'FDA industry Guidance for Drug Substance CMC Information
化学合成原料药起始物料国内外相关要求的比较
999 中国新药杂志 2014 年第 23 卷第 9 期
Chinese Journal of New Drugs 2014,23(9)
大于 0. 10%[5]。④ 起始物料与原料药之间应有多 个合成步骤,中间体未进行分离纯化的反应不算作 合成步骤[5,13]。FDA 把合成路线中最临近原料药的 化合物定义为最后中间体,并规定,从最后中间体生 成原料药的反应必须涉及共价键的形成或断裂,且 不能是简单的成酯( 例如甲酯、乙酯等) 、皂化等反 应,也不 能 是 成 盐、重 结 晶 或 拆 分 等[5,13 - 14]。FDA 还要求,如拟缩短所批准的合成路线,新的起始物料 一般应在最后中间体之前,也就是从起始物料到原 料药之间至少应有两个合成步骤[6]。⑤ 起始物料 的结构不能过于复杂,采用常用技术( 例如红外光 谱) 应能区分拟用作起始物料的化学物质与其潜在 的异构体和类似物[5]。
[Abstract] The selection and quality control of starting materials are an important part of drug substance development. In the past 20 years,this topic involves in the technical requirements for drug substance registration, and gets attention quite all time. As the publication of ICH Q11 guideline,agreement on general selection principles and quality control requirements of starting materials has been reached in ICH regions. In comparison,we are still lagging behind in the understanding of this topic. In this article,we reviewed FDA,EMA,ICH and domestic related requirements of starting materials for synthetic drug substances. Furthermore,we analyzed our existing problems focusing on the regulatory function,selection principles,and document submission requirements for starting materials. We want to provide a reference to amend the domestic related requirements for starting materials.
FDA无菌加工生产的无菌药品指南中英文对照版
监控程序
VI. COMPONENTS AND CONTAINER/CLOSURES .................................................... 27
药品成分和容器 /密封 A. Components..................................................................................................................................27
法规架构 B. Technical Framework .......................................................................................................iologics Evaluation and Research Food and Drug Administration
1401 Rockville Pike, Rockville, MD 20852-1448 /cber/guidelines.htm.
辅助洁净区域 C. Clean Area Separation ................................................................................................................ 11
净化区的隔离 D. Air Filtration ................................................................................................................................ 12
鼻用制剂FDA标准
Nasal Spray and Inhalation Solution, Suspension, and Spray Drug Products — Chemistry, Manufacturing, and Controls Documentation `U.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)July 2002CMCNasal Spray and Inhalation Solution, Suspension, and Spray Drug Products —Chemistry, Manufacturing, and ControlsDocumentationAdditional copies are available from:Office of Training and CommunicationsDivision of Drug Information, HFD-240Center for Drug Evaluation and ResearchFood and Drug Administration5600 Fishers LaneRockville, MD 20857(Tel) 301-827-4573/cder/guidance/index.htmU.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)July 2002CMCTABLE OF CONTENTSI. INTRODUCTION (1)II. BACKGROUND (2)A. Nasal Sprays (2)B. Inhalation Solutions and Suspensions (3)C. Inhalation Sprays (3)III. DRUG PRODUCT (5)A. Formulation Components (5)B. Formulation Composition (5)C. Specifications for the Formulation Components (5)D. Manufacturer (9)E.Method of Manufacture and Packaging (9)F. Specifications for the Drug Product (10)G. Container Closure Systems (22)H.Drug Product Stability (27)IV.DRUG PRODUCT CHARACTERIZATION STUDIES (32)A.Priming and Repriming in Various Orientations (33)B.Effect of Resting Time (33)C.Temperature Cycling (33)D.In Vitro Dose Proportionality (34)E.Cleaning Instructions (34)F.Device Robustness (34)G.Effect of Dosing Orientation (34)H.Effect of Varying Flow Rates (35)I.Profiling of Sprays Near Container Exhaustion (Tail Off Characteristics) (35)J.Effect of Storage on the Particle Size Distribution (35)K.Plume Geometry (36)L.Preservative Effectiveness and Sterility Maintenance (36)M.Characterization of Nebulizer Specified in the Labeling (36)N.Photostability (36)O.Stability of Primary (Unprotected) Package (37)BELING CONSIDERATIONS (37)A.Nasal and Inhalation Spray Drug Products (37)B.Inhalation Solutions and Suspensions (40)GLOSSARY OF TERMS (43)GUIDANCE FOR INDUSTRY1Nasal Spray and Inhalation Solution, Suspension, and Spray Drug Products — Chemistry, Manufacturing, and Controls Documentation This guidance represents the Food and Drug Administration's (FDA's) current thinking on this topic. It does not create or confer any rights for or on any person and does not operate to bind FDA or the public. An alternative approach may be used if such approach satisfies the requirements of the applicable statutes and regulations.I. INTRODUCTIONThis document provides guidance for industry on the chemistry, manufacturing, and controls (CMC) documentation that should be submitted in new drug applications (NDAs) and abbreviated new drug applications (ANDAs) for nasal spray and inhalation solution, suspension, and spray drug products intended for local and/or systemic effect. This guidance covers CMC information recommended for inclusion in the application regarding the drug product components, manufacturing process, and associated controls for each of these areas, but does not address the manufacture of drug substances. The guidance also provides recommendations on labeling. This guidance does not address propellant-based inhalation and nasal aerosols (also known as oral and nasal metered-dose inhalers, MDIs), inhalation powders (also known as dry powder inhalers, DPIs), and nasal powders.2This guidance sets forth information that should be provided to ensure continuing quality and performance characteristics for these drug products. The guidance does not impose mandatory requirements but does suggest approaches that are appropriate for submitting CMC-related regulatory information. The guidance provides recommendations for drug1 This guidance has been prepared by the Inhalation Drug Products Working Group of the Chemistry, Manufacturing, and Controls Coordinating Committee (CMCCC) in the Center for Drug Evaluation and Research (CDER) at the FDA.2 In November 1998 (63 FR 64270), the Agency made available a draft guidance document on Metered Dose Inhaler (MDI) and Dry Powder Inhaler (DPI) Drug Products Chemistry, Manufacturing, and Controls Documentation. When finalized, this guidance will provide CMC recommendations for MDIs and DPIs.products that are used to treat a variety of diseases and patient populations. Therefore, CMC recommendations may vary depending on the specific drug product and stage of development. For example, the recommendations in this guidance should be considered during the investigational stages and phased in by the initiation of critical clinical studies (phase 2 and phase 3 studies) to provide supporting documentation for an NDA. Applicants are encouraged to discuss significant departures from the approaches outlined in this guidance (including decisions to provide less CMC documentation than recommended) with the appropriate Agency review division before implementation to avoid expending resources on development avenues that may later be deemed inappropriate.Reference to information in Drug Master Files (DMFs) for particular portions of the CMC section of the application is appropriate if the DMF holder provides written authorization that includes specific reference (e.g., submission date, page number, item name and unique identifier) to the pertinent and up-to-date information (21 CFR314.420(d)). Refer to FDA=s Guideline for Drug Master Files (September 1989) for more information about DMFs.II. BACKGROUNDA. Nasal SpraysNasal spray drug products contain therapeutically active ingredients (drugsubstances) dissolved or suspended in solutions or mixtures of excipients (e.g.,preservatives, viscosity modifiers, emulsifiers, buffering agents) innonpressurized dispensers that deliver a spray containing a metered dose of theactive ingredient. The dose can be metered by the spray pump or could have been premetered during manufacture. A nasal spray unit can be designed for unitdosing or can discharge up to several hundred metered sprays of formulationcontaining the drug substance. Nasal sprays are applied to the nasal cavity forlocal and/or systemic effects.Although similar in many features to other drug products, some aspects of nasalsprays may be unique (e.g., formulation, container closure system, manufacturing, stability, controls of critical steps, intermediates, and drug product). Theseaspects should be considered carefully during the development program becausechanges can affect the ability of the product to deliver reproducible doses topatients throughout the product=s shelf life. Some of the unique features of nasalsprays are listed below:$Metering and spray producing (e.g., orifice, nozzle, jet) pump mechanisms and components are used for reproducible delivery of drug formulation,and these can be constructed of many parts of different design that areprecisely controlled in terms of dimensions and composition.$Energy is required for dispersion of the formulation as a spray. This is typically accomplished by forcing the formulation through the nasalactuator and its orifice.$The formulation and the container closure system (container, closure, pump, and any protective packaging) collectively constitute the drugproduct. The design of the container closure system affects the dosingperformance of the drug product.$The concept of classical bioequivalence and bioavailability may not be applicable for all nasal sprays, depending on the intended site of action.The doses administered are typically so small that blood or serumconcentrations are generally undetectable by routine analytical procedures.Additional information will be provided in a future guidance for industryon Bioavailability and Bioequivalence Studies for Nasal Aerosols andNasal Sprays for Local Action.3B. Inhalation Solutions and SuspensionsInhalation solution and suspension drug products are typically aqueous-basedformulations that contain therapeutically active ingredients and can also containadditional excipients. Aqueous-based oral inhalation solutions and suspensionmust be sterile (21 CFR 200.51). Inhalation solutions and suspensions areintended for delivery to the lungs by oral inhalation for local and/or systemiceffects and are to be used with a specified nebulizer. Unit-dose presentation isrecommended for these drug products to prevent microbial contamination duringuse. The container closure system for these drug products consists of thecontainer and closure, and can include protective packaging such as foiloverwrap. Recommendations on overwrapping of inhalation drug productspackaged in semipermeable container closure systems are provided in sectionIII.G.5.C. Inhalation SpraysAn inhalation spray drug product consists of the formulation and the containerclosure system. The formulations are typically aqueous based and, by definition,do not contain any propellant. Aqueous-based oral inhalation sprays must besterile (21 CFR 200.51). Inhalation sprays are intended for delivery to the lungsby oral inhalation for local and/or systemic effects. The products containtherapeutically active ingredients and can also contain additional excipients. The formulation can be in unit-dose or multidose presentations. The use ofpreservatives or stablilizing agents in inhalation spray formulations is3A notice of availability for the June 1999 draft guidance Bioavailability and Bioequivalence Studies for Nasal Aerosols and Nasal Sprays for Local Action published in the Federal Register on June 24, 1999 (64 FR 33869).discouraged. If these excipients are included in a formulation, their use should be justified by assessment in a clinical setting to ensure the safety and tolerability of the drug product. The dose is delivered by the integral pump components of thecontainer closure system to the lungs by oral inhalation for local and/or systemiceffects. The container closure system of these drug products consists of thecontainer, closure, and pump, and can also include protective packaging.Current container closure system designs for inhalation spray drug productsinclude both premetered and device-metered presentations using mechanical or power assistance and/or energy from patient inspiration for production of thespray plume. Premetered presentations contain previously measured doses or adose fraction in some type of units (e.g., single or multiple blisters or othercavities) that are subsequently inserted into the device during manufacture or bythe patient before use. Typical device-metered units have a reservoir containingformulation sufficient for multiple doses that are delivered as metered sprays bythe device itself when activated by the patient.Inhalation spray and nasal spray drug products have many similarities. Therefore, many of the unique features listed in section II.A for nasal sprays are alsocharacteristic of inhalation spray drug products. Moreover, the potential widearray of inhalation spray drug product designs with unique characteristics willpresent a variety of development challenges. Regardless of the design, the mostcrucial attributes are the reproducibility of the dose, the spray plume, and theparticle/droplet4 size distribution, since these parameters can affect the delivery of the drug substance to the intended biological target. Maintaining thereproducibility of these parameters through the expiration dating period andensuring the sterility of the content and the functionality of the device (e.g., spray mechanism, electronic features, sensors) through its lifetime under patient-useconditions will probably present the most formidable challenges. Therefore,changes in components of the drug product or changes in the manufacturer ormanufacturing process that can affect these parameters should be carefullyevaluated for their effect on the safety, clinical effectiveness and stability of theproduct. If such changes are made subsequent to the preparation of the batchesused in critical clinical, bioequivalence, or primary stability studies, adequatesupportive comparative data should be provided to demonstrate equivalency interms of safety, clinical effectiveness, and stability of the product.The remaining portion of this guidance will focus on specific chemistry,manufacturing, and controls information recommended for inclusion in the drugproduct section of applications for nasal spray and inhalation solution, suspension, and spray drug products.4 The term particle/droplet refers to a combination of droplets and particles or droplets alone, depending on the formulation and conditions of measurement.III. DRUG PRODUCTA. Formulation ComponentsA list of all components (i.e., ingredients) used in the manufacture of the drugproduct formulation, regardless of whether they undergo chemical change or areremoved during manufacture, should be included in the application. Eachcomponent should be identified by its established name, if any, and by itscomplete chemical name, using structural formulas when warranted for specificidentification. If any proprietary preparations or other mixtures are used ascomponents, their identity should be fully described including a completestatement of their composition and other information that will properly identifythe material.B. Formulation CompositionThe application should include a statement of the quantitative composition of the unit formula of the drug product, specifying the name and amount of each activeingredient and excipient contained in a stated quantity of the formulation. Forcomponents in the final formulation, the amounts should be expressed inconcentration (i.e., amount per unit volume or weight), as well as amount percontainer and per spray, where applicable. The target container net contentshould also be indicated. Similarly, a production batch formula representative of the one to be employed in the manufacture of the drug product should beincluded. Any calculated overage for an ingredient should be designated as such and the percentage shown. The overage should be scientifically justified anddocumented in both the unit formula and batch formula. For these products,overages can be included only for justified reproducible manufacturing lossesand/or for an ANDA product to match the overage present in the Reference Listed Drug. Any intended change in the formulation of the commercial product fromthat used in the submitted batches (e.g., critical clinical, biobatch, primarystability, production) should be clearly indicated by providing the composition of each formulation.The composition of suspension formulations may be crucial in defining thephysical stability and the performance characteristics of the drug product. Thedensity and suspension properties of the solid materials of the formulation and the potential for agglomeration should be considered. Moreover, interaction of thesuspended drug substance with the various internal container closure systemcomponents can also contribute to a nonhomogeneous distribution of drugsubstance. The above mentioned phenomena, which may be exacerbated withtime, can contribute to inconsistent particle size distribution and medication dose delivery. See also the discussions in sections III.F.1.c and III.F.2.c.C. Specifications for the Formulation Components1. Active IngredientsInformation regarding the comprehensive characterization of the physical andchemical properties of the drug substance should be included in the application.Important properties of the drug substance used in suspension formulations caninclude, but are not necessarily limited to, density, particle size distribution,particle morphology, solvates and hydrates, polymorphs, amorphous forms,solubility profile, moisture and/or residual solvent content, microbial quality,dissociation constants (pKa), and specific rotation.Appropriate acceptance criteria and tests for routine control (i.e., release, stability, and retest) should be instituted for those drug substance parameters consideredkey to ensuring reproducibility of the physicochemical properties of the drugsubstance. Specification parameters can include, as applicable, color, appearance (visual and microscopic), specific identification, moisture, residue on ignition,specific rotation, assay, impurities, microbial limits (U.S. Pharmacopeia (USP)<61>)5, melting range, particle size distribution, crystalline forms, amorphouscontent, residual solvents, and heavy metals. Some of these parameters may notbe pertinent for drug substances used in solution formulations.The purity of the drug substance and its impurity profile should be characterizedand controlled with appropriate specifications. Important impurity-relatedparameters can include organic volatile impurities and/or residual solvents,organic impurities (synthesis-related and degradation products), and inorganicimpurities (e.g., heavy metals, reagents, catalysts). Any impurity found in thedrug substance at a concentration of 0.10 percent or 1.0 milligram (mg) per dayintake (whichever is lower), relative to the parent drug substance, should beidentified. Moreover, the drug substance impurities should be appropriatelyqualified. Justification of acceptance criteria for the drug substance impuritiesshould be based on toxicological considerations and levels of impurities found in the submitted batches (e.g., critical clinical, biobatch, primary stability,production). For guidance on toxicological qualification, the