94-23378. International Conference on Harmonisation; Draft Guideline on Impurities in New Drug Substances; Availability; Notice DEPARTMENT OF HEALTH AND HUMAN SERVICES  

  • [Federal Register Volume 59, Number 183 (Thursday, September 22, 1994)]
    [Unknown Section]
    [Page 0]
    From the Federal Register Online via the Government Publishing Office [www.gpo.gov]
    [FR Doc No: 94-23378]
    
    
    [[Page Unknown]]
    
    [Federal Register: September 22, 1994]
    
    
    _______________________________________________________________________
    
    Part VIII
    
    
    
    
    
    Department of Health and Human Services
    
    
    
    
    
    _______________________________________________________________________
    
    
    
    Food and Drug Administration
    
    
    
    _______________________________________________________________________
    
    
    
    
    International Conference on Harmonisation; Draft Guideline on 
    Impurities in New Drug Substances; Availability; Notice
    DEPARTMENT OF HEALTH AND HUMAN SERVICES
    
    Food and Drug Administration
    -----------------------------------------------------------------------
    [Docket No. 94D-0325]
    
     
    International Conference on Harmonisation; Draft 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 draft 
    guideline entitled ``Impurities in New Drug Substances.'' The draft 
    guideline was prepared by the Quality Expert Working Group of the 
    International Conference on Harmonisation of Technical Requirements for 
    Registration of Pharmaceuticals for Human Use (ICH). The draft 
    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: Written comments by December 6, 1994.
    
    ADDRESSES: Submit written comments on the draft 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 CDER Executive Secretariat Staff (HFD-8), Center 
    for Drug Evaluation and Research, Food and Drug Administration, 7500 
    Standish Pl., Rockville, MD 20855.
    
    FOR FURTHER INFORMATION CONTACT:
        Regarding the draft guideline: Charles S. Kumkumian, Center for 
    Drug Evaluation and Research (HFD-102), Food and Drug Administration, 
    5600 Fishers Lane, Rockville, MD 20857, 301-594-6758.
        Regarding the ICH: Janet Showalter, Office of Health Affairs (HFY-
    50), Food and Drug Administration, 5600 Fishers Lane, Rockville, MD 
    20857, 301-443-1382.
    
    SUPPLEMENTARY INFORMATION: In recent years, many important initiatives 
    have been undertaken by regulatory authorities and industry 
    associations to promote Harmonisation of regulatory requirements. FDA 
    has participated in many meetings designed to enhance Harmonisation and 
    is committed to seeking scientifically based harmonized technical 
    procedures for pharmaceutical development. One of the goals of 
    Harmonisation is to identify and then reduce differences in technical 
    requirements for drug development.
        ICH was organized to provide an opportunity for tripartite 
    Harmonisation 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 
    Harmonisation 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 Industry 
    Associations, the Japanese Ministry of Health and Welfare, the Japanese 
    Pharmaceutical Manufacturers Association, 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 IFPMA, as well as observers from the World Health 
    Organization, the Canadian Health Protection Branch, and the European 
    Free Trade Area.
        On March 15, 1994, the ICH Steering Committee agreed that a 
    consensus draft guideline entitled ``Impurities In New Drug 
    Substances,'' which was developed by the Quality Expert Working Group 
    of the ICH, should be made available for public comment. The draft 
    guideline will be made available for comment by the European Commission 
    and Japanese Ministry of Health and Welfare, as well as by FDA, in 
    accordance with their respective consultation procedures. Comments 
    about this draft will be considered by FDA and the Expert Working 
    Group. Ultimately, FDA intends to adopt the ICH Steering Committee's 
    final guideline.
        The draft 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 
    draft guideline is not intended to apply to new drug substances used 
    during the clinical research stage of development or clinical trials. 
    The draft guideline also does not apply to biological/biotechnological 
    substances, peptides, 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 draft 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, if 
    the agency issues this guideline in final form, the guideline would not 
    be issued under the authority of current Sec. 10.90(b), and would not 
    create or confer any rights, privileges, or benefits for or on any 
    person, nor would it operate to bind FDA in any way.
        Interested persons may, on or before December 6, 1994, to the 
    Dockets Management Branch (address above) submit written comments on 
    the draft 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 draft 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 draft 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 stage of development, e.g., investigational new drugs or 
    clinical trials exemptions. Biological/biotechnological, peptide, 
    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 
    described below which qualification should not be necessary.
    
    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 need 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 arising 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, identification 
    of all recurring impurities at or above the 0.1 percent level is 
    expected in batches manufactured by the proposed commercial process. 
    Degradation products observed in stability studies at labelled 
    storage temperatures 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 also 
    to 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 
    guideline. 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 are normally 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 are based on pharmacopeial standards of 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 are 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 use. It is 
    considered acceptable to use the drug substance 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 are often 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 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
        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 these guidelines. 
    Specified impurities may be identified or unidentified and are 
    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 sensitivity 
    of the analytical methods should be commensurate with the level at 
    which the impurities are to 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,'' 
    etc.). 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 which 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 are 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:
        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 can 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, e.g., 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 which has been adequately tested in safety and/or 
    clinical studies is considered qualified. Impurities which 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 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 (patient population, drug 
    class effects, clinical considerations, etc.). 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 acceptable.
    
    BILLING CODE 4160-01-F
    
    TN22SE94.000
    
    BILLING CODE 4160-01-C
    
         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 will 
    be 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 synthetic route changes, process 
    optimization, scale-up, etc. 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 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.
        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.
        Manufacture: All operations related to the new drug substance, 
    including purchase of materials, production, quality control, 
    release, storage, and distribution of the new drug substance.
        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 are normally 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: September 15, 1994.
    William K. Hubbard,
    Interim Deputy Commissioner for Policy.
    [FR Doc. 94-23378 Filed 9-21-94; 8:45 am]
    BILLING CODE 4160-01-F
    
    
    

Document Information

Published:
09/22/1994
Entry Type:
Uncategorized Document
Action:
Notice.
Document Number:
94-23378
Dates:
Written comments by December 6, 1994.
Pages:
0-0 (1 pages)
Docket Numbers:
Federal Register: September 22, 1994