......................19859
331.22  Amended....................................................11581
331.30  (g) amended................................................11581
331.31  Redesignated as 331.80 and new (a)(3) and (4) added; eff. 
        5-13-91....................................................19859
331.80  Redesignated from 331.31 and new (a)(3) and (4) added; 
        eff. 5-13-91...............................................19859
333.120  (b)(3) added; eff. 3-15-91.................................9722
    (a)(7) revised; eff. 10-3-91...................................40381
    (a)(5)(iii) redesignated as (a)(5)(iv); new (a)(5)(iii), 
(b)(4), (5) and (6) added; eff. 12-5-91............................50172
338  Authority citation revised.....................................9079
341.3  (b) and (c) revised.........................................40382
341.74  (f) added..................................................27808
    (d)(2)(iii) revised............................................40383
346  Added; eff. 8-5-91............................................31779
357.801--357.850 (Subpart I)  Added; eff. 5-13-91..................19865
358  Added.........................................................33255
    Regulation at 55 FR 33255; effective date corrected to 8-14-91
                                                                   37403
358.150  (c)(1)(ii) and (d)(3) corrected...........................37403
358.501--358.550 (Subpart F)  Added; eff. 8-14-91..................33261
361.1  (b)(2), (c)(1), (3), (4), (d)(8) and (e) amended............11582
369.20  Amended; eff. 8-5-91.......................................31783
369.21  Amended....................................................11582
429.40  (a) amended................................................11582

[[Page 1098]]

430  Authority citation corrected..................................11110
430.4  (a)(61) added................................................2641
    (a)(62) added...................................................6634
    (a)(63) added..................................................14240
430.5  (a)(96) and (b)(98) added....................................2641
    (a)(97) and (b)(99) added.......................................6634
    (a)(98) and (b)(100) added.....................................14240
430.6  (b)(98) added................................................2641
    (b)(99) added...................................................6634
    (b)(100) added.................................................14240
431.1  (a) amended.................................................11582
431.50  Amended....................................................11582
431.51  (d) amended................................................11582
431.53  (h) amended................................................11582
433.17  Amended....................................................11582
436.212  (e)(5) added; (f) revised.................................19873
436.215  (b) table amended; (c)(12) added...........................6636
440.1a  (a)(3)(ii) introductoryt text amended......................11582
440.2a  (a)(3)(ii) introductory text amended.......................11582
440.80  Added......................................................38674
440.91  Added.......................................................5839
440.103d  (a)(3)(ii) introductory text amended.....................11582
440.103e  (a)(3)(ii) introductory text amended.....................11582
440.103f  (a)(3)(ii) introductory text amended.....................11582
440.209b  (a)(3)(ii) introductory text amended.....................11582
440.241  Redesignated as 440.241a; new 440.241 added.................277
440.241a  Redesignated from 440.241..................................277
440.241b  Added......................................................277
440.249  Redesignated as 440.249a; new 440.249 added.................279
    (b) introductory text corrected.................................2481
440.249a  Redesignated from 440.249..................................279
440.249b  Added......................................................279
440.280c  Added....................................................38675
440.290b  (a)(3)(ii) introductory text amended.....................11582
440.290c  Added.....................................................5840
441.20a  (a)(3)(ii) introductory text amended......................11582
441.220  (a)(3)(ii) introductory text amended......................11582
442.10  (a)(3)(ii) amended.........................................11582
442.12  (a)(3)(ii) amended.........................................11583
442.12a  (a)(3)(ii) amended........................................11583
442.13  (a)(3)(ii) amended.........................................11583
442.14  (a)(3)(ii) amended.........................................11583
442.16a  (a)(3)(ii) amended........................................11583
442.17  (a)(3)(ii) amended.........................................11583
442.17a  (a)(3)(ii) amended........................................11583
442.18a  (a)(3)(ii) introductory text amended......................11583
442.19  (a)(3)(ii) amended.........................................11583
442.20a  (a)(3)(ii) introductory text amended......................11583
442.50a  (a)(3)(ii) introductory text amended......................11583
442.53a  (a)(3)(ii) introductory text amended......................11583
    Technical correction...........................................24026
442.55  (a)(2)(ii) amended.........................................11583
442.55a  (a)(3)(ii) introductory text amended......................11583
442.60  Added......................................................14240
442.70a  Added......................................................6634
442.119  (a)(3)(ii) introductory text amended......................11583
442.211b  (a)(3)(ii) introductory text amended.....................11583
442.212b  (a)(3)(ii) introductory text amended.....................11583
442.213b  (a)(3)(ii) introductory text amended.....................11583
442.214b  (a)(3)(ii) introductory text amended.....................11583
442.216a  (a)(3)(ii) introductory text amended.....................11583
442.217b  (a)(3)(ii) introductory text amended.....................11583
442.223  Amended...................................................11583
442.250  (a)(3)(ii) introductory text amended......................11583
442.255b  (a)(3)(ii) introductory text amended.....................11583
442.260  Added.....................................................14242
442.270a  Added.....................................................6636
444.7  Added.......................................................38676
444.46  (a)(3)(ii) amended.........................................11584
444.206  (a)(1) amended............................................38677
444.246  (a)(3)(ii) introductory text amended......................11584
444.320d  Added.....................................................2643
444.342h  (a)(1) amended...........................................14969
444.342i  (a)(1)(ii) amended.......................................14969
444.342j  (a)(1) amended...........................................14969
444.342k  (a)(1) amended...........................................14969
444.380d  Added......................................................617
444.442g  (a)(1) amended...........................................18598

