95-20608. International Conference on Harmonisation; Draft Guideline on Stability Testing of Biotechnological/Biological Products; Availability  

  • [Federal Register Volume 60, Number 161 (Monday, August 21, 1995)]
    [Notices]
    [Pages 43501-43505]
    From the Federal Register Online via the Government Publishing Office [www.gpo.gov]
    [FR Doc No: 95-20608]
    
    
    
    -----------------------------------------------------------------------
    [Docket No. 93D-0139]
    
    
    International Conference on Harmonisation; Draft Guideline on 
    Stability Testing of Biotechnological/Biological Products; Availability
    
    AGENCY: Food and Drug Administration, HHS.
    
    ACTION: Notice.
    
    -----------------------------------------------------------------------
    
    SUMMARY: The Food and Drug Administration (FDA) is publishing a draft 
    guideline entitled ``Quality of Biotechnological Products: Stability 
    Testing of Biotechnological/Biological Products.'' This draft guideline 
    was prepared under the auspices of the International Conference on 
    Harmonisation of Technical Requirements for Registration of 
    Pharmaceuticals for Human Use (ICH). The draft guideline is intended to 
    give guidance to applicants regarding the type of stability studies 
    that should be provided in support of marketing applications for 
    biotechnological/biological products.
    
    DATES: Written comments by October 5, 1995.
    
    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 draft 
    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, as well as the 
    CBER Congressional and Consumer Affairs Branch (HFM-12), Center for 
    Biologics Evaluation and Research, 
    
    [[Page 43502]]
    Food and Drug Administration, 1401 Rockville Pike, Rockville, MD 20852.
    
    FOR FURTHER INFORMATION CONTACT: 
        Regarding the guideline: Kenneth Seamon, Center for Biologics 
    Evaluation and Research (HFM-20), Food and Drug Administration, 1401 
    Rockville Pike, Rockville, MD 20852, 301-827-0375.
        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.
        At a meeting held on March 29, 1995, the ICH Steering Committee 
    agreed that a draft guideline entitled ``Quality of Biotechnological 
    Products: Stability Testing of Biotechnological/Biological Products'' 
    should be made available for public comment. The draft guideline is the 
    product of the Quality Expert Working Group of the ICH. 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.
        This draft guideline is intended to supplement the tripartite ICH 
    guideline entitled ``Stability Testing of New Drug Substances and 
    Products,'' published in the Federal Register of September 22, 1994 (59 
    FR 48754). Biotechnological/biological products have distinguishing 
    characteristics to which consideration should be given in any well-
    defined testing program designed to confirm their stability during the 
    intended storage period. For such products, in which the active 
    components are typically proteins and/or polypeptides, maintenance of 
    molecular conformation and biological activity is dependent on 
    noncovalent as well as covalent forces. The products are particularly 
    sensitive to environmental factors such as temperature changes, 
    oxidation, light, ionic content, shear, etc. In order to ensure 
    maintenance of biological activity and to avoid degradation, stringent 
    conditions for their storage are usually necessary. This draft 
    guideline is intended to assist the applicant in developing appropriate 
    supporting stability data for a biotechnological/biological product.
        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 are acceptable to FDA. The agency 
    is now in the process of revising Sec. 10.90(b). Therefore, this 
    guideline is not being issued under the authority of Sec. 10.90(b), and 
    it does not create or confer any rights, privileges, or benefits for or 
    on any person, nor does it operate to bind FDA in any way.
        Interested persons may, on or before October 5, 1995, submit 
    written comments on the draft guideline to the Dockets Management 
    Branch (address above). 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:
    
    Quality of Biotechnological Products: Stability Testing of 
    Biotechnological/Biological Products (Q5C)
    
    ICH Expert Working Group on Quality of Biotechnology Products
    
    Annex to the Tripartite ICH Guideline for the Stability Testing of New 
    Drug Substances and Products
    
