96-64. International Conference on Harmonisation; Guideline on Impurities in New Drug Substances; Availability  

  • [Federal Register Volume 61, Number 3 (Thursday, January 4, 1996)]
    [Notices]
    [Pages 372-376]
    From the Federal Register Online via the Government Publishing Office [www.gpo.gov]
    [FR Doc No: 96-64]
    
    
    
    
    [[Page 371]]
    
    _______________________________________________________________________
    
    Part II
    
    
    
    
    
    Department of Health and Human Services
    
    
    
    
    
    _______________________________________________________________________
    
    
    
    Food and Drug Administration
    
    
    
    _______________________________________________________________________
    
    
    
    International Conference on Harmonisation, Guidelines Availability: 
    Impurities in New Drug Substances; Notice
    
    Federal Register / Vol. 61, No. 3 / Thursday, January 4, 1996 / 
    Notices
    
    [[Page 372]]
    
    
    DEPARTMENT OF HEALTH AND HUMAN SERVICES
    
    Food and Drug Administration
    [Docket No. 94D-0325]
    
    
    International Conference on Harmonisation; Guideline on 
    Impurities in New Drug Substances; Availability
    
    AGENCY: Food and Drug Administration, HHS.
    
    ACTION: Notice.
    
    -----------------------------------------------------------------------
    
    SUMMARY: The Food and Drug Administration (FDA) is publishing a 
    guideline entitled ``Impurities in New Drug Substances.'' The guideline 
    was prepared under the auspices of the International Conference on 
    Harmonisation of Technical Requirements for Registration of 
    Pharmaceuticals for Human Use (ICH). The guideline is intended to 
    provide guidance to applicants for drug marketing registration on the 
    content and qualification of impurities in new drug substances produced 
    by chemical syntheses and not previously registered in a country, 
    region, or member State.
    
    DATES: Effective January 4, 1996. Submit written comments at any time.
    
    ADDRESSES: Submit written comments on the guideline to the Dockets 
    Management Branch (HFA-305), Food and Drug Administration, rm. 1-23, 
    12420 Parklawn Dr., Rockville, MD 20857. Copies of the guideline are 
    available from the Consumer Affairs Branch (previously the CDER 
    Executive Secretariat Staff) (HFD-210), Center for Drug Evaluation and 
    Research, Food and Drug Administration, 7500 Standish Pl., Rockville, 
    MD 20855.
    FOR FURTHER INFORMATION CONTACT:
        Regarding the guideline: Robert W. Trimmer, Center for Drug 
    Evaluation and Research (HFD-625), Food and Drug Administration, 7500 
    Standish Pl., Rockville, MD 20855, 301-594-0370.
        Regarding the ICH: Janet J. Showalter, Office of Health Affairs 
    (HFY-20), Food and Drug Administration, 5600 Fishers Lane, Rockville, 
    MD 20857, 301-827-0864.
    