applicant isencouraged to refer to the following guidance documents: (1) ICH Q3AImpurities in New Drug Substances (January 1996),6 (2) NDAs: Impurities inDrug Substances (February 2000), and (3) ANDAs: Impurities in Drug Substances (November 1999). The applicant can also contact the responsible review division for guidance on toxicological qualification.For suspension formulations, the specification for drug substance should includecontrols for particle size distribution and physical properties (e.g., shape, crystal5 Sample size for microbial limits testing should be 10 grams unless otherwise justified.6 The guidance, Q3A Impurities in New Drug Substances, will be superseded by FDA’s guidance for industry, Q3A(R) Impurities in New Drug Substances, once it is issued in final form. We update guidances periodically. To make sure you have the most recent version of a guidance, check the CDER guidance page at /cder/guidance/index.htm.habit, morphology, surface texture) of the drug substance, parameters that are often critical for reproducible drug product performance. If laser diffraction methodology is used for testing the particle size distribution, it is crucial that test procedure instrumental parameters (e.g., apparatus and accessories, calculation theory, correction principles, software version, sample placement, laser trigger condition, measurement range, beam width) be defined accurately and with sufficient detail for Agency laboratories to validate the adequacy of the methodology. In addition, the potential effect of micronization processes on the levels of amorphous content and foreign particulates in the drug substance should be considered.In general, acceptance criteria for all parameters defining the physicochemical properties should be based on historical data, thereby providing continuity of quality and reproducible performance of future batches of the drug substance.For additional information on various aspects of drug substance chemistry, manufacturing, and controls documentation, see the FDA Guideline for Submitting Supporting Documentation in Drug Applications for the Manufacture of Drug Substances (February 1987).2. ExcipientsBecause of the route of administration and the sensitive nature of various patient populations using oral inhalation (solution, suspension, spray) drug products, more thorough characterization with additional comprehensive controls (e.g., strength, quality, purity), as compared to drug products for other routes of administration, should be considered for excipients used in these drug products. Moreover, for nasal and inhalation suspension formulations, additional controls should be applied to critical excipients to ensure safety and effectiveness of the drug product. Critical excipients for suspension formulations (e.g., microcrystalline cellulose for nasal sprays) are those that can affect the suspension and/or particle characteristics and, therefore, the quality, stability, or performance of the drug product. The suitability of the physicochemical properties of these critical excipients should be thoroughly investigated and documented.Unless otherwise indicated, the comments below regarding excipients pertain to nasal spray and inhalation solution, suspension, and spray drug products.The source of each excipient should be assessed, and the material supplied should meet appropriate acceptance criteria that are based on test results from a minimum of one batch used to prepare the submitted batches of drug product (e.g., critical clinical, biobatch, primary stability, production). However, for critical excipients of suspension formulations, the sources should be identified and test results from multiple batches should be provided. Likewise, when the supplier of an excipient is changed prior to submission of the application, the new supplier=s ability toprovide material of comparative quality should be assessed and supporting data should be provided.For noncompendial excipients, appropriate authorization to a DMF that provides information on the noncompendial excipient or an equivalent package of information prepared by the excipient manufacturer should be provided in the application. The information should include analytical procedures, acceptance criteria, and a brief description of the manufacture and controls.When a USP or National Formulary (NF) monograph material is used, the associated specifications may not always provide adequate assurance with regard to the assay, quality, or purity of the material or its performance in the drug product. In these cases, monograph specifications should be supplemented with appropriate controls (e.g., particle size distribution, crystal forms, amorphous content, foreign particulates) to ensure batch-to-batch reproducibility of these components. This can be particularly relevant for compendial excipients that have an impact on the purity of inhalation drug products or performance properties (e.g., droplet and particle size distribution, spray content uniformity) of suspension drug products. The additional test procedures should be included, and the acceptance criteria should reflect the data for the excipients used in the submitted batches (e.g., critical clinical, biobatch, primary stability, production). Acceptance criteria for physicochemical parameters of a qualified polymeric excipient (e.g., molecular weight distribution, viscosity) that are wider than what is reflective of the data on the submitted batches can be justified by demonstrating that the proposed ranges of the excipient attributes do not adversely affect the quality of the drug product. Justification should be based on adequate release and stability data that is specific to the drug product prepared with the excipient attributes near the limits of the allowable range.The suitability of the toxicological properties of the excipients for these drug products should be thoroughly investigated and documented. Toxicological qualification of these excipients may be appropriate under various circumstances, including (1) increased concentration of an excipient above that previously used in inhalation and nasal drug products, (2) excipients that have been used previously in humans but not by the inhalation or nasal route, and (3) novel excipients not previously used in humans in the United States. The extent of toxicological investigation to qualify the use of an excipient under such circumstances will vary, and the applicant is encouraged to contact the responsible review division to discuss an appropriate strategy for toxicological qualification.If excipients are accepted based on certificates of analysis from the manufacturers with the applicant performing a specific identification test upon receipt, the applicant should also develop validated procedures, have access to all of the manufacturer=s analytical and other test procedures, or use contract laboratories to allow them to establish the reliability of the test results at appropriate intervals, as required under 21 CFR 211.84. The applicant should confirm the supplier=sresults by (1) testing an adequate number of batches of each excipient used in preparing the submitted drug product batches (e.g., critical clinical, primary stability, biobatch, production batches) and (2) providing a commitment to test a predetermined number of batches of each excipient used in preparing postapproval drug product batches.D. ManufacturersThe name, street address, and, if available, registration number7 of each facility involved in the manufacture of the drug substance should be listed along with a statement of each manufacturer's specific operations and responsibilities. The same information should be provided for each facility involved in the manufacturing, processing, packaging, controls, stability testing, or labeling of the drug product, including all contractors (e.g., test laboratories, packagers, labelers). For sterile drug products, building numbers, filling rooms, and filling lines should also be identified. Manufacturers of critical and novel excipients should be identified by name and address.E.Method of Manufacture and PackagingA detailed description of the manufacturing, processing, and packaging procedures for the drug product should be included.All aqueous-based oral inhalation drug products must be manufactured as sterile products (21 CFR 200.51), and their sterility should be ensured through the expiration dating period.If micronization is used for the drug substance and/or excipients, the process should be fully validated and the equipment, operating conditions, and process controls should be described in detail. For example, the description of the controls for a milling operation could include the rate of feed, air pressure, air flow rate, particle size being fed, number of times a lot is micronized, re-use of carryovers from previous micronized lots. Potential contamination of the material during the micronization process should be controlled with appropriate tests and acceptance criteria. See the discussion of testing attributes specific for micronized material (e.g., particle size distribution, crystal forms, amorphous content, foreign particulates) discussed in section III.C.1.A copy of the actual (executed) batch record, including process controls, and controls for critical steps and intermediates should be submitted, as appropriate, for representative batches (e.g., critical clinical, biobatch, primary stability). A schematic diagram of the proposed production process, a list of process controls, and a master batch production and controls record should be submitted. A brief 7 Information on when registration is required and how to register is available in 21 CFR 207.。
FDA药物临床前指南-Guidance for Industry Unmarketed Investigational Drugs
Codevelopment of Two or More Unmarketed Investigational Drugs for Use in CombinationDRAFT GUIDANCEThis guidance document is being distributed for comment purposes only. Comments and suggestions regarding this draft document should be submitted within 60 days of publication in the Federal Register of the notice announcing the availability of the draft guidance. Submit comments to the Division of Dockets Management (HFA-305), Food and Drug Administration, 5630 Fishers Lane, rm. 1061, Rockville, MD 20852. All comments should be identified with the docket number listed in the notice of availability that publishes in the Federal Register.For questions regarding this draft document contact (CDER) Colleen Locicero 301-796-1114.U.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)December 2010Clinical MedicalCodevelopment of Two or More Unmarketed Investigational Drugs for Use in CombinationAdditional copies are available from:Office of CommunicationsDivision of Drug Information, WO51, Room 220110903 New Hampshire Ave.Silver Spring, MD 20993Phone: 301-796-3400; Fax: 301-847-8714druginfo@/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htmU.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)December 2010Clinical MedicalTable of Contents 1234 5 6 7 8 910111213141516171819202122 I.INTRODUCTION (1)II.BACKGROUND (2)III.DETERMINING WHETHER CODEVELOPMENT IS AN APPROPRIATEDEVELOPMENTOPTION (2)IV.NONCLINICAL CODEVELOPMENT (3)A.Demonstrating the Biological Rationale for the Combination (3)B.Nonclinical Safety Characterization (3)V.CLINICAL CODEVELOPMENT (4)A.Early Human Studies (Phase 1) (4)1.Safety of the Individual Components (4)2.Safety and Dosing of the Combination (5)B.Clinical Pharmacology (5)C.Proof of Concept Studies (Phase 2) (6)D. Confirmatory Studies (Phase 3) (8)VI.REGULATORY PROCESS ISSUES IN CODEVELOPMENT (8)A. Early Interaction with FDA (8)B. IND Submissions and Marketing Applications (9)beling Issues (9)D.Pharmacovigilance (9)232425262728Guidance for Industry1Codevelopment of Two or More Unmarketed Investigational Drugsfor Use in Combination2930 This draft guidance, when finalized, will represent the Food and Drug Administration’s (FDA’s) current31 thinking on this topic. It does not create or confer any rights for or on any person and does not operate to32 bind FDA or the public. You can use an alternative approach if the approach satisfies the requirements of33 the applicable statutes and regulations. If you want to discuss an alternative approach, contact the FDA34 staff responsible for implementing this guidance. If you cannot identify the appropriate FDA staff, call35 the appropriate number listed on the title page of this guidance.36373839404142434445464748495051525354555657 I. INTRODUCTIONThis guidance is intended to assist sponsors in the codevelopment2 of two or more novel (not previously marketed) drugs to be used in combination to treat a disease or condition. The guidance provides recommendations and advice on how to address certain scientific and regulatory issues that will arise during codevelopment. It is not intended to apply to development of fixed-dose combinations of already marketed drugs or to development of a single new investigational drug to be used in combination with an approved drug or drugs. The guidance is also not intended to apply to vaccines, gene or cellular therapies, blood products, or medical devices.3FDA’s guidance documents, including this guidance, do not establish legally enforceable responsibilities. Instead, guidances describe the Agency’s current thinking on a topic and should be viewed only as recommendations, unless specific regulatory or statutory requirements are cited. The use of the word should in Agency guidances means that something is suggested or recommended, but not required.1 This guidance has been prepared by the Office of Medical Policy in the Center for Drug Evaluation and Research (CDER) at the Food and Drug Administration.2Codevelopment herein refers to the concurrent development of two or more drug products with the intent that the products be used in combination to treat a disease or condition.3 For purposes of this guidance, the term drug includes therapeutic biological products that are regulated by CDER. Consult the Therapeutic Biologics web page for further information on the types of biological products to which this guidance applies:/Drugs/DevelopmentApprovalProcess/HowDrugsareDevelopedandApproved/ApprovalApplications/T herapeuticBiologicApplications/default.htmII. BACKGROUND585960616263646566676869707172737475767778798081828384858687888990919293949596979899 100 101 102 103 Combination therapy is an important treatment modality in many disease settings, including cancer, cardio-vascular disease, and infectious diseases. Recent scientific advances have increased our understanding of the pathophysiological processes that underlie these and other complex diseases. This increased understanding has provided further impetus for new therapeutic approaches using combinations of drugs directed at multiple therapeutic targets to improve treatment response or minimize development of resistance. In settings in which combination therapy provides significant therapeutic advantages, there is growing interest in the development of combinations of investigational drugs not previously developed for any purpose.Because the existing developmental and regulatory paradigm focuses primarily on assessment of the effectiveness and safety of a single new investigational drug acting alone, or in combination with an approved drug, FDA believes guidance is needed to assist sponsors in the codevelopment of two or more unmarketed drugs. Although interest in codevelopment has been most prominent in oncology and infectious disease settings, codevelopment also has potential application in other therapeutic settings. Therefore, this guidance is intended to describe a high-level, generally applicable approach to codevelopment of two or more unmarketed drugs. It describes the criteria for determining when codevelopment is an appropriate option, makes recommendations about nonclinical and clinical development strategies, and addresses certain regulatory process issues. III. DETERMININGWHETHERCODEVELOPMENT IS AN APPROPRIATE DEVELOPMENT OPTIONConcurrent development of two or more novel drugs for use in combination generally will provide less information about the safety and effectiveness of the individual drugs than would be obtained if the individual drugs were developed alone. How much less will vary depending on a variety of factors, including the stage of development at which the individual drug components cease to be studied independently. For example, in codevelopment scenarios in which rapid development of resistance to monotherapy is a major concern, it may not be possible or appropriate to obtain clinical data for the individual components of the combination beyond phase 1 testing. Because codevelopment will generally provide less information about the safety and effectiveness of the individual drugs, it will present greater risk compared to development of an individual drug. Therefore, FDA believes that codevelopment should ordinarily be reserved for situations that meet the following criteria:•The combination is intended to treat a serious disease or condition.•There is a compelling biological rationale for use of the combination (e.g., the agents inhibit distinct targets in the same molecular pathway, provide inhibition of both aprimary and compensatory pathway, or inhibit the same target at different binding sites to decrease resistance or allow use of lower doses to minimize toxicity).• A preclinical model (in vivo or in vitro) or short-term clinical study on an established biomarker suggests that the combination has substantial activity and provides greater thanadditive activity or a more durable response (e.g., delayed resistance) compared to the individual agents alone. 