    Introduction
    
         The principles established in the ICH harmonized tripartite 
    guideline ``Stability Testing of New Drug Substances and Products'' 
    (27 October 1993) apply in general to biotechnological/biological 
    products. However, biotechnological/biological products do have 
    distinguishing characteristics to which consideration should be 
    given in any well-defined testing program designed to confirm their 
    stability during the intended storage period. For such products, in 
    which the active components are typically proteins and/or 
    polypeptides, maintenance of molecular conformation and, hence of 
    biological activity, is dependent on noncovalent as well as covalent 
    forces. The products are particularly sensitive to environmental 
    factors such as temperature changes, oxidation, light, ionic 
    content, shear, etc. In order to ensure maintenance of biological 
    activity and to avoid degradation, stringent conditions for their 
    storage are usually necessary.
        The evaluation of stability may necessitate complex analytical 
    methodologies. Assays for biological activity, where applicable, are 
    an essential part of the pivotal stability studies. Appropriate 
    physicochemical, biochemical, and immunochemical methods for the 
    analysis of the molecular entity and the quantitative detection of 
    degradation products should also be part of the stability program 
    whenever purity and molecular characteristics of the product permit 
    use of these methodologies.
        With the above concerns in mind, the applicant should develop 
    the proper supporting stability data for a biotechnological/
    biological product and consider many external conditions which can 
    affect the product's potency, purity, and quality. Primary data to 
    support a requested storage period for either drug substance or drug 
    product should be based on long-term, real-time, real-condition 
    stability studies. Thus, the development of a proper long-term 
    stability program becomes critical to the successful development of 
    a commercial product. The purpose of this document is to give 
    guidance to applicants regarding the type of stability studies that 
    should be provided in support of marketing applications. It is 
    understood that during the review and evaluation process, continuing 
    updates of initial stability data may occur.
    
    Scope of the Annex
    
        The principles adopted and explained in this annex apply to 
    well-characterized proteins and polypeptides, their derivatives and 
    products of which they are components, and which are isolated from 
    tissues, body fluids, cell cultures, or produced using rDNA 
    technology. Thus, the document covers the 
    
    [[Page 43503]]
    generation and submission of stability data for products such as 
    cytokines (interferons, interleukins, colony-stimulating factors, 
    tumor necrosis factors), erythropoietins, plasminogen activators, 
    blood plasma factors, growth hormones and growth factors, insulins, 
    monoclonal antibodies, and vaccines consisting of well-characterized 
    proteins or polypeptides. In addition, the principles outlined in 
    the following sections may apply to other types of products, such as 
    conventional vaccines, after consultation with the appropriate 
    regulatory authorities.
        The document does not cover antibiotics, allergenic extracts, 
    heparins, vitamins, or whole blood.
    
    Terminology
    
         For the basic terms used in this annex the reader is referred 
    to the ``Glossary'' in the ICH harmonized tripartite guideline 
    ``Stability Testing of New Drug Substances and Products'' (27 
    October 1993). However, since traditional terminology used by 
    manufacturers of biotechnological/biological products does not 
    always conform to that of the tripartite guideline mentioned above, 
    traditional terms are specified in brackets to assist the reader. A 
    supplemental glossary is also included that defines certain of the 
    traditional terms used in the biologics field.
    
    Selection of Batches
    
    Drug Substance (Bulk Material)
    
        Where bulk material is to be stored after manufacture but prior 
    to formulation and final manufacturing, stability data should be 
    provided on at least three batches for which manufacture and storage 
    are representative of the manufacturing scale of production. A 
    minimum of 6 months stability data at the time of submission should 
    be submitted in cases where storage periods greater than 6 months 
    are requested. For drug substances with storage periods of less than 
    6 months, the minimum amount of stability data in the initial 
    submission will be determined on a case-by-case basis. Data from 
    pilot-plant-scale batches of a well-characterized drug substance 
    (bulk material) produced at a reduced scale of fermentation and 
    purification may be provided at the time the dossier is submitted to 
    the regulatory agencies with a commitment to place the first three 
    full-scale batches into the long-term stability program after 
    approval.
        The quality of the batches of drug substance placed into the 
    stability program should be representative of the quality of the 
    material used in preclinical and clinical studies and of the quality 
    of the material to be made at manufacturing scale. In addition, the 
    drug substance (bulk material) made at pilot-plant scale should be 
    produced by a process and stored under conditions representative of 
    that used for the manufacturing scale. The drug substance entered 
    into the stability program should be stored in containers which 
    properly represent the actual holding containers used during 
    manufacture. Scaled-down containers may be acceptable for drug 
    substance stability monitoring assuming that they are constructed of 
    the same material and make use of the same type of container/closure 
    system that is routinely used during the manufacture.
    
    Intermediates
    
        During manufacture of biotechnological/biological products, the 
    quality and control of certain intermediates may be critical to the 
    production of the final product. In general, the manufacturer should 
    identify intermediates and generate in-house data and process limits 
    that assure their stability within the bounds of the developed 
    process. While the use of pilot-plant-scale data is permissible, the 
    manufacturer should establish the suitability of such data using the 
    manufacturing-scale process.
    