    SUPPLEMENTARY INFORMATION: In recent years, many important initiatives 
    have been undertaken by regulatory authorities and industry 
    associations to promote international harmonization of regulatory 
    requirements. FDA has participated in many meetings designed to enhance 
    harmonization and is committed to seeking scientifically based 
    harmonized technical procedures for pharmaceutical development. One of 
    the goals of harmonization is to identify and then reduce differences 
    in technical requirements for drug development among regulatory 
    agencies.
        ICH was organized to provide an opportunity for tripartite 
    harmonization initiatives to be developed with input from both 
    regulatory and industry representatives. FDA also seeks input from 
    consumer representatives and others. ICH is concerned with 
    harmonization of technical requirements for the registration of 
    pharmaceutical products among three regions: The European Union, Japan, 
    and the United States. The six ICH sponsors are the European 
    Commission, the European Federation of Pharmaceutical Industries 
    Associations, the Japanese Ministry of Health and Welfare, the Japanese 
    Pharmaceutical Manufacturers Association, the Centers for Drug 
    Evaluation and Research and Biologics Evaluation and Research, FDA, and 
    the Pharmaceutical Research and Manufacturers of America. The ICH 
    Secretariat, which coordinates the preparation of documentation, is 
    provided by the International Federation of Pharmaceutical 
    Manufacturers Associations (IFPMA).
        The ICH Steering Committee includes representatives from each of 
    the ICH sponsors and the IFPMA, as well as observers from the World 
    Health Organization, the Canadian Health Protection Branch, and the 
    European Free Trade Area.
        In the Federal Register of September 22, 1994 (59 FR 48740), FDA 
    published a draft tripartite guideline entitled ``Impurities in New 
    Drug Substances.'' The notice gave interested persons an opportunity to 
    submit comments by December 6, 1994.
        After consideration of the comments received and revisions to the 
    guideline, a final draft of the guideline was submitted to the ICH 
    Steering Committee and endorsed by the three participating regulatory 
    agencies at the ICH meeting held in March 1995.
        The guideline is intended to provide guidance to applicants for 
    drug marketing registration on the content and qualification of 
    impurities in new drug substances produced by chemical syntheses and 
    not previously registered in a country, region, or member State. The 
    guideline is not intended to apply to new drug substances used during 
    the clinical research stage of development or clinical trials. The 
    guideline also does not apply to biological/biotechnological 
    substances, peptides, oligonucleotides, radiopharmaceuticals, 
    fermentation and semisynthetic products derived from that process, 
    herbal products, and crude products of animal or plant origin. 
    Impurities in new drug substances are addressed in the guideline from 
    two perspectives: (1) Chemistry aspects--classification and 
    identification of impurities, report generation, setting 
    specifications, and a brief discussion of analytical procedures; and 
    (2) safety aspects--guidance for qualifying impurities that were not 
    present in batches of the new drug substance used in safety and 
    clinical studies and/or impurity levels substantially higher than in 
    those batches.
        In the past, guidelines have generally been issued under 
    Sec. 10.90(b) (21 CFR 10.90(b)), which provides for the use of 
    guidelines to state procedures or standards of general applicability 
    that are not legal requirements but that are acceptable to FDA. The 
    agency is now in the process of revising Sec. 10.90(b). Therefore, the 
    guideline is not being issued under the authority of Sec. 10.90(b). 
    Although this guideline does not create or confer any rights on or for 
    any person, and does not operate to bind FDA in any way, it does 
    represent the agency's current thinking on the content and 
    qualification of impurities in new drug substances produced by chemical 
    syntheses and not previously registered in a country, region, or member 
    state.
        As with all of FDA's guidelines, the public is encouraged to submit 
    written comments with new data or other new information pertinent to 
    this guideline. The comments in the docket will be periodically 
    reviewed, and, where appropriate, the guideline will be amended. The 
    public will be notified of any such amendments through a notice in the 
    Federal Register.
        Interested persons may, at any time, submit to the Docket 
    Management Branch (address above) written comments on the guideline. 
    Two copies of any comments are to be submitted, except that individuals 
    may submit one copy. Comments are to be identified with the docket 
    number found in brackets in the heading of this document. The guideline 
    and received comments may be seen in the office above between 9 a.m. 
    and 4 p.m., Monday through Friday.
        The text of the guideline follows:
    
    Impurities in New Drug Substances
    
    1. Preamble
    
        This document is intended to provide guidance for registration 
    applications on the content and qualification of impurities in new 
    drug substances produced by chemical syntheses and not previously 
    registered in a region or member state. It is not intended to apply 
    to the regulation of new drug substances used during the clinical 
    research 
    
    [[Page 373]]
    stage of development. Biological/biotechnological, peptide, 
    oligonucleotide, radiopharmaceutical, fermentation and semisynthetic 
    products derived therefrom, herbal products, and crude products of 
    animal or plant origin are not covered. Impurities in new drug 
    substances are addressed from two perspectives:
        Chemistry aspects include classification and identification of 
    impurities, report generation, setting specifications, and a brief 
    discussion of analytical procedures; and
        Safety aspects include specific guidance for qualifying 
    impurities which were not present in batches of new drug substance 
    used in safety and clinical studies and/or impurity levels 
    substantially higher than in those batches. Threshold limits are 
    defined, below which, qualification is not needed.
    