[[Page 1099]]

444.542k  (a)(3)(ii) introductory text amended.....................11584
444.542l  (a)(3)(ii) introductory text amended.....................11584
    (a)(1) revised; eff. 12-5-91...................................50173
446.120d  (a)(3)(ii) introductory text amended.....................11584
446.121  Redesignated as 446.121a; new 446.121 added................6637
446.121a  Redesignated from 446.121.................................6637
446.121b  Added.....................................................6637
448.23  (a)(3)(ii) amended.........................................11584
448.123  (a)(3)(ii) introductory text amended......................11584
    Redesignated as 448.123a; new 448.123 added....................19873
448.123a  Redesignated from 448.123................................19873
448.123b  Added....................................................19873
    (b)(1)(ii) corrected...........................................22014
448.223  (a)(3)(ii) introductory text amended......................11584
448.510f  (a)(1) revised; eff. 3-15-91..............................9722
    (a)(3)(ii) introductory text amended...........................11584
448.513a  (a)(1) revised; eff. 12-5-91.............................50173
448.513c  (a)(1) revised; eff. 12-5-91.............................50173
448.513d  (a)(3)(ii) introductory text amended.....................11584
448.513e  (a)(1) revised; eff. 10-3-91.............................40381
    (a)(3)(ii) introductory text amended...........................11584
449.150d  (a)(3)(ii) introductory text amended.....................11584
    Technical correction...........................................24026
452.32a  (a)(3)(ii) introductory text amended......................11584
452.110d  (a)(3)(ii) introductory text amended.....................11584
452.115g  Added......................................................280
    Technical correction............................................2481
452.125d  (b)(1) revised...........................................14091
452.510d  (a)(3)(ii) introductory text amended.....................11584
452.910  (a)(3)(ii) amended........................................11584
    (a)(4)(ii) correctly amended............................23634, 25392
453.222  (a)(1) revised.............................................5842
    Technical correction...........................................11109
455.4a  (a)(3)(ii) introductory text amended.......................11584
455.15  (a)(3)(ii) amended.........................................11584
455.15a  (a)(3)(ii) amended........................................11584
455.40  Added.......................................................2641
    (a)(1) introductory text, (a)(1)(iii) and (b)(2) corrected.....11110
    (a)(1) introductory text corrected.............................14378
455.82a  (a)(3)(ii) amended........................................11585
455.185b  (a)(3)(ii) introductory text amended.....................11585
455.204a  Corrected.................................................9317
    (a)(3)(ii) introductory text amended...........................11585
455.510d  (a)(1) amended...........................................11585
455.540  Added......................................................2642

                                  1991

21 CFR
                                                                   56 FR
                                                                    Page
Chapter I
310.545  (a)(20) added; (d) revised................................37798
    Regulation at 56 FR 37792 effective date corrected; (d)(2) 
corrected..........................................................46823
    (a)(7) amended; (d) introductory text revised; (d)(3) added; 
eff. 12-4-92.......................................................63568
312.120  (c)(4) revised............................................22113
312.145  (b) amended................................................3776
314.445  (b) amended................................................3776
333.301--333.350 (Subpart D)  Added; eff. 8-16-92..................41019
358.701--358.750 (Subpart H)  Added................................64568
361.1  (c)(3), (4) and (d)(8) amended..............................10806
429.55  (b) revised; interim.......................................50249
442.216a  (a)(1), (2), (b)(1)(i)(a) and (ii)(a) revised..............484
    (b)(1)(ii)(a)  corrected........................................2585