    Drug Product (Final Container Product)
    
        Stability information should be provided on at least three 
    batches of final container product representative of that which will 
    be used at manufacturing scale. Where possible, batches of final 
    container product included in stability testing should be derived 
    from different batches of bulk material. A minimum of 6-months data 
    at the time of submission should be submitted in cases where storage 
    periods greater than 6 months are requested. For drug products with 
    storage periods of less than 6 months, the minimum amount of 
    stability data in the initial submission will be reviewed on a case-
    by-case basis. Product expiration dating will be based upon the 
    actual data submitted in support of the application. Since dating is 
    based upon the real-time/real-temperature data submitted for review, 
    it is expected that continuing updates of initial stability data 
    will occur during the review and evaluation process. Where pilot-
    scale batches were submitted to establish the dating for a product 
    and, in the event that product produced at manufacturing scale does 
    not meet those long-term stability specifications throughout the 
    dating period or is not representative of the material used in 
    preclinical and clinical studies, the sponsor/applicant should 
    notify the appropriate regulatory authorities to determine a 
    suitable course of action.
    
    Sample Selection Criteria
    
        Where one product is distributed in batches differing in fill 
    volume (e.g., 1 milliliter (mL), 2 mL, or 10 mL), unitage (e.g., 10 
    units, 20 units, or 50 units), or mass (e.g., 1 milligram (mg), 2 
    mg, or 5 mg) samples to be entered into the stability program may be 
    selected on the basis of a matrix system and/or by bracketing.
        Matrixing, i.e., the statistical design of a stability study in 
    which different fractions of samples are tested at different 
    sampling points, should only be applied when appropriate 
    documentation is provided that confirms that the stability of the 
    samples tested represents the stability of all samples. The 
    differences in the samples for the same drug product should be 
    identified as, for example, covering different batches, different 
    strengths, different sizes of the same closure and possibly, in some 
    cases, different container/closure systems. Matrixing should not be 
    applied to samples with differences that may affect stability, such 
    as different strengths and different containers/closures, where it 
    cannot be confirmed that the products respond similarly under 
    storage conditions.
        Where the same strength and exact container/closure system is 
    used for three or more fill contents, the applicant may elect to 
    place only the smallest and largest container size into the 
    stability program, i.e., bracketing. The design of a protocol that 
    incorporates bracketing assumes that the stability of the 
    intermediate condition samples are represented by those at the 
    extremes. In certain cases, it may be necessary to provide data that 
    demonstrate that all samples are properly represented by data 
    collected for the extremes.
    
    Stability-Indicating Profile
    
        On the whole, there is no single stability-indicating assay or 
    parameter that profiles the stability characteristics of a 
    biotechnological/biological product. Consequently, the manufacturer 
    should propose a stability-indicating profile that provides 
    assurance that changes in the identity, purity, and potency of the 
    product will be detected.
        It is also expected that, at the time of submission, applicant/
    firms have validated the methods that comprise the stability-
    indicating profile and that the data are available for review. The 
    determination of which tests should be included will be product 
    specific. The items emphasized in the following subsections are not 
    intended to be all inclusive, but represent product characteristics 
    that should typically be documented to demonstrate product stability 
    adequately.
    
    Protocol
    
         The dossier accompanying the application for marketing 
    authorization should include a detailed protocol for the assessment 
    of the stability of both drug substance and drug product in support 
    of the claimed storage conditions and expiration dating periods. The 
    protocol should include all necessary information, including well-
    defined specifications, test intervals, etc., which taken as a 
    whole, demonstrates the stability of the biotechnological/biological 
    product throughout the claimed expiration dating period. The 
    statistical methods to be used are described in the tripartite 
    guideline. It is assumed that the manufacturer of the product will 
    strictly adhere to this protocol.
    