104105106107108109110111112113114115116117118119120121122123124125126127128129130131132133134135136137138139140141142143144145146 • There is a compelling reason for why the agents cannot be developed individually (e.g., monotherapy for the disease of interest leads to resistance and/or one or both of the agents would be expected to have very limited activity when used as monotherapy). FDA recommends that sponsors consult with FDA on the appropriateness of codevelopment before initiation of clinical development of the combination. IV. NONCLINICAL CODEVELOPMENT A. Demonstrating the Biological Rationale for the Combination The biology of the disease, pathogen, or tumor type should be sufficiently understood to provide a plausible biological rationale for the use of combination therapy to treat the disease or condition. For example, in an oncology setting the biological rationale may be to intervene at different steps in the cell proliferation pathway. The biological rationale for a combination anti-infective therapy may be to target different metabolic pathways or different steps in the replication cycle of the pathogen to reduce the chance of developing resistance to the therapy or increase efficacy in treating disease caused by resistant organisms (e.g., multidrug-resistant atypical tuberculosis). Sponsors should develop evidence to support the biological rationale for the combination in an in vivo (preferable) or in vitro model. The model should compare the activity of the combination to the activity of the individual components. Ordinarily, the model should demonstrate that, compared to the individual components, the combination has substantial activity and provides greater than additive activity or a more durable response in a pathophysiological process considered pertinent to the drug’s intended use in humans. An animal model of activity generally would not be necessary. However, if there is an animal model relevant to the human disease, valuable activity data, as well as information about the relative doses of the drugs, might be obtained from evaluating the combination in that model. B. Nonclinical Safety Characterization For detailed recommendations regarding nonclinical safety characterization for two or more investigational drugs to be used in combination, sponsors should consult the recently revised International Conference on Harmonisation (ICH) Guidance on Nonclinical Safety Studies.4 Section XVII of that guidance (Combination Drug Toxicity Testing) includes a discussion of nonclinical safety studies appropriate in a combination drug development setting involving two early stage entities. The ICH guidance defines early stage entities as compounds with limited clinical experience (i.e., phase 2 studies or less), so the discussion is specifically applicable to the4 Guidance for Industry: M3(R2) Nonclinical Safety Studies for the Conduct of Human Clinical Trials andMarketing Authorization, January 2010 (this guidance is a revision of 1997 ICH guidance M3: Nonclinical Safety Studies for the Conduct of Human Clinical Trials for Pharmaceuticals).147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 type of development described in this guidance. In situations in which it is possible to obtain only limited clinical data for the individual drugs, additional nonclinical data for the individual drugs or combination may be needed before beginning human studies with the combination. (e.g., see section V.A.1).V. CLINICAL CODEVELOPMENTThis section provides a general roadmap and guiding principles for concurrent clinical development of two or more investigational drugs to be used in combination. It includes recommendations for characterizing the clinical safety and effectiveness of the combination and, to the extent needed or possible, the individual components of the combination.Note: The appropriate review division should always be consulted on the specifics of a given clinical development program.A. Early Human Studies (Phase 1)The main objectives of early studies in humans are to characterize the safety and pharmacokinetics of the individual components and then the combination and to provide data to support appropriate dosing for the combination in phase 2 testing.1. Safety of the Individual ComponentsWhenever possible, the safety profile of each individual drug should be characterized in phase 1 studies in healthy volunteers in the same manner as would be done fordevelopment of a single drug, including determination of the maximum tolerated dose(MTD), the nature of the dose limiting toxicity (DLT), and pharmacokinetic parameters.If there is a useful measure (e.g., biomarker) of pharmacologic activity, it will also beimportant to determine dose-response for that measure. If testing in healthy volunteers is not possible (e.g., if nonclinical data suggest a drug may be genotoxic or otherwiseunacceptable for studies in healthy volunteers), the safety profile of the individual drugs should be evaluated in patients with the disease of interest. These safety data will guide decisions in later studies about starting doses, dose escalation increments, and final dose selection.If it is not possible to characterize the safety of the individual drugs in humans (e.g.,where drug toxicity prevents use of healthy volunteers and monotherapy would beunethical in patients with the disease of interest), the sponsor should conduct nonclinical studies of the combination to support initial dosing of the combination in humans.The nonclinical data for the combination should include pharmacokinetic (absorption,distribution, metabolism, and excretion) and toxicokinetic data and appropriatebiomarker/target inhibition, if relevant.2. Safety and Dosing of the Combination 193194195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237For initial human effectiveness studies of the combination, the combination starting dose, dosing escalation intervals, and doses to be used in dose-response studies should bedetermined based on phase 1 safety data for the individual components, if available. Ifphase 1 safety data for the components are unavailable, nonclinical data for thecombination will be needed to determine the initial combination dose in humans (seeprevious paragraph). Phase 1 safety studies of the combination could also be conducted — for example, sequential testing in which subjects get drug A, then drug B, then AB — to support dosing in subsequent studies.B. ClinicalPharmacologyThe sponsor should conduct the same clinical pharmacology studies for each of the individual drugs in the combination as would be done if the drugs were being developed separately. In general, such studies include the assessment of bioavailability, characterization of pharmacokinetics, mass balance, the evaluation of effects of intrinsic (such as renal impairment and hepatic impairment) and extrinsic (such as food effect and drug interactions) factors on pharmacokinetics or pharmacodynamics, and exposure-response. Studies to address intrinsic and extrinsic factors could be conducted with the combination instead of the individual drugs.The evaluation of drug interaction potential follows the same sequence as in other development programs; results of in vitro drug metabolism and drug transporter studies inform the need for in vivo drug interaction studies. The role of pharmacogenomics should be investigated and incorporated into the combination drug development plan to identify potential sources of pharmacokinetic or pharmacodynamic variability.Dose-response should be evaluated for each drug of the combination. The results of such studies should be used to determine doses to further explore for the combination. If the drug products cannot be administered alone, various doses of each drug administered as the combination should be assessed.If one drug has no activity or minimal activity by itself, dose-response should be assessed when the drug products are administered in combination using a number of doses of the active drug and the inactive drug. The same approach should be used in evaluating dose-response for the combination of drugs where each drug has minimal activity when used alone.In addition to evaluating dose-response, response should be evaluated with respect to systemic drug concentration to provide insight into efficacy and safety as a function of drug exposure. Concentration-response assessments should be done in both phase 2 and phase 3 trials. To increase exposure ranges in phase 3 and to further assess dose-response, the incorporation of more than one dose of each of the drugs used in the combination in the phase 3 trials should be considered.C. Proof of Concept Studies (Phase 2) 238239240 241 242 243 244 245 246247 248 249 250 251 252 253 254 255 256 257 258 In general, phase 2 testing should accomplish the following to the extent needed for a given combination (e.g., to the extent not sufficiently established by existing data): •Demonstrate the contribution of each component of the combination to the extent possible and needed (given available nonclinical and pharmacologic data);•Provide evidence of the effectiveness of the combination; and•Optimize the dose or doses of the combination for phase 3 trials.The amount and types of clinical data needed and appropriate study designs will vary depending on the nature of the combination being developed, the disease, and other factors. For the types of combinations contemplated by this guidance, it will often be inappropriate to use monotherapy treatment arms in studies of the disease of interest, or it will be possible to administer the components of the combination as monotherapy only for short durations. In these circumstances, the study design typically employed to determine the contributions of the components to the combination — a four-arm factorial design comparing the combination to individual components and placebo or standard of care (SOC) therapy (AB v. A. v. B v. placebo or SOC) — will have limited utility. The following scenarios illustrate possible phase 2 study designs for combinations of two investigational drugs in different situations.Scenario 1: The components of the combination cannot be administered individually259 260261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278If in vivo or in vitro models, or phase 1 or other early clinical studies make clear that the components of the combination cannot be administered individually in clinical trials inthe disease of interest (e.g., because such testing would involve administering treatment known to be ineffective as monotherapy), or can’t be administered as monotherapy forthe duration needed to evaluate effectiveness (e.g., because of rapid development ofresistance), proof-of-concept evidence for the combination ordinarily should come froma study directly comparing the combination (AB) to SOC. Alternatively, if SOC isknown to be an effective therapy (not solely palliative), an add-on design could be used comparing the combination plus SOC to SOC alone.In some resistance scenarios, it may be possible to administer the individual drugs in acombination as monotherapy for a short duration, but long enough to establish proof ofconcept in humans. For example, direct-acting antivirals (DAAs) to treat chronichepatitis C virus infection can be administered as monotherapy for three days to establish antiviral activity and for initial dose exploration. For DAA studies of longer duration, the combination should be used or the individual components should be added to an activecontrol.55 See draft guidance for industry: Chronic Hepatitis C Virus Infection: Developing Direct-Acting Antiviral Agents for Treatment (section III. 4. b. – Phase 1b (proof-of-concept) trials) or consult the Division of Antiviral Drug Products in CDER for more specific recommendations.279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 Scenario 2: Each drug alone has activity and can be administered individuallyIf in vivo or in vitro models, or phase 1 or other early clinical studies indicate that each drug has some activity, but the combination appears to have greater than additive activity, and rapid development of resistance is not a concern, a four-arm, phase 2 trial comparing the combination to each drug alone and to placebo or SOC (AB v. A v. B v. SOC or placebo6) should be used to demonstrate the contribution of the components to the combination and proof of concept. As noted above, if SOC is a known effective therapy, a study design in which each of the arms is added to SOC could be used (AB + SOC v. A + SOC v. B + SOC v. placebo + SOC).An adaptive trial design with the same four treatment arms might also be used where appropriate, initially using the treatment arms described above. The single-drug arms could be terminated early if it became clear that they had much less activity than the combination. These designs could demonstrate the activity of each component of (i.e., the contribution of each component to the combination) without exposing the large numbers of patients typically required for phase 3 trials to therapeutic products with inadequate activity. For these trials, it may not be necessary to use a clinical endpoint as a primary efficacy measurement. A credible pharmacodynamic or other biomarker, such as tumor response, may be adequate.300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 Scenario 3: One drug is active alone and one is inactiveIf in vivo or in vitro models, or phase 1 or other early clinical studies suggest that one of the drugs is inactive or minimally active and one drug is modestly active, but the combination has substantial activity, the more active drug generally will require greater scrutiny and should ordinarily be studied as a single drug in a phase 2 study. The minimally active drug generally would not require study as a single drug beyond initial phase 1 safety studies. In this scenario, proof of concept and the contribution of each component could be demonstrated using a three-arm comparison of the active drug alone, SOC, and the combination (AB v. A v. SOC), or the combination and the individual drug added to SOC where SOC is a known effective therapy (AB + SOC v. A + SOC v. SOC). If the inactive drug in a combination is a pharmacokinetic or metabolic enhancer that contributes to the activity of the combination only by increasing the therapeutic concentrations of the active drug, human pharmacokinetic data may provide adequate evidence to support the enhanced activity of the combination and demonstrate the contribution of the inactive drug. A confirmatory study of the combination would usually be needed to provide evidence of effectiveness for the combination (see section V.D). Dose Finding319 320321 322Dose-finding studies could be very important to refine the combination dose or doses and select doses for phase 3 trials. Depending on the role of each component, it may be6 Note that the placebo arm is intended to show the effect size compared to non-treatment, not to show the contribution of each component.323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367useful to test multiple doses of both components to establish a best dose in terms of risks and benefits. If one component in a two-drug combination is more active than the other, it may be more important to study multiple doses of the more active drug (as part of thecombination). For the same reason, it may be more important to study multiple doses ofa drug that is significantly more toxic than the other component of the combination.Other study designs and types of studies also may be appropriate.D. Confirmatory Studies (Phase 3)If findings from in vivo or in vitro models and/or phase 2 trials adequately demonstrate the contribution of each component to the combination, phase 3 trials comparing the combination to SOC or placebo generally will be sufficient to establish effectiveness. If the contribution of the individual components is not clear and it is ethically feasible to use a component or components of the combination as monotherapy in a study arm, it may be necessary to demonstrate the contribution of the components in phase 3 studies (e.g., by use of a factorial design). For example, if phase 2 data do not provide sufficient evidence of the contribution of each component of a two drug combination, but provide strong evidence that the combination is superior to one of the components, a phase 3 trial comparing the combination to the more active component alone and SOC may be needed to demonstrate that the less active component contributes to the activity of the combination. In this and other situations, it will often be useful to study more than one dose of the more active drug in phase 3 studies.Unexpected toxicity (e.g., serious adverse events observed at higher than expected rates) in phase 2 trials is a potential complication for development of a combination and progressing to phase 3 trials. If the toxicity can be attributed to one component of the combination, it may be possible to conduct phase 3 trials with the combination using a lower dose or doses of the more toxic component. If the toxicity cannot be attributed to an individual component of the combination, additional studies may be needed to identify the more toxic component and appropriate dosing for the combination before initiating phase 3 trials. The specifics of any phase 3 design should be discussed with the appropriate FDA review division at an End-of-Phase 2 meeting.VI. REGULATORY PROCESS ISSUES IN CODEVELOPMENTSponsors should consider a number of regulatory issues when planning the codevelopment of two or more novel drugs for use in combination. Key issues are outlined below.A. Early Interaction with FDASponsors are encouraged to communicate as early as possible (e.g., pre-IND meeting) with the appropriate FDA review division when considering codevelopment of innovative combination therapy. Sponsors also are encouraged to consult FDA frequently throughout the development process. We believe such communication will help facilitate development of the combination therapy.368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398B. IND Submissions and Marketing ApplicationsDecisions about the type of IND submission(s) and marketing application(s) needed (e.g., individual component submissions, combination submission) will depend on the sponsor's overall codevelopment and marketing strategy. Until FDA has more experience with codevelopment, FDA recommends that these decisions be made on a case-by-case basis in consultation with the appropriate review division.C. Labeling IssuesFDA also anticipates that the content of labeling for the combination and/or the components will be case specific, depending on the nature of the combination, the intended uses of the individual components, the marketing strategy, and other factors. Therefore, FDA does not believe it can provide generally applicable labeling guidance at this time. Again, we recommend consultation with the appropriate review division.D. PharmacovigilanceApplicants should develop a pharmacovigilance plan that takes into account the additional postmarket risks presented by initial marketing of two or more previously unapproved drugs for use in combination (compared to risks associated with marketing of a single drug). Risk will vary, depending on the nature of the combination and how the combination is marketed. The risk assessment should consider, among other things:•Potential for use of each drug individually;•Potential for use of any of the components of the combination in combinations with other drugs; and•Drugs likely to be co-administered with the combination.Applicants should discuss their pharmacovigilance plans with the appropriate review division and the Office of Surveillance and Epidemiology.。
Stability Testing of Drug Substances and Products(FDA)