                                  1992

21 CFR
                                                                   57 FR
                                                                    Page
Chapter I
310  Technical correction....................................2136, 46067
310.201  (a)(25) removed; (b) added; eff. 12-9-93..................58374
310.305  (a) revised; (b)(2), (c)(4) and (d)(1) amended............17980
310.537  Added.....................................................29173

[[Page 1100]]

310.545  (a)(1), (2), (6)(i), (ii), (7), (8) and (12)(iv) amended 
                                                                    3526
    (a)(6)(iii), (d)(4) and (5) added; (d) introductory text and 
(1) revised; eff. 9-14-93..........................................41860
    (a)(3) amended.................................................45295
    (a)(6)(i), (d) introductory text and (1) revised; (d)(6) 
added; eff. 12-9-93................................................58374
    (a)(21), (d)(7) and (8) added; (d) introductory text revised; 
eff. 6-18-93.......................................................60423
    (a)(10)(iv) and (d)(9) added; (d) introductory text revised; 
eff. 6-18-93.......................................................60427
    (a)(22) and (d)(10) added; (d) introductory text revised; eff. 
6-18-93............................................................60431
312  Technical correction..........................................19458
312.34  (a) amended................................................13248
312.35  (a) amended................................................13249
312.42  (b)(3)(iii) and (4) added..................................13249
312.44  (b)(1)(xi) added; (b)(2)(i) revised........................13249
312.145  (b) amended...............................................10814
314  Technical correction..........................................61489
314.1  (a)(1) and (2) amended......................................17981
314.3  (b) revised.................................................17981
314.50  Introductory text amended, (a)(2) and (c)(1) amended; 
        (g)(3) added...............................................17982
314.54  Added......................................................17982
    (a)(2) corrected...............................................61612
314.55  Removed....................................................17983
314.56  Removed....................................................17983
314.60  Existing text designated as (a) and amended; (b) added.....17983
314.70  (e) added..................................................17983
314.71  (b) amended................................................17983
314.80  (a), (c)(1)(ii) and (d)(1) amended; (b) revised............17983
314.81  (b)(3)(iii) added..........................................17983
314.92--314.99 (Subpart C)  Added..................................17983
314.94  (a)(9)(iii) corrected......................................29353
314.100--314.170 (Subpart C)  Redesignated as subpart D............17983
    Heading revised................................................17987
314.100  Revised...................................................17987
314.101  Revised...................................................17987
    (d)(3) and (4) corrected.......................................29353
314.102  Revised...................................................17988
    (b) corrected..................................................29353
314.103  (a) revised; (b) and (c)(2) amended.......................17989
314.104  Revised...................................................17989
314.105  Revised...................................................17989
314.110  Revised...................................................17989
314.120  Revised...................................................17990
314.122  Added.....................................................17990
    (b) corrected..................................................29353
314.125  Heading, (a) introductory text, (b) introductory text, 
        (7), (9), (10), (12), (14), (15), (16) and (17) revised; 
        (b)(18) added..............................................17991
314.127  Added.....................................................17991
    (a)(3)(iii)(A)(1) corrected....................................29353
314.150  Revised...................................................17993
314.151  Added.....................................................17994
314.152  Revised...................................................17994
314.153  Added.....................................................17995
314.160  Revised...................................................17995
314.161  Added.....................................................17995
314.162  Added.....................................................17996
314.200--314.235 (Subpart D)  Redesignated as subpart E............17983
314.200  (a) introductory text, (b)(1) and (c)(3) revised; (c)(1) 
        and (g)(1) amended.........................................17996
314.300 (Subpart E)  Redesignated as subpart F.....................17983
314.410--314.445 (Subpart F)  Redesignated as subpart G............17983
314.430  Heading, (a) through (d), (e) introductory text, (f)(5), 
        (6) and (g) introductory text revised......................17996
314.440  Heading, (a) introductory text, (1) and (2) revised.......17997
314.500--314.560 (Subpart H)  Added................................58958
316  Added.........................................................62085
320.1  (a) and (e) revised.........................................17997
320.21--320.32 (Subpart B)  Heading revised........................17998
320.21  Revised....................................................17998
320.22  Revised....................................................17998
320.23  Revised....................................................17999
320.24  Revised....................................................17999
    (b)(4) corrected...............................................29354
320.30  Revised....................................................18000
320.31  Revised....................................................18000
320.32  Redesignated as 320.38; new 320.32 redesignated from 
        320.51 and revised.........................................18000
320.33  Redesignated from 320.52; heading and introductory text 
        revised....................................................18001
320.34  Redesignated from 320.55...................................18001