    Potency
    
        Wherever the intended use of a product is linked to a definable 
    and measurable biological activity, testing for potency should be 
    part of the stability studies. Potency studies should be performed 
    at appropriate intervals as defined in the stability protocol and 
    the results should be reported in units of biological activity 
    calibrated, whenever possible, against nationally or internationally 
    recognized standards. Where no national or international agreement 
    has been reached on units of potency, the assay results may be 
    reported in in-house derived units using an appropriately 
    characterized reference preparation.
        In some biotechnological/biological products, potency is 
    dependent upon the conjugation of the active ingredient(s) to a 
    second moiety or binding to an adjuvant. Dissociation of the active 
    ingredient(s) from the carrier used in conjugates or adjuvants 
    
    [[Page 43504]]
    should be examined in real-time/real-temperature studies (including 
    conditions encountered during shipment). The assessment of the 
    stability of such products may be associated with difficulties 
    since, in some cases, in vitro tests for biological activity and 
    physicochemical characterisation are impractical or provide 
    inaccurate results. Appropriate strategies (e.g., testing the 
    product prior to conjugation/binding, release of the active compound 
    from the second moiety, in vivo assays, etc.) or the use of an 
    appropriate surrogate test should be considered to overcome the 
    inadequacies of in vitro testing.
    
    Purity and Molecular Characterization
    
        The degree of purity, as well as individual and total upper 
    limits for degradation products of the biotechnological/biological 
    product entered into the stability studies, should be reported and 
    documented whenever possible. Limits of acceptable degradation 
    should be derived from the analytical profiles of batches of the 
    drug substance and drug product used in the preclinical and clinical 
    studies.
        For physicochemically well-defined drug substances and/or drug 
    products, the use of relevant physicochemical, biochemical, and 
    immunochemical analytical methodologies should permit a 
    comprehensive characterisation of the active ingredient (e.g., 
    molecular size, charge, hydrophobicity, etc.) and the accurate 
    detection of degradation changes that may result from deamidation, 
    oxidation, sulfoxidation, aggregation, or fragmentation during 
    storage. As examples, methods that may contribute to this include 
    electrophoresis (SDS-PAGE, immunoelectrophoresis, Western blot, 
    isoelectrofocusing), high-resolution chromatography (reversed-phase 
    chromatography, gel filtration, ion exchange, affinity 
    chromatography, etc.), and peptide mapping.
        Wherever significant qualitative or quantitative changes 
    indicative of degradation product formation are detected during 
    long-term, accelerated, and/or stress stability studies, 
    consideration should be given to potential hazards and to the need 
    for characterization and quantification of degradation products 
    within the long-term stability program. Acceptable limits should be 
    proposed and justified, taking into account the levels observed in 
    material used in preclinical and clinical studies.
        For substances that cannot be properly characterized or products 
    for which an exact analysis of the purity level cannot be 
    meaningfully determined through routine analytical methods, the 
    applicant should propose and justify alternative testing procedures.
    
    Other Product Characteristics
    
        The following product characteristics, though not specifically 
    relating to biotechnological/biological products, should be 
    monitored and reported for the drug product in its final container:
        Visual appearance of the product (colour and opacity for 
    solutions/suspensions; colour, texture, and dissolution time for 
    powders), visible particulates in solutions or after the 
    reconstitution of powders or lyophilized cakes, pH, and moisture 
    level of powders and lyophilized products.
        Sterility testing or alternatives (e.g., container/closure 
    integrity testing) should be performed at a minimum initially and at 
    the end of the proposed shelf-life.
        Additives (stabilizers, preservatives, etc.) or excipients may 
    degrade during the dating period of the drug product. If there is 
    any indication during preliminary stability studies that reaction or 
    degradation of such materials adversely affect the quality of the 
    drug product, these items may need to be monitored during the 
    stability program.
        The container/closure has the potential to adversely affect the 
    product and should be carefully evaluated. Closure configurations, 
    vial liners seal-types should also be considered (see below).
    
    Storage Conditions
    
    Temperature
    
         Since most finished biotechnological/biological products need 
    precisely defined storage temperatures, the storage conditions for 
    the real-time/real-temperature stability studies may be confined to 
    the recommended storage temperature.
    
    Humidity
    
         Biotechnological/biological products are generally distributed 
    in containers protecting them against humidity. Therefore, where it 
    can be demonstrated that the proposed containers (and conditions of 
    storage) afford sufficient protection against high and low humidity, 
    stability tests at different relative humidities can usually be 
    omitted. Where humidity-protecting containers are not used, 
    appropriate stability data should be provided.
    