ANDAs: Stability Testing of Drug Substances and Products U.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)June 2013GenericsANDAs: Stability Testing of Drug Substances and ProductsAdditional copies are available from:Office of CommunicationsDivision of Drug Information, WO51, Room 2201Center for Drug Evaluation and ResearchFood and Drug Administration10903 New Hampshire Ave., Silver Spring, MD 20993Phone: 301-796-3400; Fax: 301-847-8714druginfo@/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htmU.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)June 2013GenericsTABLE OF CONTENTSI.INTRODUCTION (1)II.BACKGROUND (1)III.DISCUSSION (2)Guidance for Industry1ANDAs: Stability Testing of Drug Substances and ProductsThis guidance represents the Food and Drug Administration’s (FDA’s) current thinking on this topic. It does not create or confer any rights for or on any person and does not operate to bind FDA or the public. You can use an alternative approach if the approach satisfies the requirements of the applicable statutes and regulations. If you want to discuss an alternative approach, contact the FDA staff responsible for implementing this guidance. If you cannot identify the appropriate FDA staff, call the appropriate number listed on the title page of this guidance.I. INTRODUCTIONThis guidance recommends that abbreviated new drug applications (ANDAs) submitted under section 505(j) of the Federal Food, Drug and Cosmetic Act, and the drug master files (DMFs) that support ANDAs, follow the stability recommendations provided in the International Conference on Harmonisation (ICH) stability guidances.FDA’s guidance documents, including this guidance, do not establish legally enforceable responsibilities. Instead, guidances describe the Agency’s current thinking on a topic and should be viewed only as recommendations, unless specific regulatory or statutory requirements are cited. The use of the word should in Agency guidances means that something is suggested or recommended, but not required.II. BACKGROUNDOver the past few years, the Office of Generic Drugs (OGD) has received numerous inquiries about what stability data FDA expects in ANDA submissions. Currently, the only published direction from OGD is contained in a 1995 letter to industry which states that OGD will accept ICH recommended long-term room temperature conditions for stability studies (i.e., 25±2°C,60±5% RH). Although adequate in the context of other guidance existing at that time, this recommendation is no longer sufficient to serve as a basis for stability testing for ANDAs. The following existing ICH guidances address stability for new drug substances and products:1.Q1A (R2) Stability Testing of New Drug Substances and Products.2.Q1B Photostability Testing of New Drug Substances and Products.3.Q1C Stability Testing for New Dosage Forms.1 This guidance has been prepared by the Office of Generic Drugs, Office of Pharmaceutical Science in the Center for Drug Evaluation and Research (CDER) at the Food and Drug Administration.4.Q1D Bracketing and Matrixing Designs for Stability Testing of New DrugSubstances and Products.5.Q1E Evaluation of Stability Data.2In the discussion that follows, these guidances are referred to as ICH stability guidances.III. DISCUSSIONAlthough the ICH stability guidances were developed by ICH to provide guidance on the information that should be provided in new drug applications to ensure the stability of new drug substances and drug products, we believe the recommendations also should be applied to ANDAs.When following the ICH stability recommendations, you, the applicant, should:1.Submit data from three pilot scale batches or two pilot scale batches and one smallscale batch. If the size of the pilot scale batch does not follow ICH recommendations,the applicant should provide a justification.2.At the time of submission, provide 6 months of data that include accelerated andlong-term conditions. FDA recommends following ICH guidelines with respect toutilization of intermediate conditions to support shelf-life.e multiple lots of drug substance as appropriate.4.Manufacture and package the drug product using principles that are representative ofthe commercial process.5.Provide a fully packaged primary batch.e drug product from all three primary batches when using bracketing and matrixingdesigns under ICH Q1D.7.Provide statistical analysis of the data as appropriate, in accordance with ICH Q1E,Appendix A.If you choose not to follow the above recommendations, you should provide FDA with a justification for the approach you intend to follow in your ANDA submissions to ensure stability.2 We update guidances periodically. To make sure you have the most recent version of these guidances, check the FDA Drugs guidance Web page at/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htm.。
FDA 行业指南 中英对照 待完成
Guidance for IndustryContainer Closure Systems for Packaging Human Drugs and Biologics Chemistry, Manufacturing and Controls Documentation行业指南人用药品及生物制品的包装容器和封装系统:化学,生产和控制文件指南发布者:美国FDA下属的CDER及CBER发布日期:May 1999TABLE OF CONTENTS目录I.INTRODUCTION介绍II.BACKGROUND 背景A.Definitions 定义B.CGMP, CPSC and USP Requirements on Containers and Closures. CGMP, CPSC和USP对容器和密封的要求C.Additional Considerations 其他需要考虑的事项III.QUALIFICATION AND QUALITY CONTROL OF PACKAGING COMPONENTS包装组件的合格要求以及质量控制A.Introduction 介绍B.General Considerations 通常要求rmation That Should Be Submitted in Support of an Original Application for Any DrugProduct 为支持任何药品的原始申请所必须提供的信息D.Inhalation Drug Products 吸入性药品E.Drug Products for Injection and Ophthalmic Drug Products 注射剂和眼科用药F.Liquid-Based Oral and Topical Drug Products and Topical Delivery Systems 液体口服和外用药品和外用给药系统G.Solid Oral Dosage Forms and Powders for Reconstitution 口服固体剂型和待重新溶解的粉末H.Other Dosage Forms 其他剂型IV.POSTAPPROVAL PACKAGING CHANGES 批准后的包装变更V.TYPE III DRUG MASTER FILES 药品主文件第III类A.General Comments 总体评述rmation in a Type III DMF 第III类DMF中包括的信息VI.BULK CONTAINERS 大包装容器A.Containers for Bulk Drug Substances 用于原料药的容器B.Containers for Bulk Drug Products 用于散装药品的容器ATTACHMENT A 附件AREGULATORY REQUIREMENTS 药政要求ATTACHMENT B 附件BCOMPLIANCE POLICY GUIDES THAT CONCERN PACKAGING 关于包装,所适用的政策指南ATTACHMENT C 附件CEXTRACTION STUDIES “提取性”研究ATTACHMENT D 附件DABBREVIATIONS 缩略语ATTACHMENT E 附件EREFERENCES 参考文献GUIDANCE FOR INDUSTRY1Container Closure Systems for Packaging Human Drugs and BiologicsChemistry, Manufacturing and Controls DocumentationI.INTRODUCTION介绍This document is intended to provide guidance on general principles2 for submitting information on packaging materials used for human drugs and biologics.3 This guidance supersedes the FDA Guideline for Submitting Documentation for Packaging for Human Drugs and Biologics , issued in February 1987 and the packaging policy statement issued in a letter to industry dated June 30, 1995 from the Office of Generic Drugs.4 This guidance is not intended to describe the information that should be providedabout packaging operations associated with drug product manufacture. 本文件目的是为递交人用药品和生物制品的包装信息提供总体原则指南。
FDA药物稳定性指导原则
Q1A(R2) Stability Testing of New Drug Substancesand ProductsU.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)Center for Biologics Evaluation and Research (CBER)November 2003ICHRevision 2Q1A(R2) Stability Testing of New Drug Substancesand ProductsAdditional copies are available from:Office of Training and CommunicationDivision of Drug Information, HFD-240Center for Drug Evaluation and ResearchFood and Drug Administration5600 Fishers LaneRockville, MD 20857(Tel) 301-827-4573/cder/guidance/index.htmorOffice of Communication, Training andManufacturers Assistance, HFM-40Center for Biologics Evaluation and ResearchFood and Drug Administration1401 Rockville Pike, Rockville, MD 20852-1448/cber/guidelines.htm.(Tel) Voice Information System at 800-835-4709 or 301-827-1800U.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)Center for Biologics Evaluation and Research (CBER)November 2003ICHRevision 2TABLE OF CONTENTSI. INTRODUCTION (1) (1)A. Objectives of the Guidance (1.1) (1)B. Scope of the Guidance (1.2) (2)C. General Principles (1.3) (2)II. GUIDANCE (2) (2)A. Drug Substance (2.1) (2)1. General (2.1.1) (2)2. Stress Testing (2.1.2) (3)3. Selection of Batches (2.1.3) (3)4. Container Closure System (2.1.4) (3)5. Specification (2.1.5) (3)6. Testing Frequency (2.1.6) (4)7. Storage Conditions (2.1.7) (4)8. Stability Commitment (2.1.8) (6)9. Evaluation (2.1.9) (7)B. Drug Product (2.2) (8)1. General (2.2.1) (8)2. Photostability Testing (2.2.2) (8)3. Selection of Batches (2.2.3) (8)4. Container Closure System (2.2.4) (8)5. Specification (2.2.5) (9)6. Testing Frequency (2.2.6) (9)7. Storage Conditions (2.2.7) (10)8. Stability Commitment (2.2.8) (14)9. Evaluation (2.2.9) (15)10. Statements/Labeling (2.2.10) (16)GLOSSARY (3) (17)REFERENCES (4) (21)ATTACHMENT List Of Revision 2 Changes (22)iGuidance for Industry1Q1A(R2) Stability Testing of New DrugSubstances and ProductsThis guidance represents the Food and Drug Administration's (FDA's) current thinking on this topic. It does not create or confer any rights for or on any person and does not operate to bind FDA or the public. You can use an alternative approach if the approach satisfies the requirements of the app licable statutes and regulations. If you want to discuss an alternative approach, contact the FDA staff responsible for implementing this guidance. If you cannot identify the appropriate FDA staff, call the appropriate number listed on the title page of this guidance.I. INTRODUCTION (1) 2This guidance is the second revision of Q1A Stability Testing of New Drug Substances and Products, which was first published in September 1994 and revised in August 2001. The purpose of this revision is to harmonize the intermediate storage condition for zones I and II with the long-term condition for zones III and IV recommended in the ICH guidance Q1F Stability Data Package for Registration Applications in Climatic Zones III and IV. The changes made in this second revision are listed in the attachment to this guidance.A. Objectives of the Guidance (1.1)This guidance is intended to define what stability data package for a new drug substance or drug product is sufficient for a registration application within the three regions of the European Union (EU), Japan, and the United States. It does not seek to address the testing for registration in or export to other areas of the world. The guidance exemplifies the core stability data package for new drug substances and products, but leaves sufficient flexibility to encompass the variety of different practical situations that may be encountered due to specific scientific considerations and characteristics of the materials being evaluated. Alternative approaches ca n be used when there are scientifically justifiable reasons.1This guidance was developed within the Expert Working Group (Quality) of the International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH) and has been subject to consultation by the regulatory parties, in accordance with the ICH process. This docume nt was endorsed by the ICH Steering Committee at Step 4 of the ICH process, February 2003. At S tep 4 of the process, the final draft is recommended for adoption to the regulatory bodies of the European Union, Japan, and the United States.2 Arabic numbers reflect the organizational breakdown in the document endorsed by the ICH Steering Committee at Step 4 of the ICH process.1B. Scope of the Guidance (1.2)The guidance addresses the information to be submitted in registration applications for new molecular entities and associated drug products. This guidance does n ot currently seek to cover the information to be submitted for abbreviated or abridged applications, variations, or clinical trial applications.Specific details of the sampling and testing for particular dosage forms in their proposed container closures are not covered in this guidance.Further guidance on new dosage forms and on biotechnological/biological products can be found in ICH guidances Q1C Stability Testing for New Dosage Forms and Q5C Quality of Biotechnological Products: Stability Testing of Biotechnological/Biological Products, respectively.C. General Principles (1.3)The purpose of stability testing is to provide evidence on how the quality of a drug substance or drug product varies with time under the influence of a variety of environmental factors, such as temperature, humidity, and light, and to establish a retest period for the drug substance or a shelf life for the drug product and recommended storage conditions.The choice of test conditions defined in this guidance is based on an analysis of the effects of climatic conditions in the three regions of the EU, Japan, and the United States. The mean kinetic temperature in any part of the world can be derived from climatic data, and the world can be divided into four climatic zones, I-IV. This guidance addresses climatic zones I and II. The principle has been established that stability information generated in any one of the three regions of the EU, Japan, and the United States would be mutually acceptable to the other two regions, provided the information is consistent with this guidance and the labeling is in accord with national/regional requirements.FDA's guidance documents, including this guidance, do not establish legally enforceable responsibilities. Instead, guidances descri be the Agency's current thinking on a topic and should be viewed only as recommendations, unless specific regulatory or statutory requirements are cited. The use of the word should in Agency guidances means that something is suggested or recommended, but not required.II. GUIDANCE (2)A. Drug Substance (2.1)1. General (2.1.1)Information on the stability of the drug substance is an integral part of the systematic approach to stability evaluation.22. Stress Testing (2.1.2)Stress testing of the drug substance can help identify the likely degradation products, which can in turn help establish the degradation pathways and the intrinsic stability of the molecule and validate the stability indicating power of the analytical procedures used. The nature o f the stress testing will depend on the individual drug substance and the type of drug product involved. Stress testing is likely to be carried out on a single batch of the drug substance. The testing should include the effect of temperatures (in 10°C increments (e.g., 50°C, 60°C) above that for accelerated testing), humidity (e.g., 75 percent relative humidity or greater) where appropriate, oxidation, and photolysis on the drug substance. The testing should also evaluate the susceptibility of the drug substance to hydrolysis across a wide range of pH values when in solution or suspension. Photostability testing should be an integral part of stress testing. The standard conditions for photostability testing are described in ICH Q1B Photostability Testing of New Drug Substances and Products.Examining degradation products under stress conditions is useful in establishing degradation pathways and developing and validating suitable analytical procedures. However, such examination may not be necessary for certain degradation products if it has been demonstrated that they are not formed under accelerated or long-term storage conditions.Results from these studies will form an integral part of the information provided to regulatory authorities.3. Selection of Batches (2.1.3)Data from formal stability studies should be provided on at least three primary batches of the drug substance. The batches should be manufactured to a minimum of pilot scale by the same synthetic route as production batches and using a method of manufacture and procedure that simulates the final process to be used for production batches. The overall quality of the batches of drug substance placed on formal stability studies should be representative of the quality of the material to be made on a production scale.Other supporting data can be provided.4. Container Closure System (2.1.4)The stability studies should be conducted on the drug substance packaged in a container closure system that is the same as or simulates the packaging proposed for storage and distribution.5. Specification (2.1.5)Specification, which is a list of tests, references to analytical procedures, and proposed acceptance criteria, is addressed in ICH Q6A Specifications: Test Procedures and Acceptance Criteria for New Drug Substances and New Drug Products: Chemical Substances and Q6B3Specifications: Test Procedures and Acceptance Criteria for New Drug Substances and New Drug Products: Biotechnological/Biological Products. In addition, specification for degradation products in a drug substance is discussed in ICH Q3A Impurities in New Drug Substances. Stability studies should include testing of those attributes of the drug substance that are susceptible to change during storage and are likely to influence qu ality, safety, and/or efficacy. The testing should cover, as appropriate, the physical, chemical, biological, and microbiological attributes. Validated stability-indicating analytical procedures should be applied. Whether and to what extent replication should be performed should depend on the results from validation studies.6. Testing Frequency (2.1.6)For long-term studies, frequency of testing should be sufficient to establish the stability profile of the drug substance. For drug substances with a proposed retest period of at least 12 months, the frequency of testing at the long-term storage condition should normally be every 3 months over the first year, every 6 months over the second year, and annually thereafter through the proposed retest period.At the accelerated storage condition, a minimum of three time points, including the initial and final time points (e.g., 0, 3, and 6 months), from a 6-month study is recommended. Where an expectation (based on development experience) exists that the results from accelerated studies are likely to approach significant change criteria, increased testing should be conducted either by adding samples at the final time point or including a fourth time point in the study design.When testing at the intermediate storage condition is called for as a result of significant change at the accelerated storage condition, a minimum of four time points, including the initial and final time points (e.g., 0, 6, 9, 12 months), from a 12-month study is recommended.7. Storage Conditions (2.1.7)In general, a drug substance should be evaluated under storage conditions (with appropriate tolerances) that test its thermal stability and, if applicable, its sensitivity to moisture. The storage conditions and the lengths of studies chosen should be sufficient to cover storage, shipment, and subsequent use.The long-term testing should cover a minimum of 12 months’ duration on at least three primary batches at the time of submission and should be continued for a period of time su fficient to cover the proposed retest period. Additional data accumulated during the assessment period of the registration application should be submitted to the authorities if requested. Data from the accelerated storage condition and, if appropriate, from the intermediate storage condition can be used to evaluate the effect of short-term excursions outside the label storage conditions (such as might occur during shipping).Long-term, accelerated, and, where appropriate, intermediate storage conditions for drug substances are detailed in the sections below. The general case should apply if the drug substance4is not specifically covered by a subsequent section. Alternative storage conditions can be used if justified.a. General case (2.1.7.1)Study Storage condition Minimum time period coveredby data at submission12 monthsLong-term* 25°C ± 2°C/60% RH ± 5% RHor30°C ± 2°C/65% RH ± 5% RHIntermediate** 30°C ± 2°C/65% RH ± 5% RH 6 monthsAccelerated 40°C ± 2°C/75% RH ± 5% RH 6 months* It is up to the applicant to decide whether long-term stability sturdies are performed at25°C ± 2°C/60% RH ± 5% RH or 30°C ± 2°C/65% RH ± 5% RH.