    2. Classification of Impurities
    
        Impurities may be classified into the following categories:
         Organic Impurities (Process and Drug Related)
         Inorganic Impurities
         Residual Solvents
        Organic impurities may arise during the manufacturing process 
    and/or storage of the new drug substance. They may be identified or 
    unidentified, volatile or nonvolatile, and include:
         Starting Materials
         By-Products
         Intermediates
         Degradation Products
         Reagents, Ligands, and Catalysts
        Inorganic impurities may derive from the manufacturing process. 
    They are normally known and identified, and include:
         Reagents, Ligands, and Catalysts
         Heavy Metals
         Inorganic Salts
         Other Materials (e.g., Filter Aids, Charcoal, etc.)
         Solvents are organic or inorganic liquids used during the 
    manufacturing process. Since these are generally of known toxicity, 
    the selection of appropriate controls is easily accomplished.
        Excluded from this document are: Extraneous contaminants which 
    should not occur in new drug substances and are more appropriately 
    addressed as good manufacturing practice issues; polymorphic form, a 
    solid state property of the new drug substance; and enantiomeric 
    impurities.
    
    3. Rationale for the Reporting and Control of Impurities
    
    3.1 Organic Impurities
    
        The applicant should summarize those actual and potential 
    impurities most likely to arise during the synthesis, purification, 
    and storage of the new drug substance. This summary should be based 
    on sound scientific appraisal of the chemical reactions involved in 
    the synthesis, impurities associated with raw materials which could 
    contribute to the impurity profile of the new drug substance, and 
    possible degradation products. This discussion may only include 
    those impurities that may reasonably be expected based on knowledge 
    of the chemical reactions and conditions involved.
        In addition, the applicant should summarize the laboratory 
    studies conducted to detect impurities in the new drug substance. 
    This summary should include test results of batches manufactured 
    during the development process and batches from the proposed 
    commercial process, as well as results of intentional degradation 
    studies used to identify potential impurities that arise during 
    storage. Assessment of the proposed commercial process may be 
    deferred until the first batch is produced for marketing. The 
    impurity profile of the drug substance lots intended for marketing 
    should be compared with those used in development and any 
    differences discussed.
        The studies conducted to characterize the structure of actual 
    impurities present in the new drug substance at or above an apparent 
    level of 0.1 percent (e.g., calculated using the response factor of 
    the drug substance) should be described. Note that all recurring 
    impurities at or above the 0.1 percent level in batches manufactured 
    by the proposed commercial process should be identified. Degradation 
    products observed in stability studies at recommended storage 
    conditions should be similarly identified. When identification of an 
    impurity is not feasible, a summary of the laboratory studies 
    demonstrating the unsuccessful effort should be included in the 
    application. Where attempts have been made to identify impurities 
    below the 0.1 percent level, it is useful to also report the results 
    of these studies.
        Identification of impurities below apparent levels of 0.1 
    percent is generally not considered necessary. However, 
    identification should be attempted for those potential impurities 
    that are expected to be unusually potent, producing toxic or 
    pharmacologic effects at a level lower than 0.1 percent. In all 
    cases, impurities should be qualified as described later in this 
    guide. Although it is common practice to round analytical results of 
    between 0.05 and 0.09 percent to the nearest number (i.e., 0.1 
    percent), for the purpose of these guidelines, such values would not 
    be rounded to 0.1 percent and these impurities would not require 
    identification.
    
    3.2 Inorganic Impurities
    
        Inorganic impurities normally are detected and quantitated using 
    pharmacopeial or other appropriate procedures. Carry over of 
    catalysts to the new drug substance should be evaluated during 
    development. The need for inclusion or exclusion of inorganic 
    impurities in the new drug substance specifications should be 
    discussed. Limits should be based on pharmacopeial standards or 
    known safety data.
    
    3.3 Solvents
    
        The control of residues of the solvents used in the 
    manufacturing process for the new drug substance should be 
    discussed. Any solvents which may appear in the drug substance 
    should be quantified using analytical procedures with an appropriate 
    level of sensitivity. Pharmacopeial or other appropriate procedures 
    should be utilized. Limits should be based on pharmacopeial 
    standards or known safety data taking into consideration dose, 
    duration of treatment, and route of administration. Particular 
    attention should be given to quantitation of toxic solvents used in 
    the manufacturing process.
    