    Accelerated and Stress Conditions
    
        As previously noted, the expiration dating generally is based 
    upon the real-time/real-temperature data. However, it is strongly 
    suggested that studies be conducted on the drug substance and drug 
    product under accelerated and stress conditions. Studies under 
    accelerated conditions may provide useful support data for 
    establishing the expiration date, provide product stability 
    information for future product development (e.g., preliminary 
    assessment of proposed manufacturing changes such as change in 
    formulation, scale-up, etc.), assist in validation of analytical 
    methods for the stability program, or generate information which may 
    help elucidate the degradation profile of the drug substance or drug 
    product. Studies under stress conditions may be useful in 
    determining whether accidental exposures to conditions other than 
    those recommended (e.g., during transportation) are deleterious to 
    the product and also for evaluating which specific test parameters 
    may be the best indicators of product stability. Studies of the 
    exposure of the drug substance or drug product to extreme conditions 
    may help to reveal patterns of degradation; if so, such changes 
    should be monitored under recommended storage conditions. While the 
    tripartite guideline describes the conditions of the accelerated and 
    stress study, the applicant should note that those conditions may 
    not be appropriate for biotechnological/biological products. 
    Conditions should be carefully selected on a case-by-case basis.
    
    Light
    
         Applicants should consult the appropriate regulatory 
    authorities on a case-by-case basis to determine guidance for 
    testing.
    
    Container/Closure
    
        Changes in the quality of the product may occur due to the 
    interactions between the formulated biotechnological/biological 
    product and container/closure. Where the lack of interactions cannot 
    be excluded in liquid products (other than sealed ampules), 
    stability studies should include samples maintained in the inverted 
    or horizontal position (i.e., in contact with the closure), as well 
    as in the upright position, to determine the effects of the closure 
    on product quality. Data should be supplied for all different 
    container/closure combinations that will be marketed.
         In addition to the standard data necessary for a conventional 
    single-use vial, the applicant should demonstrate that the closure 
    used with a multiple-dose vial is capable of withstanding the 
    conditions of repeated insertions and withdrawals so that the 
    product retains its full potency, purity, and quality for the 
    maximum period specified in the instructions-for-use on containers, 
    packages, and/or package inserts. Such labeling should be in 
    accordance with relevant national/regional requirements.
    
    Stability After Reconstitution of Freeze-Dried Product
    
         The stability of freeze-dried products after their 
    reconstitution should be demonstrated for the conditions and the 
    maximum storage period specified on containers, packages, and/or 
    package inserts. Such labeling should be in accordance with relevant 
    national/regional requirements.
    
    Testing Frequency
    
        The shelf-lives of biotechnological/biological products may vary 
    from days to several years. Thus, it is difficult to draft uniform 
    guidelines regarding the stability study duration and testing 
    frequency that would be applicable to all types of biotechnological/
    biological products. With only a few exceptions, however, the shelf-
    lives for existing products and potential future products will be 
    within the range of 0.5 to 5 years. Therefore, the recommendations 
    that follow are based upon expected shelf-lives in that range, and 
    take account of the fact that, frequently, degradation of 
    biotechnological/biological products is not governed by the same 
    factors during different intervals of a long storage period.
         When shelf lives of 1 year or less are proposed, the real-time 
    stability studies generally should be conducted monthly for the 
    first 3 months and at 3-month intervals thereafter.
         For products with proposed shelf-lives of greater than 1 year, 
    the studies should be conducted every 3 months during the first year 
    of storage, every 6 months during the second year, and annually 
    thereafter.
    
    [[Page 43505]]
    
         While the testing intervals listed above are appropriate in the 
    preapproval or prelicense stage, reduced testing may be appropriate 
    after approval or licensure where data are available that 
    demonstrate adequate stability. Where data exist that indicate the 
    stability of a product is not compromised, the applicant is 
    encouraged to submit a protocol that supports elimination of 
    specific test intervals (e.g., 9-month testing) for postapproval/
    postlicensure, long-term studies.
    
    Specifications
    
         Although biotechnological/biological products may be subject to 
    significant losses of activity and to physicochemical degradation 
    during storage, international and national regulations have provided 
    little guidance with respect to distinct release and end-of-shelf-
    life specifications. Recommendations for maximum acceptable losses 
    of activity or limits for physicochemical changes (degradation) 
    during the proposed shelf-life have not been developed for 
    individual types or groups of biotechnological/biological products 
    but are considered on a case-by-case basis. Each product should 
    retain its specifications within established limits for safety, 
    purity, and potency throughout its proposed shelf-life. These 
    specifications and limits should be derived from all available 
    information using the appropriate statistical methods. The use of 
    different specifications for release and expiration should be 
    supported by sufficient data to demonstrate that clinical 
    performance is not affected. The proposals should be in accordance 
    with the principles outlined in the appropriate section of the 
    tripartite guideline.
    