[[Page 1101]]

320.35  Redesignated from 320.56...................................18001
320.36  Redesignated from 320.62...................................18001
320.38  Redesignated from 320.32...................................18000
320.50  Removed....................................................18000
320.51  Redesignated as 320.32.....................................18000
320.52  Redesignated as 320.33.....................................18001
320.53  Removed....................................................18001
320.54  Removed....................................................18001
320.55  Redesignated as 320.34.....................................18001
320.56  Redesignated as 320.35.....................................18001
320.57  Removed....................................................18001
320.58  Removed....................................................18001
320.59  Removed....................................................18001
320.60  Removed....................................................18001
320.61  Removed....................................................18001
320.62  Redesignated as 320.36.....................................18001
341.3  (e) added; eff. 12-9-93.....................................58374
341.12  Added; eff. 12-9-93........................................58374
341.72  Added; eff. 12-9-93........................................58374
341.78  (b) amended; (c)(1) and (2) redesignated as (c)(2) and 
        (1); new (c)(2) revised; (c)(3) added......................29177
341.90  (e) through (q) added; eff. 12-9-93........................58376
348  Added; eff. 6-19-93...........................................27656
358  Technical correction...........................................2136
    Authority citation revised.....................................44494
358.550  (d)(2) revised............................................44494
369.20  Amended; eff. 12-9-93......................................58376
369.21  Amended; eff. 12-9-93......................................58376
433.1  (d)(2) amended..............................................18001

                                  1993

21 CFR
                                                                   58 FR
                                                                    Page
Chapter I
310  Authority citation revised.............................46745, 54462
310.201  (a)(29) removed; eff. 9-23-94.............................49898
310.530  Added.....................................................47610
310.531  Added; eff. 5-16-94.......................................60336
310.536  Added.....................................................46754
310.538  Added.....................................................47605
310.544  Added.....................................................31241
310.545  (a)(8) and (18) text redesignated as (a)(8)(i) and 
        (18)(i); new (a)(8)(i) heading, (10)(v), (vi) and (vii), 
        (18)(i) heading, (ii), (iii), (vi), (22)(ii), (23), (24), 
        (25) and (d)(11) added; (d) introductory text and (1) 
        revised....................................................27641
    (a)(19) removed................................................31241
    (a)(22)(iii) and (d)(12) added; (d) introductory text revised 
                                                                   46745
    (a)(26), (d)(13) and (14) added; (d) introductory text 
revised; eff. 9-2-94...............................................46748
    (a)(13) removed................................................46754
    (a)(11) removed................................................47605
    (a)(22)(iv) and (d)(15) added; (d) introductory text revised; 
eff. 9-23-94.......................................................49898
    (a)(8)(iii) and (d)(21) added; (d) introductory text revised; 
eff. 4-21-94.......................................................54455
    (a)(18)(ii) amended; (d)(11) revised; (d)(22) added; eff. 10-
21-94..............................................................54462
    (a)(5) removed; eff. 5-16-94...................................60337
312.57  (c) revised................................................25926
314  Authority citation revised....................................47351
    Technical correction............................................4078
314.50  Introductory text and (h)(2) revised; (d)(1)(ii) 
        redesignated as (d)(1)(ii)(a); (d)(1)(ii)(b), (c), 
        (d)(1)(v), (h)(3) and (4) added............................47351
314.54  (a)(1)(i) revised; (a)(2) amended; (a)(4) added............47351
314.60  (c) added..................................................47352
314.70  (a) revised................................................47352
    (b)(2)(xi), (xii) and (d)(9) added; eff. 9-13-95...............47959
314.71  (b) revised................................................47352
314.94  Introductory text amended; (a)(9)(i) and (d)(4) revised; 
        (d)(5) added...............................................47352
314.96  (b) redesignated as (c); new (b) added.....................47352
314.125  (b)(17) revised...........................................25926
314.127  (b) revised...............................................25927
314.150  (b)(9) revised............................................25927
314.440  (a)(1) and (2) amended; (a)(4) added......................47352
316.10  (b)(10) corrected...........................................6167
316.27  (a)(2)(ii) corrected........................................6167

[[Page 1102]]