** If 30°C ± 2°C/65% RH ± 5% RH is the long-term condition, there is no intermediate condition.If long-term studies are conducted at 25°C ± 2°C/60% RH ± 5% RH and significant change occurs at any time during 6 months’ testing at the accelerated storage condition, additional testing at the intermediate storage condition should be conducted and evaluated againstsigni ficant change criteria. Testing at the intermediate storage condition should include all tests, unless otherwise justified. The initial application should include a minimum of 6 months’ data from a 12-month study at the intermediate storage condition.Significant change for a drug substance is defined as failure to meet its specification.b. Drug substances intended for storage in a refrigerator (2.1.7.2)Study Storage condition Minimum time period coveredby data at submission Long-term 5°C ± 3°C 12 monthsAccelerated 25°C ± 2°C/60% RH ± 5% RH 6 monthsData from refrigerated storage should be assessed according to the evaluation section of this guidance, except where explicitly noted below.If significant change occurs between 3 and 6 months’ testing at the accelerated storage condition, the proposed retest period should be based on the real time data available at the long-term storage condition.If significant change occurs within the first 3 months’ testing at the accelerated storageconditi on, a discussion should be provided to address the effect of short-term excursions outside the label storage condition (e.g., during shipping or handling). This discussion can be supported, if appropriate, by further testing on a single batch of the drug substance for a period shorter than53 months but with more frequent testing than usual. It is considered unnecessary to continue to test a drug substance through 6 months when a significant change has occurred within the first 3 months.c. Drug substances intended for storage in a freezer (2.1.7.3)Study Storage condition Minimum time period coveredby data at submission Long-term -20°C ± 5°C 12 monthsFor drug substances intended for storage in a freezer, the retest period should be based on the real time data obtained at the long-term storage condition. In the absence of an accelerated storage condition for drug substances intended to be stored in a freezer, testing on a single batch at an elevated temperature (e.g., 5°C ± 3°C or 25°C ± 2°C) for an appropriate time period should be conducted to address the effect of short-term excursions outside the proposed label storage condition (e.g., during shipping or handling).d. Drug substances intended for storage below -20°C (2.1.7.4)Drug substances intended for storage below -20°C should be treated on a case-by-case basis.8. Stability Commitment (2.1.8)When available long-term stability data on primary batches do not cover the proposed retest period granted at the time of approval, a commitment should be made to continue the stability studies postapproval to firmly establish the retest period.Where the submission includes long-term stability data on three production batches covering the proposed retest period, a postapproval commitment is considered unnecessary. Otherwise, one of the following commitments should be made:• If the submission includes data from stability studies on at least three production batches, a commitment should be made to continue these studies through theproposed retest period.• If the submission includes data from stability studies on fewer than three production batches, a commitment should be made to continue these studiesthrough the proposed retest period and to place additional production batches, to atotal of at least three, on long-term stability studies through the proposed retestperiod.• If the submission does not include stability data on production batches, a commitment should be made to place the first three production batches on long-term stability studies through the proposed retest period.6The stability protocol used for long-term studies for the stability commitment should be the same as that for the primary batches, unless otherwise scientifically justified.9. Evaluation (2.1.9)The purpose of the stability study is to establish, based on testing a minimum of three batches of the drug substance and evaluating the stability information (including, as appropriate, results of the physical, chemical, biological, and microbiological tests), a retest period applicable to all future batches of the drug substance manufactured under similar circumstances. The degree of variability of individual batches affects the confidence that a future production batch will remain within specification throughout the assigned re test period.The data may show so little degradation and so little variability that it is apparent from looking at the data that the requested retest period will be granted. Under these circumstances, it is normally unnecessary to go through the formal statistical analysis; providing a justification for the omission should be sufficient.An approach for analyzing the data on a quantitative attribute that is expected to change with time is to determine the time at which the 95 percent, one-sided confidence limit for the mean curve intersects the acceptance criterion. If analysis shows that the batch-to-batch variability is small, it is advantageous to combine the data into one overall estimate. This can be done by first applying appropriate statistical tests (e.g., p values for level of significance of rejection of more than 0.25) to the slopes of the regression lines and zero time intercepts for the individual batches. If it is inappropriate to combine data from several batches, the overall retest period should be based on the minimum time a batch can be expected to remain within acceptance criteria.The nature of any degradation relationship will determine whether the data should be transformed for linear regression analysis. Usually the relationship ca n be represented by a linear, quadratic, or cubic function on an arithmetic or logarithmic scale. Statistical methods should be employed to test the goodness of fit of the data on all batches and combined batches (where appropriate) to the assumed degradation line or curve.Limited extrapolation of the real time data from the long-term storage condition beyond the observed range to extend the retest period can be undertaken at approval time if justified. This justification should be based, for example, o n what is known about the mechanism of degradation, the results of testing under accelerated conditions, the goodness of fit of any mathematical model, batch size, and/or existence of supporting stability data. However, this extrapolation assumes that the same degradation relationship will continue to apply beyond the observed data.Any evaluation should cover not only the assay, but also the levels of degradation products and other appropriate attributes.710. Statements/Labeling (2.1.10)A storage statement should be established for the labeling in accordance with relevantnational/regional requirements. The statement should be based on the stability evaluation of the drug substance. Where applicable, specific instructions should be provided, particul a rly for drug substances that cannot tolerate freezing. Terms such as ambient conditions or room temperature should be avoided.A retest period should be derived from the stability information, and a retest date should be displayed on the container label if appropriate.B. Drug Product (2.2)1. General (2.2.1)The design of the formal stability studies for the drug product should be based on knowledge of the behavior and properties of the drug substance, results from stability studies on the drug substance, and experience gained from clinical formulation studies. The likely changes on storage and the rationale for the selection of attributes to be tested in the formal stability studies should be stated.2. Photostability Testing (2.2.2)Photostability testing should be conducted on at least one primary batch of the drug product if appropriate. The standard conditions for photostability testing are described in ICH Q1B.3. Selection of Batches (2.2.3)Data from stability studies should be provided on at least three primary batches of the drug product. The primary batches should be of the same formulation and packaged in the same container closure system as proposed for marketing. The manufacturing process used for primary batches should simulate that to be applied to production batches and should provide product of the same quality and meeting the same specification as that intended for marketing. Two of the three batches should be at least pilot scale batches, and the third one can be smaller if justified. Where possible, batches of the drug product should be manufactured by using different batches of the drug substance.Stability studies should be performed on each individual strength and container size of the drug product unless bracketing or matrixing is applied.Other supporting data can be provided.4. Container Closure System (2.2.4)Stability testing should be conducted on the dosage form packaged in the container closure system proposed for marketing (including, as appropriate, any se condary packaging and8container label). Any available studies carried out on the drug product outside its immediate container or in other packaging materials can form a useful part of the stress testing of the dosage form or can be considered as supporting information, respectively.5. Specification (2.2.5)Specification, which is a list of tests, references to analytical procedures, and proposed acceptance criteria, including the concept of different acceptance criteria for release and shelf life specifications, is addressed in ICH Q6A and Q6B. In addition, specification for degradation products in a drug product is addressed in ICH Q3B Impurities in New Drug Products.Stability studies should include testing of those attributes of the drug product tha t are susceptible to change during storage and are likely to influence quality, safety, and/or efficacy. The testing should cover, as appropriate, the physical, chemical, biological, and microbiological attributes, preservative content (e.g., antioxidant, antimicrobial preservative), and functionality tests (e.g., for a dose delivery system). Analytical procedures should be fully validated and stability indicating. Whether and to what extent replication should be performed will depend on the results of validation studies.Shelf life acceptance criteria should be derived from consideration of all available stability information. It may be appropriate to have justifiable differences between the shelf life and release acceptance criteria based on the stability evaluation and the changes observed on storage. Any differences between the release and shelf life acceptance criteria for antimicrobial preservative content should be supported by a validated correlation of chemical content and preservative effectiveness demonstrated during drug development on the product in its final formulation (except for preservative concentration) intended for marketing. A single primary stability batch of the drug product should be tested for antimicrobial preservative effecti v eness (in addition to preservative content) at the proposed shelf life for verification purposes, regardless of whether there is a difference between the release and shelf life acceptance criteria for preservative content.6. Testing Frequency (2.2.6)For long-term studies, frequency of testing should be sufficient to establish the stability profile of the drug product. For products with a proposed shelf life of at least 12 months, the frequency of testing at the long-term storage condition should normally be every 3 months over the first year, every 6 months over the second year, and annually thereafter through the proposed shelf life.At the accelerated storage condition, a minimum of three time points, including the initial and final time points (e.g., 0, 3, and 6 months), from a 6-month study is recommended. Where an expectation (based on development experience) exists that results from accelerated testing are likely to approach significant change criteria, increased testing should be conducted either by adding samples at the final time point or by including a fourth time point in the study design.9When testing at the intermediate storage condition is called for as a result of significant change at the accelerated storage condition, a minimum of four time points, including the initial and final time points (e.g., 0, 6, 9, 12 months), from a 12-month study is recommended.Reduced designs (i.e., matrixing or bracketing), where the testing frequency is reduced or certain factor combinations are not tested at all, can be applied if justified.7. Storage Conditions (2.2.7)In general, a drug product should be evaluated under storage conditions (with appropriate tolerances) that test its thermal stability and, if applicable, its sensitivity to moisture or potential for solvent loss. The storage conditions and the lengths of studies chosen should be sufficient to cover storage, shipment, and subsequent use.Stability testing of the drug product after constitution or dilution, if applicable, should be conducted to provide information for the labeling on the preparation, storage condition, and in-use period of the constituted or diluted product. This testing should be performed on the constituted or diluted product through the proposed in-use period on primary batches as part of the formal stability studies at initial and final time points, and if full shelf life, long-term datawill not be available before submission, at 12 months or the last time point for which data will be available. In general, thi s testing need not be repeated on commitment batches.The long-term testing should cover a minimum of 12 months’ duration on at least three primary batches at the time of submission and should be continued for a period of time sufficient to cover the proposed shelf life. Additional data accumulated during the assessment period of the registration application should be submitted to the authorities if requested. Data from the accelerated storage condition and, if appropriate, from the intermediate storage condition can be used to evaluate the effect of short-term excursions outside the label storage conditions (such as might occur during shipping).Long-term, accelerated, and, where appropriate, intermediate storage conditions for drug products are detailed in the sections below. The general case should apply if the drug product is not specifically covered by a subsequent section. Alternative storage conditions can be used if justified.10。
FDA行业指南中英对照待完成
FDA行业指南中英对照待完成FDA Industry Guidance - FDA行业指南Introduction - 引言Scope - 范围This guidance applies to manufacturers, distributors, importers, and other participants in the food, drug, and medical device industries. - 本指南适用于食品、药品和医疗器械行业的制造商、分销商、进口商及其他参与者。
Definitions - 定义For the purposes of this guidance, the following definitions apply: - 为了本指南的目的,将适用以下定义:1. Food - 食品Any article used for food or drink for man or animals. - 任何用于人类或动物的食品或饮料。
2. Drug - 药品Any substance intended for use in the diagnosis, cure, treatment, or prevention of disease. - 任何用于诊断、治愈、治疗或预防疾病的物质。
3. Medical Device - 医疗器械Any instrument, apparatus, or device intended for use in the diagnosis, cure, mitigation, treatment, or prevention of disease. - 任何用于诊断、治愈、缓解、治疗或预防疾病的仪器、装置或设备。
4. Manufacturer - 制造商Product Quality Requirements - 产品质量要求Manufacturers should ensure that products meet the appropriate quality standards established by the FDA. - 制造商应确保产品符合FDA制定的适当质量标准。
FDA-guidance for industry-Mass Spectrometry for Confirmation of the Identity of Animal Drug Residues
U.S. Department of Health and Human Services Food and Drug Administration Center for Veterinary Medicine June 6.2001
-
Draft Guidance for Industry: Mass Spectrometty for Confirmation of the Identity of Animal Drug Residues
118
Guidance for Industry
Mass, Spectrometty for Confirmation of the Identity of Animal Drug Residues
DRAFT GUIDANCE
This draft guidance
document
is being distቤተ መጻሕፍቲ ባይዱibuted
2
submitted to CVM: The document should also help in making decisions about appropriate methodology in various regulatory situations and ensuring consistency in work do,ne for CVM’s purposes. This document sets guidance standards and performance specifications as-targets. CVM recommends that methods meet or exceed these standards.
This draft document is intended to provide specific guidance for the development, evaluation, and application of mass specfromefric methods for confirming the identify of animal drtig residues. If represents the current thinking of the FDA Center for Veterinary Medicine on this matter. If does not create or confer any rights for or on any person and does not operate to bind the FDA or public. An alternative approach may be used if such approach satisfies the requirements of the applicable statute and regulations.
最新FDA 行业指南 中英对照 待完成
F D A行业指南中英对照待完成Guidance for IndustryContainer Closure Systems for Packaging Human Drugs and Biologics Chemistry, Manufacturing and Controls Documentation行业指南人用药品及生物制品的包装容器和封装系统:化学,生产和控制文件指南发布者:美国FDA下属的CDER及CBER发布日期:May 1999TABLE OF CONTENTS目录I. INTRODUCTION介绍II. BACKGROUND 背景A. Definitions 定义B. CGMP, CPSC and USP Requirements on Containers and Closures. CGMP, CPSC和USP对容器和密封的要求C. Additional Considerations 其他需要考虑的事项III. QUALIFICATION AND QUALITY CONTROL OF PACKAGING COMPONENTS包装组件的合格要求以及质量控制A. Introduction 介绍B. General Considerations 通常要求C. Information That Should Be Submitted in Support of an Original Application for AnyDrug Product 为支持任何药品的原始申请所必须提供的信息D. Inhalation Drug Products 吸入性药品E. Drug Products for Injection and Ophthalmic Drug Products 注射剂和眼科用药F. Liquid-Based Oral and Topical Drug Products and Topical Delivery Systems 液体口服和外用药品和外用给药系统G. Solid Oral Dosage Forms and Powders for Reconstitution 口服固体剂型和待重新溶解的粉末H. Other Dosage Forms 其他剂型IV. POSTAPPROVAL PACKAGING CHANGES 批准后的包装变更V. TYPE III DRUG MASTER FILES 药品主文件第III类A. General Comments 总体评述B. Information in a Type III DMF 第III类DMF中包括的信息VI. BULK CONTAINERS 大包装容器A. Containers for Bulk Drug Substances 用于原料药的容器B. Containers for Bulk Drug Products 用于散装药品的容器ATTACHMENT A 附件AREGULATORY REQUIREMENTS 药政要求ATTACHMENT B 附件BCOMPLIANCE POLICY GUIDES THAT CONCERN PACKAGING 关于包装,所适用的政策指南ATTACHMENT C 附件CEXTRACTION STUDIES “提取性”研究ATTACHMENT D 附件DABBREVIATIONS 缩略语ATTACHMENT E 附件EREFERENCES 参考文献GUIDANCE FOR INDUSTRY1Container Closure Systems for Packaging Human Drugs and Biologics Chemistry, Manufacturing and Controls DocumentationI.INTRODUCTION介绍This document is intended to provide guidance on general principles2 for submitting information on packaging materials used for human drugs and biologics.3 This guidance supersedes theFDA Guideline for Submitting Documentation for Packaging for Human Drugs and Biologics ,issued in February 1987 and the packaging policy statement issued in a letter to industry dated June 30, 1995 from the Office of Generic Drugs.4 This guidance is not intended to describe the information that should be provided about packaging operations associated with drug productmanufacture. 本文件目的是为递交人用药品和生物制品的包装信息提供总体原则指南。
FDA 行业指南 中英对照 待完成