    4. Analytical Procedures
    
        The registration application should include documented evidence 
    that the analytical procedures are validated and suitable for the 
    detection and quantitation of impurities. Differences in the 
    analytical procedures used during development and proposed for the 
    commercial product should be discussed in the registration 
    application.
        Organic impurity levels can be measured by a variety of 
    techniques, including those which compare an analytical response for 
    an impurity to that of an appropriate reference standard or to the 
    response of the new drug substance itself. Reference standards used 
    in the analytical procedures for control of impurities should be 
    evaluated and characterized according to their intended uses. The 
    drug substance may be used to estimate the levels of impurities. In 
    cases where the response factors are not close, this practice may 
    still be acceptable, provided a correction factor is applied or the 
    impurities are, in fact, being overestimated. Specifications and 
    analytical procedures used to estimate identified or unidentified 
    impurities often are based on analytical assumptions (e.g., 
    equivalent detector response, etc.). The assumptions should be 
    discussed in the registration application.
    
    5. Reporting Impurity Content of Batches
    
        Analytical results should be provided for all batches of the new 
    drug substance used for clinical, safety, and stability testing, as 
    well as batches representative of the proposed commercial process. 
    The content of individual identified and unidentified and total 
    impurities observed in these batches of the new drug substance 
    should be reported with the analytical procedures indicated. A 
    tabulation (e.g., spreadsheet) of the data is recommended. 
    Impurities should be designated by code number or by an appropriate 
    descriptor, e.g., retention time. Levels of impurities which are 
    present but are below the validated limit of quantitation need not 
    be reported. When analytical procedures change during development, 
    reported results should be linked with the procedure used, with 
    appropriate validation information provided. Representative 
    chromatograms should be provided. Chromatograms of such 
    representative batches, from methods validation studies showing 
    separation and detectability of impurities (e.g., on spiked 
    samples), along with any other impurity tests routinely performed, 
    can serve as the representative impurity profiles. The applicant 
    should ensure that complete impurity profiles (i.e., chromatograms) 
    of individual batches are available if requested. A tabulation 
    should be provided which links the specific new drug substance batch 
    to each safety study and each clinical study in which it has been 
    used.
        For each batch of the new drug substance, the report should 
    include:
         Batch Identity and Size
         Date of Manufacture
         Site of Manufacture
         Manufacturing Process
         Impurity Content, Individual and Total
        
    [[Page 374]]
    
         Use of Batches
         Reference to Analytical Procedure Used
    
    6. Specification Limits for Impurities
    
         The specifications for a new drug substance should include 
    limits for impurities. Stability studies, chemical development 
    studies, and routine batch analyses can be used to predict those 
    impurities likely to occur in the commercial product. The selection 
    of impurities to include in the new drug substance specifications 
    should be based on the impurities found in batches manufactured by 
    the proposed commercial process. Those impurities selected for 
    inclusion in the specifications for the new drug substance are 
    referred to as ``specified impurities'' in this guideline. Specified 
    impurities may be identified or unidentified and should be 
    individually listed in the new drug substance specifications.
         A rationale for the inclusion or exclusion of impurities in the 
    specifications should be presented. This rationale should include a 
    discussion of the impurity profiles observed in the safety and 
    clinical development batches, together with a consideration of the 
    impurity profile of material manufactured by the proposed commercial 
    process. Specific identified impurities should be included along 
    with recurring unidentified impurities estimated to be at or above 
    0.1 percent. For impurities known to be unusually potent or to 
    produce toxic or unexpected pharmacological effects, the 
    quantitation/detection limit of the analytical methods should be 
    commensurate with the level at which the impurities must be 
    controlled. For unidentified impurities, the procedure used and 
    assumptions made in establishing the level of the impurity should be 
    clearly stated. Unidentified impurities included in the 
    specifications should be referred to by some appropriate qualitative 
    analytical descriptive label (e.g., ``unidentified A,'' 
    ``unidentified with relative retention of 0.9''). Finally, a general 
    specification limit of not more than 0.1 percent for any unspecified 
    impurity should be included.
        Limits should be set no higher than the level that can be 
    justified by safety data, and, unless safety data indicate 
    otherwise, no lower than the level achievable by the manufacturing 
    process and the analytical capability. In other words, where there 
    is no safety concern, impurity specifications should be based on 
    data generated on actual batches of the new drug substance allowing 
    sufficient latitude to deal with normal manufacturing and analytical 
    variation, and the stability characteristics of the new drug 
    substance. Although normal manufacturing variations are expected, 
    significant variation in batch-to-batch impurity levels may indicate 
    that the manufacturing process of the new drug substance is not 
    adequately controlled and validated.
        In summary, the new drug substance specifications should 
    include, where applicable, limits for:
         Organic Impurities:
          Each Specified Identified Impurity
          Each Specified Unidentified Impurity at or above 0.1 
    percent
          Any Unspecified Impurity, with a limit of not more 
    than 0.1 percent
          Total Impurities
         Residual Solvents
         Inorganic Impurities
        A summation of assay value and impurity levels generally may be 
    used to obtain mass balance for the test sample. The mass balance 
    need not add to exactly 100 percent because of the analytical error 
    associated with each analytical procedure. The summation of impurity 
    levels plus the assay value may be misleading, for example, when the 
    assay procedure is nonspecific (e.g., potentiometric titrimetry) and 
    the impurity level is relatively high.
    