320.31  (c) revised; (d) added; (e) and (f) removed................25927
320.38  Revised....................................................25927
320.63  Revised....................................................25928
330.3  Added; eff. 9-13-95.........................................47959
331.30  (d) introductory text revised; (h) added; eff. 8-26-94.....45208
333.201--333.280 (Subpart C)  Added; eff. 9-23-94..................49898
341.74  (c)(4)(v) and (vi) added; eff. 10-20-94....................54236
341.76  (c)(4) revised; eff. 10-20-94..............................54242
347  Added; eff. 10-21-94..........................................54462
358.601--358.650 (Subpart G)  Added; eff. 12-14-94.................65455
430.4  (a)(64) added...............................................26652
    (a)(65) added..................................................26656
    (a)(66) added..................................................26659
    (a)(67) added..................................................26663
    (a)(68) added..................................................26666
430.5  (a)(99) and (b)(101) added..................................26652
    (a)(100) and (b)(102) added....................................26656
    (a)(101) and (b)(103) added....................................26659
    (a)(102) and (b)(104) added....................................26663
    (a)(103) and (b)(105) added....................................26666
430.6  (b)(101) added..............................................26653
    (b)(102) added.................................................26656
    (b)(103) added.................................................26659
    (b)(104) added.................................................26663
    (b)(105) added.................................................26666
436.215  (b) table amended; (c)(13) added..........................26653
    (b) table amended; (c)(14) added...............................26656
    (b) table amended; (c)(15) added...............................26660
    (b) table amended; (c)(16) added...............................26667
436.368  Added.....................................................26660
441.220  Redesignated as 441.220b..................................26669
    Added..........................................................26670
441.220a  Added....................................................26670
441.220b  Redesignated from 441.220................................26669
442.80  Added......................................................26660
442.180  Added.....................................................26661
442.180a  Added....................................................26661
442.180b  Added....................................................26661
443  Added.........................................................26667
444.380e  Added....................................................26671
450.30  Added......................................................26664
450.230  Added.....................................................26665
452.50  Added......................................................26653
452.60  Added......................................................26657
452.150  Added.....................................................26654
452.160  Added.....................................................26658

                                  1994

21 CFR
                                                                   59 FR
                                                                    Page
Chapter I
310  Technical corrections.........................................49350
310.201  (a)(13) removed; eff. 1-30-95..............................4218
310.545  (a)(6)(ii) redesignated as (a)(6)(ii)(A); new 
        (a)(6)(ii)(A) heading, (B) and (d)(23) added; (d) 
        introductory text and (1) revised; eff. 8-23-95............43409
310.546  Added.....................................................43252
314  Authority citation revised....................................13200
314.50  (h) redesignated as (k); new (h), (i) and (j) added........50361
    (b) corrected..................................................60051
    (d)(5)(vi)(a) amended..........................................13200
314.52  Added......................................................50362
314.53  Added......................................................50363
314.54  (a)(1)(vii) added..........................................50364
314.70  (f) added..................................................50364
314.94  (a)(12) added..............................................50364
314.95  Added......................................................50366
314.101  (e) revised...............................................50366
314.107  Added.....................................................50367
314.108  Added.....................................................50368
314.200  (c)(1) amended............................................14364
314.300  (b)(4) amended............................................14365
330.1  (i) added....................................................4000
    (g) amended....................................................14365
331.30  (d)(1) removed.............................................60556
336.50  (c)(1) revised; eff. 4-11-95...............................16982
338.50  (c)(3) revised; eff. 4-11-95...............................16983
341.3  (f), (g) and (h) added; eff. 8-23-95........................43409
341.12  (h) added; eff. 1-30-95.....................................4218
341.14  (a)(5) and (6) added; eff. 6-5-95..........................29174
341.20  Added; eff. 8-23-95........................................43409
341.72  (c)(4) heading and (6)(iii) heading revised; (d)(8) added; 
        eff. 1-30-95................................................4218
341.74  (c)(4)(vii), (viii), (ix), (d)(1)(iv) and (v) added; eff. 
        6-5-95.....................................................29174

[[Page 1103]]