Container Closure Systems for Packaging Human Drugs and Biologics Chemistry, Manufacturing and Controls Documentation行业指南人用药品及生物制品的包装容器和封装系统:化学,生产和控制文件指南发布者:美国FDA下属的CDER及CBER发布日期:May 1999TABLE OF CONTENTS目录I.INTRODUCTION介绍II.BACKGROUND 背景A.Definitions 定义B.CGMP, CPSC and USP Requirements on Containers and Closures. CGMP, CPSC和USP对容器和密封的要求C.Additional Considerations 其他需要考虑的事项III.QUALIFICATION AND QUALITY CONTROL OF PACKAGING COMPONENTS包装组件的合格要求以及质量控制A.Introduction 介绍B.General Considerations 通常要求rmation That Should Be Submitted in Support of an Original Application forAny Drug Product 为支持任何药品的原始申请所必须提供的信息D.Inhalation Drug Products 吸入性药品E.Drug Products for Injection and Ophthalmic Drug Products 注射剂和眼科用药F.Liquid-Based Oral and Topical Drug Products and Topical Delivery Systems 液体口服和外用药品和外用给药系统G.Solid Oral Dosage Forms and Powders for Reconstitution 口服固体剂型和待重新溶解的粉末H.Other Dosage Forms 其他剂型IV.POSTAPPROVAL PACKAGING CHANGES 批准后的包装变更V.TYPE III DRUG MASTER FILES 药品主文件第III类A.General Comments 总体评述rmation in a Type III DMF 第III类DMF中包括的信息VI.BULK CONTAINERS 大包装容器A.Containers for Bulk Drug Substances 用于原料药的容器B.Containers for Bulk Drug Products 用于散装药品的容器ATTACHMENT A 附件AREGULATORY REQUIREMENTS 药政要求ATTACHMENT B 附件BCOMPLIANCE POLICY GUIDES THAT CONCERN PACKAGING 关于包装,所适用的政策指南ATTACHMENT C 附件CEXTRACTION STUDIES “提取性”研究ATTACHMENT D 附件DABBREVIATIONS 缩略语ATTACHMENT E 附件EREFERENCES 参考文献GUIDANCE FOR INDUSTRY1Container Closure Systems for Packaging Human Drugs and Biologics Chemistry, Manufacturing and Controls DocumentationI.INTRODUCTION介绍This document is intended to provide guidance on general principles2 for submitting information on packaging materials used for human drugs and biologics.3 This guidance supersedes the FDA Guideline for Submitting Documentation for Packaging for Human Drugs and Biologics , issued in February 1987 and the packaging policy statement issued in a letter to industry dated June 30, 1995 from the Office of Generic Drugs.4 This guidance is not intended to describe the information that should be provided about packaging operations associated with drug product manufacture. 本文件目的是为递交人用药品和生物制品的包装信息提供总体原则指南。
FDA Guidance for Industry Botanical Drug Products
Guidance for Industry Botanical Drug ProductsU.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)June 2004ChemistryGuidance for Industry Botanical Drug ProductsCopies of this Guidance are available from:Division of Drug Information (HFD-240),Office of Training and Communications,Center for Drug Evaluation and Research (CDER),Food and Drug Administration5600 Fishers Lane, Rockville, MD 20857, (Tel) 301-827-4573Internet at /cder/guidance/index.htmU.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)June 2004ChemistryTABLE OF CONTENTSI.INTRODUCTION (1)II.BACKGROUND (2)III.GENERAL REGULATORY APPROACHES (3)A.Marketing Under OTC Drug Monograph Versus Approved NDA (4)B.CMC Information for Botanical Drug Products (5)C.CMC and Toxicology Information to Support Initial Studies (5)D.Applicability of Combination Drug Regulations (6)IV.MARKETING A BOTANICAL DRUG UNDER AN OTC DRUG MONOGRAPH (6)V.MARKETING A BOTANICAL DRUG UNDER AN NDA (7)VI.INDS FOR BOTANICAL DRUGS (7)A.IND Information for Different Categories of Botanicals (7)B.Basic Format for INDs (9)1.Cover Sheet (see § 312.23(a)(1)) (9)2.Table of Contents (see § 312.23(a)(2)) (9)3.Introductory Statement and General Investigational Plan (see § 312.23(a)(3)) (9)4.Investigator′s Brochure (see § 312.23(a)(5)) (9)5.Protocols (§ 312.23(a)(6)) (9)6.Chemistry, Manufacturing, and Controls (§ 312.23(a)(7)) (10)7.Pharmacological and Toxicological Information (§ 312.23(a)(8)) (13)8.Previous Human Experience With the Product (§ 312.23(a)(9)) (13)VII.INDS FOR PHASE 1 AND PHASE 2 CLINICAL STUDIES OF LAWFULLY MARKETED BOTANICAL PRODUCTS WITHOUT SAFETY CONCERNS (13)A.Description of Product and Documentation of Human Use (13)1.Description of Botanicals Used (§ 312.23(a)(3)(i)) (14)2.History of Use (§ 312.23(a)(3)(ii),(a)(9)) (14)3.Current Marketed Use (§ 312.23(a)(3)(ii), (a)(9)) (14)B.Chemistry, Manufacturing, and Controls (14)1.Botanical Raw Material (§ 312.23(a)(7)(i)) (14)2.Botanical Drug Substance (§ 312.23(a)(7)(iv)(a)) (15)3.Botanical Drug Product (§ 312.23(a)(7)(iv)(b)) (15)4.Animal Safety Test (§ 312.23(a)(8)) (16)5.Placebo (§ 312.23(a)(7)(iv)(c)) (16)beling (§ 312.23(a)(7)(iv)(d)) (16)7.Environmental Assessment or Claim of Categorical Exclusion (§ 312.23(a)(7)(iv)(e)) (17)C.Pharmacology/Toxicology Information (17)1.All Marketed Botanical Products (17)2.Foreign-Marketed Botanical Products (18)D.Bioavailability (18)E.Clinical Considerations (18)VIII.INDS FOR PHASE 1 AND PHASE 2 CLINICAL STUDIES FOR NONMARKETED BOTANICAL PRODUCTS AND PRODUCTS WITH KNOWN SAFETY CONCERNS (19)A.Description of Product and Documentation of Human Use (19)1.Description of Botanicals Used (§ 312.23(a)(3)(i)) (19)2.History of Use (If Any) (§ 312.23(a)(3)(ii), (a)(9)) (19)3.Current Investigational Use (If Any) (§ 312.23(a)(3)(ii), (a)(9)) (20)B.Chemistry, Manufacturing, and Controls (20)1.Botanical Raw Material (§ 312.23(a)(7)(i)) (20)2.Botanical Drug Substance (§ 312.23(a)(7)(iv)(a)) (21)3.Botanical Drug Product (§ 312.23(a)(7)(iv)(b)) (23)4.Placebo (see section VII.B.5) (25)beling (see section VII.B.6) (25)6.Environmental Assessment or Claim of Categorical Exclusion (25)C.Nonclinical Safety Assessment (25)1.Traditional Preparations (25)2.Others (26)3.Products with Known Safety Issues (26)D.Bioavailability (26)E.Clinical Considerations (27)IX.INDS FOR PHASE 3 CLINICAL STUDIES OF ALL BOTANICAL PRODUCTS (27)A.Description of Product and Documentation of Human Experience (27)B.Chemistry, Manufacturing, and Controls (28)1.Expanded Clinical Studies (28)2.End-of-Phase 3 Clinical Studies and Pre-NDA Considerations (32)C.Nonclinical Safety Assessment (34)1.Repeat-Dose General Toxicity Studies (35)2.Nonclinical Pharmacokinetic/Toxicokinetic Studies (35)3.Reproductive Toxicology (36)4.Genotoxicity Studies (36)5.Carcinogenicity Studies (36)6.Special Pharmacology/Toxicology Studies (37)7.Regulatory Considerations (37)D.Bioavailability and Clinical Pharmacology (37)E.Clinical Considerations (38)GLOSSARY (39)QUESTIONS AND ANSWERS (42)ATTACHMENT A: REGULATORY APPROACHES FOR MARKETING BOTANICAL DRUG PRODUCTS (47)Guidance for Industry1Botanical Drug ProductsThis guidance represents the Food and Drug Administration's (FDA's) current thinking on this topic. It does not create or confer any rights for or on any person and does not operate to bind FDA or the public. An alternative approach may be used if such approach satisfies the requirements of the applicable statutes and regulations. If you want to discuss an alternative approach, contact the FDA staff responsible for implementing this guidance. If you cannot identify the appropriate FDA staff, call the appropriate number listed on the title page of this guidance.I.INTRODUCTIONThis guidance explains when a botanical drug may be marketed under an over-the-counter (OTC) drug monograph and when FDA regulations require approval for marketing of a new drug application (NDA), submitted under section 505(b) of the Federal Food, Drug, and Cosmetic Act (the Act), 21 U.S.C. 355(b). In addition, this document provides sponsors with guidance on submitting investigational new drug applications (INDs) for botanical drug products, including those botanical products (or botanicals) currently lawfully marketed as foods (including conventional foods and dietary supplements) in the United States.This guidance also discusses several areas in which, because of the unique nature of botanicals, FDA finds it appropriate to apply regulatory policies that differ from those applied to synthetic, semisynthetic, or otherwise highly purified or chemically modified drugs (including antibiotics derived from microorganisms). This latter group of drug substances is referred to in this guidance as synthetic or highly purified drugs. Therefore, when the recommendations on a specific topic discussed in this guidance differ from those in other existing guidances (e.g., Submitting Supporting Documentation in Drug Applications for the Manufacture of Drug Substances, 1987), 2 this guidance takes precedence. In particular, this guidance states that applicants may submit reduced documentation of nonclinical (preclinical) safety and of chemistry, manufacturing, and controls (CMC) to support an IND for initial clinical studies of 1This guidance has been prepared by working groups in the Medical Policy, Pharmacology and Toxicology, and Complex Drug Substances Coordinating Committees in the Center for Drug Evaluation and Research (CDER) at the Food and Drug Administration (FDA).2 FDA has issued a draft guidance entitled Drug Substance: Chemistry, Manufacturing, and Controls Information, which, when finalized, will replace the 1987 guidance (see 69 FR 929, January 7, 2004).botanicals that have been legally marketed in the United States and/or a foreign country as dietary supplements without any known safety concerns.FDA's guidance documents, including this guidance, do not establish legally enforceable responsibilities. Instead, guidances describe the Agency's current thinking on a topic and should be viewed only as recommendations, unless specific regulatory or statutory requirements are cited. The use of the word should in Agency guidances means that something is suggested or recommended, but not required.II.BACKGROUNDBotanical products are finished, labeled products that contain vegetable matter as ingredients.3 A botanical product may be a food (including a dietary supplement), a drug (including a biological drug), a medical device (e.g., gutta-percha), or a cosmetic under the Act. An article is generally a food if it is used for food (21 U.S.C. 312(f)(1)). Whether an article is a drug, medical device, or cosmetic under the Act turns on its “intended use” (21 U.S.C. 312(g)(1)(B) and (C), (h)(2) and (3), (i)). “Intended use” is created by claims made by or on behalf of a manufacturer or distributor of the article to prospective purchasers, such as in advertising, labeling, or oral statements.For the purposes of this document, the term botanicals includes plant materials, algae, macroscopic fungi, and combinations thereof. It does not include:•Materials derived from genetically modified botanical species (i.e., by recombinant DNA technology or cloning).•Fermentation products (i.e., products produced by fermentation of yeast, bacteria, and other microscopic organisms, including when plants are used as a substrate, and products produced by fermentation of plant cells), even if such products are previously approvedfor drug use or accepted for food use in the United States (e.g., antibiotics, amino acids, and vitamins).•Highly purified substances (e.g., paclitaxel) or chemically modified substances (e.g., estrogens synthesized from yam extracts) derived from botanical sources.This guidance addresses all botanical drug products (in all dosage forms) that are regulated under the Act, except those also regulated under section 351 of the Public Health Service Act (42U.S.C. 262). Although this guidance does not address drugs that contain animals or animal parts (e.g., insects, annelids, shark cartilage) and/or minerals, either alone or in combination with botanicals, many scientific principles described in this guidance may also apply to those products. When a drug product contains botanical ingredients in combination with either (1) a synthetic or highly purified drug or (2) a biotechnology derived or other naturally derived drug, this guidance only applies to the botanical portion of the product.3Botanical product and other terms used in this guidance are defined in the Glossary for use in this guidance only; these definitions may not be appropriate in other contexts.III.GENERAL REGULATORY APPROACHESMany botanical products are used widely in the United States. Depending on its labeling and intended use, a botanical product can be a food, a dietary supplement, and/or a drug. Botanicals used for food and consumed primarily for their taste, aroma, or nutritive value (e.g., lettuce, herbs used as seasonings) are regulated as foods. Botanicals can also be dietary supplements if they are labeled as dietary supplements and otherwise meet the dietary supplement definition in section 201(ff) of the Act (21 U.S.C. 321(ff)).If a botanical product is intended for use in diagnosing, mitigating, treating, or curing disease, it is a drug under section 201(g)(1)(B) of the Act and is subject to regulation as such. If a botanical product is intended to prevent disease, it is also a drug under section 201(g)(1)(B), except that a product that bears a health claim authorized in accordance with section 403(r) of the Act (21 U.S.C. 343(r)) is not a drug solely because its labeling contains such a claim. If the intended use of a botanical product is to affect the structure or function of the human body, it may be regulated either as a dietary supplement or as a drug, depending on the circumstances.Under the Dietary Supplement Health and Education Act of 1994 (DSHEA), an orally ingested product that meets the definition of a “dietary supplement” under section 201(ff) of the Act may be lawfully marketed with a statement that (1) claims a benefit related to a classical nutrient deficiency disease (and discloses the prevalence of the disease in the United States), (2) describes how the product is intended to affect the structure or function of the human body, (3) characterizes the documented mechanism by which the product acts to maintain such structure or function, or (4) describes general well-being from consumption of the product (section403(r)(6)(A) of the Act).4 A dietary supplement statement of the type described above may not claim to diagnose, mitigate, treat, cure, or prevent a specific disease or class of diseases (section 403(r)(6) of the Act).5If a botanical product is intended to affect the structure or function of the body but does not meet the definition of a dietary supplement, or does not meet the requirements for making a structure/function claim under section 403(r)(6) of the Act, it is subject to regulation as a drug under section 201(g)(1)(C) of the Act. As noted above, a botanical product is subject to regulation as a drug under section 201(g)(1)(B) of the Act if it is intended for use in diagnosing, mitigating, treating, curing, or preventing disease (except for a product marketed with certain health claims authorized under section 403(r) of the Act). Under section 505(b) of the Act, a 4The manufacturer must have substantiation that such statement is truthful and not misleading (section403(r)(6)(B) of the Act) and must notify FDA that the statement is being used no later than 30 days after the first marketing of the dietary supplement with the statement (section 403(r)(6) of the Act). In addition, the statement must be accompanied by the following disclaimer: “This statement has not been evaluated by the Food and Drug Administration. This product is not intended to diagnose, treat, cure, or prevent any disease” (section 403(r)(6)(C) of the Act). FDA regulations at 21 CFR 101.93(b)-(e) prescribe the required format and placement of the disclaimer in dietary supplement labeling.5FDA regulations at § 101.93(g) define disease for purposes of this provision and set forth what types of statements FDA will consider to be claims to diagnose, mitigate, treat, cure, or prevent disease.drug must be marketed under an approved NDA6 unless the product is excluded from the definition of a new drug under section 201(p) of the Act. Certain products that FDA determines are generally recognized as safe and effective in accordance with section 201(p) may be marketed under FDA′s OTC drug monograph system.A.Marketing Under OTC Drug Monograph Versus Approved NDAA botanical drug product may be marketed in the United States under (1) an OTC drugmonograph or (2) an approved NDA or ANDA. A botanical product that has beenmarketed in the United States for a material time and to a material extent for a specificOTC drug indication may be eligible for inclusion in an OTC drug monograph codifiedin21 CFR parts 331-358. The manufacturer would need to submit a petition in accordancewith 21 CFR 10.30 to amend the monograph to add the botanical substance as a newactive ingredient.Under current regulations, if there is no marketing history in the United States or aforeign country for a botanical drug product,7 if available evidence of safety andeffectiveness does not warrant inclusion of the product in an OTC drug monograph, or if the proposed indication would not be appropriate for nonprescription use, themanufacturer must submit an NDA to obtain FDA approval to market the product for the proposed use (sections 201(p) and 505 of the Act). An NDA for a botanical drug couldseek approval for either prescription or OTC use, depending on the indication andcharacteristics of the product and whether it is safe for use outside of the supervision of a practitioner licensed by law to administer it. If existing information on the safety andeffectiveness of a botanical drug product is insufficient to support an NDA, werecommend that new clinical studies be conducted to demonstrate safety andeffectiveness.8When a final OTC drug monograph is published for a specific use of a botanical drug,any person may market a product containing the same substance and for the same use,provided the labeling and other active ingredients (if present) are in accord with allrelevant monographs and other applicable regulations. In contrast, when a product isapproved under an NDA, the approval is specific to the drug product that is the subject of the application (the applicant’s drug product), and the applicant may be eligible for6Under section 505(j) of the Act, a botanical drug product may also be marketed as a generic drug under an abbreviated new drug application (ANDA). The generic version of the previously approved drug would have to be both pharmaceutically equivalent and bioequivalent to such drug. For information on the submission of ANDAs, see FDA regulations in 21 CFR parts 314 and 320 as well as Agency guidance documents.7FDA has issued a final rule that establishes criteria and procedures by which conditions may become eligible for inclusion in the OTC drug monograph system (67 FR 3060, January 23, 2002). Among other things, the final rule addresses how FDA considers foreign marketing data in determining whether a drug has been used under particular conditions to a material extent and for a material time (as required under section 201(p) of the Act) to qualify for inclusion in an OTC drug monograph.8See 21 CFR 312.20 (concerning requirement for an IND).marketing exclusivity for either 5 years (if it is a new chemical entity) or 3 years from the time of approval, even in the absence of patent protection. A new botanical drug (containing multiple chemical constituents) may qualify as a new chemical entity under § 314.108(a). If a product qualifies as a new chemical entity, during the period of exclusivity, FDA will not approve, or in some cases even review, certain competitor products unless the second sponsor conducts all studies necessary to demonstrate the safety and effectiveness of its product and submits a 505(b)(1) application. Therefore, if a person wishing to market a botanical drug product that is not included in an existing OTC drug monograph desires marketing exclusivity for the product, the person should seek approval of an NDA rather than petition the Agency to amend a monograph. Attachment A contains a schematic showing different regulatory approaches that can be taken for marketing botanical drug products in the United States, including OTC drug monograph and NDA procedures.B.CMC Information for Botanical Drug ProductsBotanical drug products have certain unique characteristics that should be taken into account in the application of FDA regulations and guidance. Botanical drugs are derived from vegetable matter and are usually prepared as complex mixtures. Their chemical constituents are not always well defined. In many