    Labeling
    
        For most biotechnological/biological drug substances and drug 
    products, precisely defined storage temperatures are recommended. 
    Specific recommendations should be stated, particularly for drug 
    substances and drug products that cannot tolerate freezing. These 
    conditions, and where appropriate, recommendations for protection 
    against light and/or humidity, should appear on containers, 
    packages, and/or package inserts. Such labeling should be in 
    accordance with relevant national/regional requirements.
    
    Glossary
    
    Conjugated Product
    
         A conjugated product is made up of an active ingredient 
    (peptide, carbohydrate, etc.) bound covalently or noncovalently to a 
    carrier (protein, peptide, inorganic mineral, etc.) with the 
    objective of improving the efficacy or stability of the product.
    
    Degradation Product
    
        Any material resulting from modification of the active 
    ingredients, additives, and/or excipients present in a drug 
    substance or drug product which occurs due to processing or storage 
    (e.g., by deamidation, oxidation, aggregation, proteolysis, etc.). 
    Degradation products are considered impurities, although some 
    degradation products may be active.
    
    Impurity
    
        Any process-generated substance present in raw materials, drug 
    substance, or drug product that is not considered to be active 
    ingredient, additives, or excipients.
    
    Intermediate
    
        A material produced during a manufacturing process that is not 
    the drug substance or the drug product but whose manufacture is 
    critical to the successful production of the drug substance or the 
    drug product. Generally, an intermediate will be quantifiable and 
    specifications will be established to determine the successful 
    completion of the manufacturing step prior to continuation of the 
    manufacturing process. This includes material that may undergo 
    further molecular modification or be held for an extended period of 
    time prior to further processing.
    
    Manufacturing-Scale Production
    
         Manufacture at the scale typically encountered in a facility at 
    the largest capacity intended for product production for marketing.
    
    Pilot-Plant Scale
    
         The production of the drug substance or drug product by a 
    procedure fully representative of and simulating that to be applied 
    at manufacturing scale. The methods of cell expansion, harvest, and 
    product purification should be identical except for the scale of 
    production.
    
    Potency
    
         Expression of the predicted capacity of a product to achieve 
    its intended role; it is based on the measurement of some attribute 
    of the product and is determined by a suitable quantitative 
    laboratory method. In general, potencies of biotechnological/
    biological products tested by different laboratories can be compared 
    in a meaningful way only if expressed in relation to that of an 
    appropriate reference material. For that purpose a reference 
    material calibrated directly or indirectly against the corresponding 
    national or international reference material is included in the 
    assay. The reference material should have some known relationship 
    with the product, the therapeutic, preventive, or diagnostic 
    capacity of which has been studied in humans.
    
    Purity
    
         Purity is a relative term with respect to biotechnological/
    biological products. The purity may be expressed as the amount 
    (weight/weight) of the desired protein of a homogeneous amino acid 
    sequence usually expressed on a percentage basis. However, due to 
    the effects of glycosylation, deamidation, etc., the absolute purity 
    of a biotechnological/biological product is extremely difficult to 
    determine. Therefore, the purity of biotechnological/biological 
    products may be evaluated by determining the amounts of known 
    impurities, such as host cell proteins, DNA, other impurities, and/
    or degradation products. Thus, the purity of a biotechnological/
    biological product is typically assessed by more than one method and 
    the purity value derived is method-dependent. For example, the 
    purity values derived from a chromatographic and by an 
    electrophoretic method may be different but equally valid for a 
    given batch of biotechnological/biological product because each 
    method focuses on a different aspect of this biological entity.
    
    Well-Characterized (Biotechnological/Biological) Product
    
         A product whose structural features (including amino acid 
    sequences, as well as physicochemical, biochemical, biological and/
    or immunochemical properties) have been elucidated using a set of 
    modern, bioanalytical, and testing methods.
    
        Dated: August 14, 1995.
    William K. Hubbard,
    Acting Deputy Commissioner for Policy.
    [FR Doc. 95-20608 Filed 8-18-95; 8:45 am]
    BILLING CODE 4160-01-F
    
    

Document Information

Published:
08/21/1995
Entry Type:
Notice
Action:
Notice.
Document Number:
95-20608
Dates:
Written comments by October 5, 1995.
Pages:
43501-43505 (5 pages)
Docket Numbers:
Docket No. 93D-0139
PDF File:
95-20608.pdf