    (d)(1)(iv) and (v) corrected; eff. 6-5-95......................36051
341.80  Added; eff. 8-23-95........................................43409
341.90  (r) and (s) added; eff. 6-5-95.............................29174
    (r) and (s) corrected; eff. 6-5-95.............................36051
    (l) added; eff. 1-30-95.........................................4218
343  Announcement..................................................37421
346.18  (b) revised................................................28767
346.50  (b)(2)(vi) heading and (d)(8) heading amended..............28767
347.10  (c) revised................................................28768
347.50  (b)(3) heading, (c)(2) heading and (d)(3) heading amended 
                                                                   28768
358  Document availability.........................................62569
358.150  (d)(3) revised; eff. 11-23-95.............................60317
358.703  (e) added; eff. 1-30-95....................................4001
358.710  (a)(6) redesignated as (a)(7); new (a)(6) added; eff. 1-
        30-95.......................................................4001
369.20  Amended; eff. 8-23-95......................................43412
369.21  Amended; eff. 1-30-95.......................................4218
430.4  (a)(69) added...............................................40806
430.5  (a)(104) and (b)(106) added.................................40806
430.6  (b)(106) added..............................................40806
    (b)(89) revised.................................................8133
436.215  (b) table amended; (c)(18) added..........................40806
    (b) table amended...............................................8133
    (b) table amended; (c)(17) added................................8856
436.369  Added.....................................................40806
436.370  Added.....................................................40807
441.20a  (a)(1)(i) revised..........................................8133
442.7  Added........................................................8857
442.52  Added......................................................26940
442.69  Added; eff. 4-18-94........................................12546
442.107  Added......................................................8857
442.107a  Added.....................................................8857
442.107b  Added.....................................................8857
442.216a  (a)(1), (b)(1)(ii)(a) and (4) revised.....................8398
442.253  Redesignated as 442.253a; new 442.253 heading added.......26941
442.253a  Redesignated from 442.253................................26941
442.253b  Added....................................................26941
442.270  Added; eff. 4-18-94.......................................12546
442.270b  Added; eff. 4-18-94......................................12546
444.320c  (a)(1) amended............................................8398
444.342a  (a)(1) amended............................................8398
444.342c  (a)(1) amended............................................8398
444.342d  (a)(1) amended............................................8399
444.342i  (a)(1)(ii) amended........................................8399
444.342j  (a)(1) amended............................................8399
444.380a  (a)(1) amended............................................8399
444.380c  (a)(1) amended............................................8399
448.330  (a)(1) amended.............................................8399
450.10  Added......................................................50485
450.24  (a)(1)(i), (ii), (v), (3)(i), (b)(1) and (2) revised; 
        (a)(1)(viii) and (b)(8) added; (b) introductory text 
        amended.....................................................9639
450.224a  (a)(1), (3)(i)(a), (b), (b)(1) and (3) revised; (b) 
        introductory text amended; (b)(4) removed...................9641
450.224b  (a)(1), (3)(i)(A), (B), (b)(1) and (4) revised; (b) 
        introductory text amended; (b)(2) removed...................9641
452.160  Redesignated as 452.160a; new 452.160 added...............52078
452.160a  Redesignated from 452.160................................52078
452.160b  Added....................................................52078
455.86  Added.......................................................8400
455.88  Added......................................................40807
    (a)(1) corrected...............................................46479
455.185a  (a)(1) amended............................................8399
455.188  Added.....................................................40808
455.285c  Added.....................................................8400

                                  1995

21 CFR
                                                                   60 FR
                                                                    Page
Chapter I
310  Technical correction...................................17611, 57927
310.201  (a)(10) and (15) removed; eff. 10-7-96....................52507
310.543  Added.....................................................20165
310.545  (a)(7) corrected; CFR correction...........................5313
    (a)(15) and (d)(1) revised; (d)(18) added; eff. 8-15-95.........8920
    (a)(9) removed; (d)(1) revised.................................20165
    (d)(1) corrected...............................................20898
    (a)(6)(iv), (d)(19) and (20) added.............................38642
    (a)(15)(ii) stayed in part.....................................42436
    (a)(2) redesignated as (a)(2)(i); (a)(2)(i) heading, (ii) and 
(d)(24) added; (d) introductory text and (1) revised; eff. 10-7-96
                                                                   52507
328  Added; eff. 3-13-96...........................................13595

[[Page 1104]]

355  Added; eff. 10-7-96...........................................52507
    Technical correction...........................................57927
355.50  (a) corrected..............................................57927
369  Technical correction..........................................57927
369.21  Amended; eff. 10-7-96......................................52510
429.55  (b) revised; interim.......................................56516
430.4  (a)(70) added...............................................58230
430.5  (a)(105) and (b)(107) added.................................58230
430.6  (b)(107) added..............................................58230
436.105  Table corrected; CFR correction...........................66898
436.106  Table corrected; CFR correction...........................66898
436.215  (b) table amended; (c)(9) revised.........................27221
    (b) table amended; (c)(19) added...............................58230
442.52  (b)(1)(iv) and (3) revised.................................33712
442.54  Added......................................................58231
442.119  Redesignated as 442.119a; new 442.119 added...............27222
442.119a  Redesignated from 442.119................................27222
442.119b  Added....................................................27222
442.154  Added.....................................................58232
442.154a  Added....................................................58232
442.154b  Added....................................................58233
450.10  Stayed.....................................................11027
    Removed........................................................16377
453.522d  Added....................................................49508