    7. Qualification of Impurities
    
        Qualification is the process of acquiring and evaluating data 
    which establishes the biological safety of an individual impurity or 
    a given impurity profile at the level(s) specified. The applicant 
    should provide a rationale for selecting impurity limits based on 
    safety considerations. The level of any impurity present in a new 
    drug substance that has been adequately tested in safety and/or 
    clinical studies is considered qualified. Impurities that are also 
    significant metabolites present in animal and/or human studies do 
    not need further qualification. A level of a qualified impurity 
    higher than that present in a new drug substance can also be 
    justified based on an analysis of the actual amount of impurity 
    administered in previous safety studies.
        If data are not available to qualify the proposed specification 
    level of an impurity, studies to obtain such data may be needed when 
    the usual qualification threshold limits given below are exceeded:
    
                                                                            
    ------------------------------------------------------------------------
                 Maximum daily dose                Qualification threshold  
    ------------------------------------------------------------------------
     2 grams (g)/day...............  0.1 percent or 1 milligram  
                                                 per day intake (whichever  
                                                 is lower)                  
    > 2 g/day.................................  0.05 percent                
    ------------------------------------------------------------------------
    
         Higher or lower threshold limits for qualification of 
    impurities may be appropriate for some individual drugs based on 
    scientific rationale and level of concern, including drug class 
    effects and clinical experience. For example, qualification may be 
    especially important when there is evidence that such impurities in 
    certain drugs or therapeutic classes have previously been associated 
    with adverse reactions in patients. In these instances, a lower 
    qualification threshold limit may be appropriate. Conversely, a 
    higher qualification threshold limit may be appropriate for 
    individual drugs when the level of concern for safety is less than 
    usual based on similar considerations (e.g., patient population, 
    drug class effects, clinical considerations). Technical factors 
    (manufacturing capability and control methodology) may be considered 
    as part of the justification for selection of alternative threshold 
    limits. Proposals for alternative threshold limits are considered on 
    a case-by-case basis.
        The ``Decision Tree for Safety Studies'' (Attachment I) 
    describes considerations for the qualification of impurities when 
    thresholds are exceeded. In some cases, decreasing the level of 
    impurity below the threshold may be simpler than providing safety 
    data. Alternatively, adequate data may be available in the 
    scientific literature to qualify an impurity. If neither is the 
    case, additional safety testing should be considered. The studies 
    desired to qualify an impurity will depend on a number of factors, 
    including the patient population, daily dose, route, and duration of 
    drug administration. Such studies are normally conducted on the new 
    drug substance containing the impurities to be controlled, although 
    studies using isolated impurities are seen as acceptable.
    
    BILLING CODE 4160-01-F
    
    [[Page 375]]
    [GRAPHIC][TIFF OMITTED]TN04JA96.025
    
    
    
    BILLING CODE 4160-01-C
    
    [[Page 376]]
    
        a If considered desirable, a minimum screen for genotoxic 
    potential should be conducted. A study to detect point mutations and 
    one to detect chromosomal aberrations, both in vitro, are seen as an 
    acceptable minimum screen.
        b If general toxicity studies are desirable, study(ies) 
    should be designed to allow comparison of unqualified to qualified 
    material. The study duration should be based on available relevant 
    information and performed in the species most likely to maximize the 
    potential to detect the toxicity of an impurity. In general, a 
    minimum duration of 14 days and a maximum duration of 90 days are 
    seen as acceptable.
    