cases, the active constituent in a botanical drug is not identified, nor is its biological activity well characterized. Therefore, the CMC documentation that should be provided for botanical drugs will often be different from that for synthetic or highly purified drugs, whose active constituents can be more readily chemically identified and quantified. For example, FDA would expect an NDA for a synthetic or highly purified drug to identify the active ingredient. However, it would not be essential for the sponsor of a botanical drug to identify the active constituents (although FDA recommends that this be done if feasible). Even if the sponsor were to eventually identify the active constituents in the NDA, the active constituents might not be identified during the IND stage.Because of the complex nature of a typical botanical drug and the lack of knowledge of its active constituent(s), FDA may rely on a combination of tests and controls to ensure the identity, purity, quality, strength, potency, and consistency of botanical drugs. These tests and controls include (1) multiple tests for drug substance and drug product (e.g., spectroscopic and/or chromatographic fingerprints, chemical assay of characteristic markers, and biological assay), (2) raw material and process controls (e.g., strict quality controls for the botanical raw materials and adequate in-process controls), and (3) process validation (especially for the drug substance).C.CMC and Toxicology Information to Su pport Initial StudiesMany botanical products are legally available in the United States as dietary supplements. Given the wide availability of such products outside of clinical trials, it is important to assess the effectiveness of such products. To support initial clinical trials, the nonclinical pharmacology and toxicology information that must be provided under 21 CFR 312.22(b) for legally available botanical products with no known safety issues (seesection VI.A) may be markedly reduced compared to that expected for synthetic orhighly purified new drugs that are not legally marketed and for which there is no priorhuman experience. In most cases, additional toxicology and CMC data will not berequired for such initial trials.D.Applicability of Combination Drug RegulationsBotanical drug products that are derived from a single part of a plant (e.g., leaves, stems, roots, or seeds), or from a single species of alga or macroscopic fungus (e.g., amushroom), are not considered to be fixed-combination drugs within the meaning of21 CFR 300.50 and 330.10(a)(4)(iv). Consequently, they do not have to meet therequirements for combination drugs, principally the need to demonstrate that eachcomponent or active ingredient makes a contribution to claimed effects.Botanical drugs composed of multiple parts of a single species of plant, alga, ormacroscopic fungus, or of parts from different species of plants algae, or macroscopicfungi, currently are subject to the combination drug requirements. However, FDA isconsidering revising its regulations to allow for the exemption of such botanical drugsfrom application of the combination drug requirements under certain circumstances. IV.MARKETING A BOTANICAL DRUG UNDER AN OTC DRUG MONOGRAPHA botanical product that has been marketed in the United States for a material time and to a material extent for a specific OTC indication may be eligible for consideration in the OTC drug monograph system. Currently, there are several botanical drugs, including cascara, psyllium, and senna, that are included in the OTC drug review. For a botanical drug substance to be included in an OTC drug monograph, there must be published data establishing general recognition of safety and effectiveness, usually including results of adequate and well-controlled clinical studies (see §§ 314.126(b) and 330.10). Requirements related to safety, effectiveness, and labeling for drugs to be included in an OTC drug monograph are set forth in 21 CFR part 330.A request to amend an OTC drug monograph to include a botanical substance must be submitted by citizen petition in accordance with §§ 10.30 and 330.10(a)(12). There should be publicly available quality standards for such a botanical drug substance in the drug section (i.e., not in the National Formulary or other nondrug sections) of the United States Pharmacopeia (USP).9 In the absence of a USP drug monograph, the petitioner should include suitable quality standards for the botanical drug substance in its citizen petition and simultaneously propose adoption of those standards in the USP. Additional criteria and procedures by which a botanical drug substance may become eligible for inclusion in the OTC drug monograph system are set forth in § 330.14. FDA regulations on current good manufacturing practices (CGMPs) apply to all OTC drug monograph products, including any listed botanical drug products (see § 330.1(a)).9However, a botanical drug’s conformance to the standards of the USP or any other official compendium does not establish that the botanical is safe, effective, and not misbranded for its intended use as a drug.For further information on the OTC drug monograph approach to marketing a botanical drug product, sponsors are encouraged to contact CDER′s Division of Over-the-Counter Drug Products (HFD-560).V.MARKETING A BOTANICAL DRUG UNDER AN NDAA botanical drug product that is not generally recognized as safe and effective for its therapeutic claims is considered a new drug under section 201(p) of the Act. Section 505(a) of the Act requires any person wishing to market a botanical drug product that is a new drug to obtain FDA approval of an NDA or ANDA for that product. According to section 505(d) of the Act and§ 314.50, an NDA must contain substantial evidence of effectiveness derived from adequate and well-controlled clinical studies, evidence of safety, and adequate CMC information. The format of an NDA submission and the requirements for its various sections are set forth in part 314 and discussed in several CDER guidance documents.VI.INDS FOR BOTANICAL DRUGSIf available information is insufficient to support an NDA for a botanical drug, the sponsor will need to develop further data. An IND is required under section 505(i) of the Act and21 CFR part 312 (unless exempt under § 312.2(b)) when a botanical product is studied in the United States for a drug use (see section 201(g) of the Act), even if such study is intended solely for research purposes. Under § 312.22, an IND must contain sufficient information to demonstrate that the drug product is safe for testing in humans and that the clinical protocol is properly designed for its intended objectives.A.IND Information for Different Categories of BotanicalsUnder § 312.22(b), the amount of information that must be submitted in an IND for aparticular drug product depends on, among other things, the novelty of the drug, theextent to which it has been studied previously, the drug product′s known or suspectedrisks, and the developmental phase of the drug. Sections VII and VIII of this guidancedescribe the information that we recommend a sponsor provide in meeting therequirements in § 312.23 for an IND for initial (i.e., phase 1 and phase 2) clinical studies of a botanical drug. As noted above, for botanicals legally marketed under the DSHEA, there will often be very little new CMC or toxicological data needed to initiate suchtrials, as long as there are no known safety issues associated with the product and it is to be used at approximately the same doses as those currently or traditionally used orrecommended. A botanical drug is considered to have a known safety issue when FDA has evidence that it produces serious and/or possibly life-threatening effects. Nonclinical evaluation to characterize toxicities may be appropriate for products with known safety issues. For example, nonclinical data may be appropriate to help establish safe doses and to determine ways to better monitor potential toxicities in humans. Such nonclinicalstudies may be needed early in development (see § 312.23(a)(8)).Properly conducted early clinical investigations, including controlled effectiveness trials in phase 2, will allow a determination of whether there is a clinical effect worth pursuing and will provide a more systematic evaluation of safety than previously available. If a botanical drug product shows promise of effectiveness in such early trials, the potential for wider use for particular purposes will create a need for greater assurance of product quality and consistency and for expanded (i.e., phase 3) clinical studies of safety and effectiveness (§ 312.22(b)). IND information appropriate for expanded clinical studies of botanical drugs is discussed in section IX.Under § 312.22(b), the IND sponsor of a botanical product that has been previously marketed but not in the United States must provide sufficient additional information to assist FDA in determining the safety of the product for use in initial clinical studies (section VII). Such additional information is appropriate under that regulation because these products are not already marketed in the United States and evidence of safety should be provided before patients are exposed to them.This guidance also addresses the type of information that should be provided under§ 312.22 in INDs for initial studies on botanical products that have not been lawfully marketed anywhere or have known safety issues (section VIII). In contrast to botanical products that have been marketed in some form, considerably less information may be available on the safety of a new botanical product that has not been marketed anywhere as a food or dietary supplement and has not been tested as a drug in humans. Consequently, it is appropriate that, under § 312.22(b), sponsors of INDs for initial trials of botanical products that have not previously been lawfully marketed anywhere, or for which there are known safety issues, provide certain additional information to FDA. The information to be provided in an IND for a botanical drug product is illustrated schematically in Attachment B and discussed in this section and sections VII-IX below. FDA encourages sponsors of INDs for initial studies of botanical drugs to seek input from CDER review divisions (organized based on the therapeutic classes of the drugs) to ensure that the appropriate information is submitted and that the clinical protocols are well designed. Many guidance documents specific to particular indications or dosage forms are also available from the respective review divisions.FDA may place an IND for initial studies of a botanical drug on clinical hold (i.e., an order issued by the Agency to delay a proposed clinical study) if it finds that the IND does not contain sufficient information required under § 312.23 to assess the risk to subjects of the proposed studies (§ 312.42(b)(1)(iv)). However, the lack of any specific item of information listed in § 312.23 for a phase 1 study will not necessarily justify imposing a clinical hold. Possible grounds for a clinical hold are set forth in § 312.42(b) and discussed in CDER′s guidance for industry on Content and Format of Investigational New Drug Applications (INDs) for Phase 1 Studies of Drugs, Including Well-Characterized, Therapeutic, Biotechnology-derived Products (November 1995).。
FDA行业指南 委托生产质量协议 中英文
Contract Manufacturing Arrangements for Drugs: Quality AgreementsGuidance for Industry行业指南:药品委托生产安排:质量协议U.S. Department of Health and Human Services Food and Drug AdministrationCenter for Drug Evaluation and Research (CDER)Center for Biologics Evaluation and Research (CBER)Center for Veterinary Medicine (CVM)November 2016Pharmaceutical Quality/Manufacturing Standards (CGMP)Guidance for IndustryAdditional copies are available from:Office of Communications, Division of Drug Information Center for Drug Evaluation and ResearchFood and Drug Administration10001 New Hampshire Ave., Hillandale Bldg., 4th Floor Silver Spring, MD 20993-0002Phone: 855-543-3784 or 301-796-3400; Fax: 301-431-6353Email: druginfo@/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htmand/orOffice of Communication, Outreach and Development Center for Biologics Evaluation and ResearchFood and Drug Administration10903 New Hampshire Ave., Bldg. 71, Room 3128Silver Spring, MD 20993-0002Phone: 800-835-4709 or 240-402-8010Email: ocod@/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/default.htmand/orPolicy and Regulations Staff, HFV-6Center for Veterinary Medicine Food and Drug Administration7519 Standish Place, Rockville, MD 20855/AnimalVeterinary/GuidanceComplianceEnforcement/GuidanceforIndustry/default.htmU.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER)Center for Biologics Evaluation and Research (CBER)Center for Veterinary Medicine (CVM)November 2016Pharmaceutical Quality/Manufacturing Standards (CGMP)TABLE OF CONTENTS 目录TABLE OF CONTENTS 目录 (3)I. INTRODUCTION前言 (4)II. DEFINING THE WHO AND WHAT OF CONTRACT MANUFACTURING指定委托生产的人和事 (6)III. RESPONSIBILITIES OF PARTIES INVOLVED IN CONTRACT MANUFACTURING委托生产所涉及各方的职责7 IV. DOCUMENTING CGMP ACTIVITIES IN QUALITY AGREEMENTS在质量协议中记录CGMP活动 (10)A.What Is a Quality Agreement? 什么是质量协议? (10)B.Elements of a Quality Agreement 质量协议的要素 (11)V. ILLUSTRATIVE SCENARIOS案例 (17)A.Owners and Contract Facilities Are Both Responsible for CGMP 所有者和受托场所是否都对CGMP负责?18B.CGMPs Apply to all Contract Facilities, Including Analytical Testing Laboratories 适用于所有合同场所,包括分析化验室的CGMP (19)C.Owners and Contract Facilities Perform Change Control Activities 所有者和受托方实施变更控制活动20VI. RECOMMENDATIONS建议 (21)Contract Manufacturing Arrangements for Drugs:QualityAgreementsGuidance for Industry1行业指南:药品委托生产安排:质量协议I.INTRODUCTION 前言This guidance describes FDA’s current thinking on defining, establishing, and documentingmanufacturing activities of the parties involved in contract drug manufacturing subject to current good manufacturing practice (CGMP) requirements. In particular, we describe how partiesinvolved in contract drug manufacturing can use quality agreements to delineate theirmanufacturing activities to ensure compliance with CGMP.本指南讲述了FDA当前对于受到CGMP约束的药品委托生产所涉及各方如何定义、设立和记录生产活动的看法,尤其是药品委托各方如何使用质量协议来描绘其生产活动,以确保符合CGMP。
美国FDA分析方法验证指引中英文对照
美国FDA分析方法验证指南中英文对照美国FDA分析方法验证指南中英文对照八、、I.INTRODUCTIONThis guida nee provides recomme ndati ons to applica nts on submitt ing an alytical procedures, validati on data, and samples to support the docume ntati on of the identity, strength, quality, purity, and potency of drug substances and drug products.1.绪论本指南旨在为申请者提供建议,以帮助其提交分析方法,方法验证资料和样品用于支持原料药和制剂的认定,剂量,质量,纯度和效力方面的文件。
This guida nce is in ten ded to assist applica nts in assembli ng in formati on, submitt ing samples, and prese nti ng data to support an alytical methodologies. The recomme ndati ons apply to drug substa nces and drug products covered in new drug applicati ons (NDAs), abbreviated new drug applicati ons (ANDAs), biologics license applications (BLAs), product license applications (PLAs), and supplements to these即plicatio ns.本指南旨在帮助申请者收集资料,递交样品并资料以支持分析方法。
FDA新药用辅料非临床研究考虑要点
FDA新药用辅料非临床研究考虑要点审评五部国家食品药品监督管理局药品审评中心【摘要】新药用辅料的研究在新药开发中占重要地位。
在我国新药的开发过程中,新辅料已有出现,在评价工作中也遇到了申报单位就相关问题的咨询,而我国目前尚无针对辅料开发临床前研究的指导原则。
在这种情况下,无论是新药研发者还是评价者都缺乏科学理论的指导,是我国研发与评价的一个空白。
FDA已经发布了新辅料非临床研究指导原则草案,是FDA相应部门评价与企业开发的重要参考,并已经积累了一些经验。
本文主要参考FDA指导原则,结合我国的实际情况,讨论了新辅料的非临床研究考虑要点,以期在探索中积累经验,为将来制定指导原则打下基础。
【关键词】药用辅料;非临床安全性1 前言本文件所指的新药用辅料为在化学的和生物来源的治疗或诊断用药物中使用的新辅料,具有以下两个特征:(1)在拟用剂量时不会发挥治疗作用(即使这些成分可能用以改善药物的转运,如增强吸收或控制有效药物释放);(2)根据目前推荐的暴露水平、暴露时间和给药途径等现有安全资料尚未被充分证实安全。
新辅料包括填充剂、稀释剂、保湿剂、溶剂、乳化剂、防腐剂、矫味剂、吸收增强剂、缓释基质以及着色剂等。
本文中新药用辅料这一术语不适用于白蛋白等大分子化合物,或生物制品中使用的氨基酸和糖等大分子化合物,也不适用于生产工艺或产品相关的杂质(如降解产物、沥出液和溶剂残渣)或外源性污染物。
2 总体考虑辅料可能是毒性物质,对药物中拟用的新辅料进行风险-效益评估,并对其建立容许性和安全性限度是十分重要的。
新辅料的应用需要安全资料的支持,可通过合理计划,以相对高效的方式评价辅料的毒理学性质。
例如,新药开发的同时,在所有动物试验中增加辅料组,同时开发新辅料。
一些辅料已有的人体资料可以替代非临床安全性资料,因此,在已有相关途径人体暴露资料时,可能不需要按以下提出的完整组合的毒理学试验进行评价。
如果现有资料不能完全支持拟用状况,将需要提供附加的安全性资料。
FDA草药法规
美国FDA与中草药相关的规定一、2004 年 6 月FDA 对“Guidance for Industry—Botanical Drug Products” 中“anicalProducts”的定义如下:Botanical products are finished, labeled products that contain vegetable matter as ingredients.A botanical product may be a food (including a dietary supplement), a drug (including abiological drug), a medical device (e.g., gutta-percha), or a cosmetic under the Act. An article is generally a food if it is used for food (21 U.S.C. 312(f)(1)). Whether an article is a drug, medical device, or cosmetic under the Act turns on its “intended use” (21 U.S.C.312(g)(1)(B) and (C), (h)(2) and (3), (i)). “Intended use” is created by claims made by or on behalf of a manufacturer or distributor of the article to prospective purchasers, such as inadvertising, labeling, or oral statements.For the purposes of this document, the term botanicals includes plant materials, algae,macroscopic fungi, and combinations thereof. It does not include:•Materials derived from genetically modified botanical species (i.e., by recombinantDNA technology or cloning).•Fermentation products (i.e., products produced by fermentation of yeast, bacteria, and other microscopic organisms, including when plants are used as a substrate, and products produced by fermentation of plant cells), even if such products are previously approved for drug use or accepted for food use in the United States (e.g., antibiotics, amino acids, and vitamins).•Highly purified substances (e.g., paclitaxel) or chemically modified substances (e.g.,estrogens synthesized from yam extracts) derived from botanical sources.Note:关于“highly purified substance”的具体界定,我已发了两次email向FDA咨询,但目前尚未得到答复。
美国FDA药品中英文翻译
美国FDA药品中英文翻译美国FDA药品中英文翻译1.处方药广告 PRESCRIPTION DRUG ADVERTISING2.处方药销售 PRESCRIPTION DRUG MARKETING3.对批发处方药销售商颁发州执照的指南 GUIDELINES FOR STATE LICENSING OF WHOLESALE PRESCRIPTION DRUG DISTRIBUTORS (奥咨达医疗器械咨询)4.人用固体口服剂型药品的印码 IMPRINTING OF SOLID ORAL DOSAGE FORM DRUG PRODUCTS FOR HUMAN USE5.药品生产者的登记与商业销售的药品的列表 REGISTRATION OF PRODUCERS OF DRUGS AND LISTING OF DRUGS IN COMMERCIAL DISTRIBUTION(只专注于医疗器械领域)6.处方药的药物治疗指导 MEDICATION GUIDES FOR PRESCRIPTION DRUG PRODUCTS7.制造、加工、包装或者保存药品的现行良好制造规范;总则 CURRENT GOOD MANUFACTURING PRACTICE IN MANUFACTURING, PROCESSING, PACKING, OR HOLDING OF DRUGS; GENERAL8.对完成的药品的现行良好制造规范 CURRENT GOOD MANUFACTURING PRACTICE FOR FINISHED PHARMACEUTICALS9.药房配药 PHARMACY COMPOUNDING10. 对含药饲料的现行良好制造规范 CURRENT GOOD MANUFACTURING PRACTICE FOR MEDICATED FEEDS11.对A型含药物品的现行良好制造规范 CURRENT GOOD MANUFACTURING PRACTICE FOR TYPE A MEDICATED ARTICLES12.对特殊人用药品的特殊要求 SPECIAL REQUIREMENTS FOR SPECIFIC HUMAN DRUGS13.管制的药品 CONTROLLED DRUGS14.药品;正式名称与已确定的名称 DRUGS; OFFICIAL NAMES AND ESTABLISHED NAMES。
- 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
- 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
- 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。