                                  1996

21 CFR
                                                                   61 FR
                                                                    Page
310.101  Removed...................................................29476
310.304  Removed...................................................29477
310.545  (a)(6)(ii)(B) stayed in part...............................9571
    (a)(6)(iv)(C) and (d)(25) added; (d) introductory text revised
                                                                   25142
312.2  (b)(6) added................................................51529
312.20  (c) added..................................................51529
312.23  (f) added..................................................51529
312.30  Introductory text amended..................................51529
312.42  (b)(5) added...............................................51530
312.53  (c)(1)(vi)(d) revised......................................57280
312.54  Added......................................................51530
312.60  Amended....................................................51530
312.62  (b) revised................................................57280
312.130  (d) added.................................................51530
314.430  (d) redesignated as (d)(1); (d)(2) added..................51530
328.10  (f) redesignated as (g); new (f) added.....................58630
331  Comment period extension......................................38046
331.10   (a) revised; eff. 2-10-97..................................4823
331.20  Removed; new 331.20 redesignated from 331.21 and revised; 
        eff. 2-10-97................................................4823
331.21  Redesignated as 331.20; new 331.21 redesignated from 
        331.29 and revised; eff. 2-10-97............................4823
331.22  Removed; eff. 2-10-97.......................................4823
331.23  Removed; eff. 2-10-97.......................................4823
331.24  Removed; eff. 2-10-97.......................................4823
331.25  Removed; eff. 2-10-97.......................................4823
331.26  Removed; eff. 2-10-97.......................................4823
331.29  Redesignated as 331.21; eff. 2-10-97........................4823
331.30  (c)(5) and (f) removed; (c)(6), (7) and (g) redesignated 
        as (c)(5), (6) and (f).....................................17806
331.80  (a)(1) revised; eff. 2-10-97................................4823
332  Authority citation revised.....................................8838
332.3  Added; eff. 3-5-97...........................................8838
332.30--332.31 (Subpart D)  Redesignated as 332.30--332.31 
        (Subpart C); eff. 3-5-97....................................8838
332.30  (a), (b) and (c) redesignated as (b), (c) and (d); heading 
        and new (b) revised; new (a) added; eff. 3-5-97.............8838
333.150  (c)(2) heading and (3) added; eff. 11-17-97...............58472
341.70  Added......................................................15703
341.76  (d)(2)(i) and (e) removed; (d)(2)(ii) redesignated as 
        (d)(2) heading.............................................25146
355.3  (e) revised; eff. 4-7-97....................................52286
355.10  (a)(1) heading, (b)(1) heading, (2) heading and (c)(1) 
        heading amended; eff. 4-7-97...............................52286
355.50  Regulation at 60 FR 52508 eff. date delayed to 4-7-97......52285
    (c)(1) and (2) revised; (d)(1)(i) heading and (ii) heading 
amended; eff. 4-7-97...............................................52287
355.70  (a) stayed in part 9-23-96 through 10-7-97.................65946
369.21  (d) amended; interim.......................................20101
452.150  Redesignated as 452.150a; new 452.150 added...............34726
452.150a  Redesignated from 452.150................................34726

[[Page 1105]]