    8. New Impurities
    
        During the course of a drug development program, the qualitative 
    impurity profile of the new drug substance may change, or a new 
    impurity may appear as a result of, for example, synthetic route 
    changes, process optimization, or scale-up. New impurities may be 
    identified or unidentified. Such changes call for consideration of 
    the need for qualification of the level of the impurity, unless it 
    is below the threshold values as noted above. When a new impurity 
    exceeds the threshold, the ``Decision Tree for Safety Studies'' 
    should be consulted. Safety studies should compare the new drug 
    substance containing a representative level of the new impurity with 
    previously qualified material, although studies using the isolated 
    impurity are also seen as acceptable (these studies may not always 
    have clinical relevance).
    
    9. Glossary
    
        Chemical Development Studies: Studies conducted to scale-up, 
    optimize, and validate the manufacturing process for a new drug 
    substance.
        Enantiomers: Compounds with the same molecular formula as the 
    drug substance, which differ in the spatial arrangement of atoms 
    within the molecule and are nonsuperimposable mirror images.
        Extraneous Substance: An impurity arising from any source 
    extraneous to the manufacturing process.
        Herbal Products: Medicinal products containing, exclusively, 
    plant material and/or vegetable drug preparations as active 
    ingredients. In some traditions, materials of inorganic or animal 
    origin may also be present.
        Identified Impurity: An impurity for which a structural 
    characterization has been achieved.
        Impurity: Any component of the new drug substance which is not 
    the chemical entity defined as the new drug substance.
        Impurity Profile: A description of the identified and 
    unidentified impurities present in a new drug substance.
        Intermediate: A material produced during steps of the synthesis 
    of a new drug substance which must undergo further molecular change 
    before it becomes a new drug substance.
        Ligand: An agent with a strong affinity to a metal ion.
        New Drug Substance: The designated therapeutic moiety which has 
    not been previously registered in a region or member state (also 
    referred to as a new molecular entity or new chemical entity). It 
    may be a complex, simple ester, or salt of a previously approved 
    drug substance.
        Polymorphism: The occurrence of different crystalline forms of 
    the same drug substance.
        Potential Impurity: An impurity which, from theoretical 
    considerations, may arise from or during manufacture. It may or may 
    not actually appear in the new drug substance.
        Qualification: The process of acquiring and evaluating data 
    which establishes the biological safety of an individual impurity or 
    a given impurity profile at the level(s) specified.
        Reagent: A substance, other than a starting material or solvent, 
    which is used in the manufacture of a new drug substance.
        Safety Information: The body of information that establishes the 
    biological safety of an individual impurity or a given impurity 
    profile at the level(s) specified.
        Solvent: An inorganic or an organic liquid used as a vehicle for 
    the preparation of solutions or suspensions in the synthesis of a 
    new drug substance.
        Specified Impurity: Identified or unidentified impurity that is 
    selected for inclusion in the new drug substance specifications and 
    is individually listed and limited in order to assure the safety and 
    quality of the new drug substance.
        Starting Material: A material used in the synthesis of a new 
    drug substance which is incorporated as an element into the 
    structure of an intermediate and/or of the new drug substance. 
    Starting materials normally are commercially available and of 
    defined chemical and physical properties and structure.
        Toxic Impurity: Impurities having significant undesirable 
    biological activity.
        Unidentified Impurity: An impurity which is defined solely by 
    qualitative analytical properties (e.g., chromatographic retention 
    time).
        Validated Limit of Quantitation: For impurities at a level of 
    0.1 percent, the validated limit of quantitation should be less than 
    or equal to 0.05 percent. Impurities limited at higher levels may 
    have higher limits of quantitation.
    
        Dated: December 21, 1995.
    William K. Hubbard,
    Associate Commissioner for Policy Coordination.
    [FR Doc. 96-64 Filed 1-3-96; 8:45 am]
    BILLING CODE 4160-01-F
    
    

Document Information

Effective Date:
1/4/1996
Published:
01/04/1996
Department:
Food and Drug Administration
Entry Type:
Notice
Action:
Notice.
Document Number:
96-64
Dates:
Effective January 4, 1996. Submit written comments at any time.
Pages:
372-376 (5 pages)
Docket Numbers:
Docket No. 94D-0325
PDF File:
96-64.pdf