452.150b  Added....................................................34726

                                  1997

21 CFR
                                                                   62 FR
                                                                    Page
300  Authority citation revised....................................51516
310  Authority citation revised....................................51516
310.305  (a) and (f)(1) revised; (b)(5) and (c)(4) removed; (c)(5) 
        and (6) redesignated as (c)(4) and (5); new (c)(4) amended
                                                                   34167
    (a), (d)(2), (3) introductory text and (i) amended; (b), (c), 
(d)(1), (3)(ii) and (4) revised; eff. 4-6-98.......................52249
310.502  Revised; eff. 4-14-97.....................................12084
310.504  Removed; eff. 4-14-97.....................................12084
310.506  Removed; eff. 4-14-97.....................................12084
310.507  Removed; eff. 4-14-97.....................................12084
310.508  Removed; eff. 4-14-97.....................................12084
310.509  Revised; eff. 4-14-97.....................................12084
310.510  Removed; eff. 4-14-97.....................................12085
310.513  Removed; eff. 4-14-97.....................................12085
310.518  Added; eff. 7-15-97........................................2250
    (b)(2) and (c)(5) corrected; eff. 7-15-97......................15111
310.525  Removed; eff. 4-14-97.....................................12085
310.526  Removed; eff. 4-14-97.....................................12085
312  Meeting.......................................................46198
    Technical correction...........................................47760
    Authority citation revised.....................................51516
312.20  (c) amended................................................32479
312.23  (a)(7)(iv)(e) revised......................................40599
312.32  (a), (b), (c)(1)(i), (2) and (d)(3) revised; (c)(3) and 
        (e) amended; eff. 4-6-98...................................52250
312.70  (a) and (b) amended........................................46876
314  Meeting................................................44891, 46198
    Technical correction...........................................47760
    Authority citation revised.....................................51516
    Clarification..................................................63268
314.50  (d)(1)(iii) revised........................................40599
314.70  (g) added..................................................39900
314.80  (c)(1)(ii) removed; (c)(1)(iii) and (iv) redesignated as 
        (c)(1)(ii) and (iii); (a), new (c)(1)(ii) introductory 
        text, (d)(1) and (l) amended; (f)(1) revised...............34168
    (a), (c) introductory text, (1), (f)(1), (3)(ii) and (4) 
revised; (b), (c)(2)(ii)(b), (d)(2), (f) heading, (2) and (3) 
amended; (j) removed; (k) and (l) redesignated as (j) and (k); 
eff. 4-6-98........................................................52251
314.101  (d)(4) revised............................................40599
314.110  (a)(3) and (b) amended....................................43639
314.120  (a)(3) amended............................................43639
314.440  (a)(3) amended............................................43639
316  Authority citation revised....................................51516
320  Authority citation revised....................................51516
329  Authority citation revised....................................51516
330  Authority citation revised....................................51516
331  Authority citation revised....................................51516
331.10  (a) correctly revised; CFR correction......................52659
332  Authority citation revised....................................51516
333  Authority citation revised....................................51516
336  Authority citation revised....................................51516
338  Authority citation revised....................................51516
340  Authority citation revised....................................51516
341  Authority citation revised....................................51516
341.12  (h) correctly revised; CFR correction.......................6866
341.76  (d)(2) heading correctly removed............................9684
344  Authority citation revised....................................51517
346  Authority citation revised....................................51517
347  Authority citation revised....................................51517
348  Authority citation revised....................................51517
349  Authority citation revised....................................51517
352  Announcement..................................................23350
355  Authority citation revised....................................51517
357  Authority citation revised....................................51517

[[Page 1106]]

358  Authority citation revised....................................51517
361  Authority citation revised....................................51517
369  Authority citation revised....................................51517
429  Authority citation revised....................................51517
430  Authority citation revised....................................51517
431  Authority citation revised....................................51517
432  Authority citation revised....................................51517
433  Authority citation revised....................................51517
436  Authority citation revised....................................51517
436.215  (c)(16)(iv) and (18)(iv) corrected; CFR correction........52659
440  Authority citation revised....................................51517
441  Authority citation revised....................................51517
442  Authority citation revised....................................51517
443  Authority citation revised....................................51517
444  Authority citation revised....................................51517
446  Authority citation revised....................................51517
448  Authority citation revised....................................51517
449  Authority citation revised....................................51517
450  Authority citation revised....................................51518
452  Authority citation revised....................................51518
453  Authority citation revised....................................51518
455  Authority citation revised....................................51518
460  Authority citation revised....................................51518

                                  1998

     (Regulations published January 1, 1998, through April 1, 1998)

21 CFR
                                                                   63 FR
                                                                    Page
310.547  Added; eff. 4-20-98.......................................13528
312.33  (a)(2) revised; eff. 8-10-98................................6862
312.53  (c)(4) added; eff. 2-2-99...................................5252
312.57  (b) and (c) redesignated as (c) and (d); new (b) added; 
        eff. 2-2-99.................................................5252
312.64  (d) added; eff. 2-2-99......................................5252
314.50  (k) redesignated as (l); new (k) added; eff. 2-2-99.........5252
    (d)(5)(v) and (vi)(a) amended; eff. 8-10-98.....................6862
314.60  (a) amended; eff. 2-2-99....................................5252
314.80  (k) corrected..............................................14611
314.94  (a)(13) added; eff. 2-2-99..................................5252
314.200  (d)(3) amended; eff. 2-2-99................................5252
314.300  (b)(6) introductory text amended; eff. 2-2-99..............5252
320.36  Existing text designated as (a); (b) added; eff. 2-2-99.....5252
330.10  (f) added; eff. 2-2-99......................................5253