169Guidance for IndustryDrug Substance Chemistry, Manufacturing,and Controls InformationSubmit comments on this guidance at any time. Submit written comments to the Division of Dockets Management (HFA-305), Food and Drug Administration, 5630 Fishers Lane, Room 1061, Rockville, MD 20852. Comments may also be submitted electronically on the Internet at . All written comments should be identified with the Docket No. FDA-2003-D-0243 (formerly 2003D-0571).For further information regarding this document, contact Alem Ghiorghis, Center for Veterinary Medicine (HFV-143), Food and Drug Administration, 7500 Standish Place, Rockville, MD 20855, (240) 276-8266, E-mail: alem.ghiorghis@.Additional copies of this guidance document may be requested from the Communications Staff (HFV-12), Center for Veterinary Medicine, Food and Drug Administration, 7519 Standish Place, Rockville, MD 20855, and may be viewed on the Internet at either/AnimalVeterinary/default.htm or .U.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Veterinary Medicine (CVM)August 6, 2010TABLE OF CONTENTS1I.INTRODUCTION (1)II.BACKGROUND (3)A.The Common Technical Document — Quality (CTD-Q) Format (3)B.Content of an Application (4)C.Additional Guidance (4)D.References to Other Applications or Master Files (MFs) (4)1.Other Applications (4)2.Master Files (MFs) (4)III.GENERAL INFORMATION (S.1) (6)A.Nomenclature (S.1.1) (7)B.Structure (S.1.2) (7)C.General Properties (S.1.3) (7)IV.MANUFACTURE (S.2) (8)A.Manufacturers (S.2.1) (8)B.Description of Manufacturing Process and Process Controls (S.2.2) (9)1.Flow Diagram (9)2.Description of the Manufacturing Process and Process Controls (10)3.Reprocessing, Reworking, Recycling, Regeneration, and Other Operations (13)C.Control of Materials (S.2.3) (16)1. Starting Materials (16)2.Reagents, Solvents, and Auxiliary Materials (17)3.Diluents (18)D.Controls of Critical Steps and Intermediates (S.2.4) (18)E.Process Validation and/or Evaluation (S.2.5) (20)F.Manufacturing Process Development (S.2.6) (21)V.CHARACTERIZATION (S.3) (21)A.Elucidation of Structure and Other Characteristics (S.3.1) (21)1.Elucidation of Structure (21)2.Physicochemical Characterization (22)3.Biological and Other Relevant Characteristics (23)B.Impurities (S.3.2) (23)VI.CONTROL OF DRUG SUBSTANCE (S.4) (25)1 Alphanumeric designations in parentheses that follow headings show where information should be placed in applications that are submitted in Common Technical Document (CTD) format.A.Specification (S.4.1) (25)B.Analytical Procedures (S.4.2) (30)C.Validation of Analytical Procedures (S.4.3) (31)D.Batch Analyses (S.4.4) (31)1.Batch Analysis Reports (31)2.Collated Batch Analyses Data (32)E.Justification of Specification (S.4.5) (32)VII.REFERENCE STANDARDS OR MATERIALS (S.5) (35)VIII.CONTAINER CLOSURE SYSTEM (S.6) (35)IX.STABILITY (S.7) (35)A.Stability Summary and Conclusions (S.7.1) (35)B.Postapproval Stability Protocol and Stability Commitment (S.7.2) (35)C.Stability Data (S.7.3) (36)1.Primary Stability Studies (36)2.Supporting Stability Studies (36)3.Stress Studies (37)X.APPENDICES (A) (37)A.Facilities and Equipment (A.1) (37)B.Adventitious Agents Safety Evaluation (A.2) (38)1.Nonviral Adventitious Agents (39)2.Viral Adventitious Agents (39)XI.REGIONAL INFORMATION (R) (40)A.Executed Production Records (R.1.S) (40)parability Protocols (R.2.S) (40)C.Methods Validation Package (R.3.S) (40)XII.LITERATURE REFERENCES (3.3) (41)ATTACHMENT 1: (42)STARTING MATERIALS FOR SYNTHETIC DRUG SUBSTANCES (42)ATTACHMENT 2: (50)STARTING MATERIALS OF PLANT OR ANIMAL ORIGIN (50)GLOSSARY (53)Guidance for Industry2Drug SubstanceChemistry, Manufacturing, and Controls InformationThis guidance represents the Food and Drug Administration's (FDA's) current thinking on this topic. It does not create or confer any rights for or on any person and does not operate to bind FDA or the public. You can use an alternative approach if the approach satisfies the requirements of the applicable statutes and regulations. If you want to discuss an alternative approach, contact the FDA staff responsible for implementing this guidance. If you cannot identify the appropriate FDA staff, call the appropriate number listed on the title page of this guidance.I. INTRODUCTIONInformation on the chemistry, manufacturing, and controls (CMC) for the drug substance must be submitted to support the approval of original new animal drug applications (NADAs) and abbreviated new animal drug applications (ANADAs).3 This guidance provides recommendations on the CMC information for drug substances that should be submitted to support these applications. The guidance is structured to facilitate the preparation of applications submitted in Common Technical Document (CTD) format.This guidance addresses the information to be submitted for drug substances to ensure continued drug substance and drug product quality (i.e., the identity, strength, quality, purity, and potency). This guidance provides recommendations on the information that should be included for the following topics:•Nomenclature, structure, and general drug substance properties•Manufacture•Characterization•Control of drug substance•Reference standards or materials•Container closure system•StabilityThe recommendations provided in this guidance apply to the following types of drug substances: 2 This guidance has been prepared by the Office of New Animal Drug Evaluation in the Center for Veterinary Medicine at the Food and Drug Administration.3 See 21 CFR 514.1(b).•Drug substances manufactured by chemical synthesis•Highly purified and well-characterized drug substances derived from plants or animals 4•Semisynthetic drug substances manufactured by the chemical modification of a highly purified and well-characterized intermediate derived from plants or animals •The synthetic portion of the manufacturing process for semisynthetic drug substances manufactured by the chemical modification of an intermediate produced by conventional fermentation.The guidance does not provide specific recommendations relating to the following: •Monoclonal antibodies•Peptides•Oligonucleotides•Radiopharmaceuticals•Medical gases•Drug substances that are not well characterized (e.g., botanicals, some proteins) derived from plants or animals•Drug substances derived using transgenic technology•Drug substances derived directly from or manufacturing operations involving fermentation (conventional fermentation or using rDNA technology) or tissue or cellculture.More detailed guidance on the content of an application may be available in separate guidance documents (see section II.C). Applicants with drug substances not specifically covered by this (Drug Substance guidance) or another guidance can apply the content recommendations in this guidance, as scientifically appropriate, and/or can contact the appropriate review teams for guidance.FDA’s guidance documents, including this guidance, do not establish legally enforceable responsibilities. Instead, guidances describe the Agency’s current thinking on a topic and should be viewed only as recommendations, unless specific regulatory or statutory requirements are cited. The use of the word should in Agency guidances means that something is suggested or recommended, but not required.4 For purposes of this guidance, d rug substances derived from plants or animals does not include materials produced by plant cell fermentation, animal cell or tissue culture, or through use of transgenic technology (e.g., biotechnology-derived protein drug products).II. BACKGROUNDA. The Common Technical Document — Quality (CTD-Q) FormatIn November 2000, the International Conference on Harmonisation of TechnicalRequirements for Registration of Pharmaceuticals for Human Use (ICH) issuedharmonized guidance for the format of drug product applications (i.e., CommonTechnical Document (CTD)). The CTD describes a format for applications that(supplemented with regional information) can be used for submission to the regulatoryauthorities in the United States, European Union, and Japan. One focus of this effort was harmonizing the format for quality information (i.e., chemistry, manufacturing, andcontrols) that will be submitted in an application. FDA’s guidance on M4Q: The CTD — Quality describes the format for the quality information submitted in Module 3 of anapplication and provides additional information on formatting aspects of an application.The information in animal drug applications is commonly presented in the order of therequired CMC information specified under section § 514.1(b)(4) and (5). Although theCTD-Q format was developed for human drugs, the drug substance information tosupport NADAs and ANADAs can be formatted according to the CTD-Q format or any alternative format that provides the appropriate information to support the application.Applicants should review FDA’s guidance on M4Q: The CTD — Quality and otherrelated CTD guidance documents for detailed formatting recommendations on preparing an application in CTD format.This Drug Substance guidance has been organized in a format conforming to Module 3 of the CTD, and it provides CMC content recommendations specific to drug substance,including recommendations for the Appendices, Regional Information, and LiteratureReferences sections. Alphanumeric designations in parentheses corresponding to theCTD format follow relevant headings and text to show where information is to be placed in the CTD.5 Recommendations specific to drug product, including recommendations for the Appendices, Regional Information and Literature References sections, will beprovided in the Drug Product guidance.•Multiple Drug Substances in an ApplicationWhen an application is submitted for a drug product involving two or more drugsubstances (e.g., combination drug product, copackaged drug products), information for each drug substance should be presented separately in the application. Informationpresented separately means one complete S section for one drug substance followed byother complete S sections for additional drug substances. All of the information pertinent to each one of the drug substances (general information, manufacture, characterization,5 Arabic numbers have been assigned to specific sections of the CTD. For example, the designation 3.2 before S, P, A, and R indicates Module 3, Body of Data section 2. Where this guidance discusses Module 3, Body of Data section 2, for brevity, the initial designation 3.2 is not repeated throughout the rest of the guidance (e.g., 3.2.S.1.3 reads S.1.3).control, standards, container closure system, and stability) should be provided in a single section.B. Content of an ApplicationThe application should include information in every S subsection for each of the drugsubstances and manufacturing schemes (e.g., alternative processes, manufacturing site)intended for approval under the application. Information should be provided in theAppendices, Regional Information, and Literature References sections for each of thedrug substances and manufacturing schemes, as appropriate. If an Appendices orRegional Information subsection or the Literature References section is not applicable,this should be stated in the application.GuidanceC. AdditionalThis guidance references ICH guidance documents cited in the CTD-Q and FDA’sguidances on general technical topics (i.e., stability, container closure systems, analytical procedures and methods validation, sterilization process validation, drug master files, and environmental assessments) rather than incorporating this detailed information. Theseguidances are referenced in the text and/or listed at the end of a section. An applicantshould refer to these guidances for recommendations on the detailed information thatshould be included in the application to address the general technical topic.FDA continues to update existing and publish new guidance documents. An applicantshould use current guidance when preparing an NADA or ANADA submission.6D. References to Other Applications or Master Files (MFs)1. OtherApplicationsCVM discourages the reference of NDAs or ANDAs for drug substance information. In these instances, CVM recommends that the drug substance information be included in a master file or incorporated in the applicant’s NADA or ANADA. Copies of letters ofauthorization (LOAs) should be submitted in the appropriate section of an NADA orANADA.2. Master Files (MFs)This guidance describes chemistry, manufacturing, and controls information for drugsubstances that should be submitted to the Agency as part of the process of seeking theapproval of an NADA or ANADA. When a drug substance is manufactured by a firm6Current guidance documents are available on the Internet/AnimalVeterinary/GuidanceComplianceEnforcement/GuidanceforIndustry/default.htm.other than the applicant, much of this information is frequently provided by reference to one or more Type II MFs rather than directly in an application. The CMC information ina Type II MF can be organized in CTD-Q format. Under FDA's regulations, anapplication can incorporate by reference all or part of the contents of any MF to address particular drug substance issues if the MF holder provides written authorization (i.e.,LOA) to the applicant and the authorization is included in the application (in theappropriate section of an NADA or ANADA). The authorization must specificallyidentify the material being incorporated by reference (514.1(a)). The incorporatedmaterial should be identified by name, reference number, volume and page number of the MF, and date of submission. See the guidance Preparation and Submission of Veterinary Master Files for more information.Both the applicant and the drug substance manufacturer (MF holder) contribute toestablishing and maintaining the identity, strength, quality, purity, and potency of theapplicant's drug products by manufacturing and controlling the drug substance inaccordance with the information submitted in the application and, by reference, in theMF. The following recommendations pertain to location of information in the MF and/or application when an applicant and Type II MF holder are different firms.•General Information (S.17): Both the MF and the application should include this information. These sections should contain similar, though not necessarily identical,information. For example, if an applicant performed screening studies andestablished the existence of multiple polymorphs, information concerning thesepolymorphs might be present in the application but not in the MF.•Manufacture (S.2): The application should identify in S.2.1 the manufacturers of each drug substance with appropriate administrative information (see section IV.A).The MF should include this information for its manufacturing operations and anycontract facilities that are used (e.g., intermediate manufacturers, laboratories). Ingeneral, a MF can be referenced for the information recommended in S.2.2 throughS.2.6. However, the information should be augmented by the applicant, asappropriate. For example, if the applicant micronizes drug substance purchased froma MF holder the information on the micronization process should be included in theapplication.•Characterization (S.3): In general, a MF can be referenced for this information.However, the information should be augmented by the applicant, as appropriate. Forexample, characterization information on physical properties critical to the applicant’sproduct, such as solid state form or particle size distribution, should be included inS.3.1 by the applicant under certain circumstances (e.g., applicant manipulates thephysical property (micronizes), the MF holder has not characterized the physicalproperty). Furthermore, information on an applicant’s studies to characterizeimpurities (S.3.2) can be warranted to support the applicant’s drug substance controls.7 Alphanumeric designations in parentheses that follow headings show where information should be placed in applications that are submitted in Common Technical Document (CTD) format.•Control of Drug Substance (S.4): In general, information recommended in S.4 should be provided in both the MF and the application. However, reference to a MFcan be appropriate for some of the information in S.4.2 through S.4.5 if the MFholder and applicant are working together to develop the drug substance controls.Both the MF and the application should include a drug substance specification(S.4.1). The MF could include more than one drug substance specification if theholder sells different technical grades of the drug substance (e.g., micronized andnonmicronized).•Reference Standards (S.5): In general, information should be provided in both the MF and the application. However, reference to a MF can be appropriate for some ofthe information if the MF holder and applicant are working together to develop thereference standard.•Container Closure System (S.6): In general, MFs can be referenced for this information. However, the information should be augmented by the applicant, asappropriate.•Stability (S.7): In general, MFs can be referenced for this information. However, the information should be augmented by the applicant, as appropriate. For example,an applicant might perform stress studies to support the analytical procedures it usedto control the drug substance.•Appendices (A): In general, MFs can be referenced for this information. However, the information should be augmented by the applicant, as appropriate.•Regional Information (R): Comparability protocols can be included in both the MF and application (R.2.S). A methods validation package should be included in theapplication (R.3.S).•Literature References (3.3): Both the MF and the application should include literature references as warranted.Type II MFs for drug substance intermediates can also be submitted in the CTD-Qformat. However, not all sections of the CTD-Q format would apply (e.g., S.4). TheCMC information provided to support an intermediate should be appropriate for theparticular situation (e.g., process, complexity of the molecule).III. GENERAL INFORMATION (S.1)General information on the nomenclature, structure, and general properties of the drug substance, should be provided in S.1.A. Nomenclature(S.1.1)All appropriate names or designations for the drug substance should be provided in S.1.1.Any codes, abbreviations, or nicknames used in the application to identify the drugsubstance should also be listed, including the following, if they exist or have beenproposed. A name that has not yet been finalized should be identified as proposed in the list.•United States Adopted Name (USAN)•Compendial name8•Chemical names (e.g., Chemical Abstracts Service (CAS), International Union of Pure and Applied Chemistry (IUPAC))•Company names or laboratory codes•Other nonproprietary names (e.g., International Nonproprietary Name (INN), British Approved Name (BAN), Japanese Accepted Name (JAN))•Chemical Abstracts Service (CAS) Registry NumberB. Structure(S.1.2)Information on the chemical structure of the drug substance should be provided in S.1.2.This information should include:•one or more drawings to show the overall chemical structure of the drug substance, including stereochemistry•molecular formula•molecular weightFor a naturally derived protein drug substance, the information should include:•the schematic amino acid sequence indicating glycosylation sites or other posttranslational modifications• a general description of the molecule (e.g., shape, disulfide bonds, subunit composition)•number of amino acid residues•molecular weight(S.1.3)PropertiesC. GeneralA list should be provided of the general physicochemical properties of the drugsubstance. Other relevant properties of the drug substance should also be listed.Relevant properties are those physical, chemical, biological and microbiologicalattributes relating to the identity, strength, quality, purity, and/or potency of the drug8 A compendial name is a name that appears in an official compendium as defined in the Federal Food, Drug, and Cosmetic Act (e.g., United States Pharmacopeia (USP)) (§ 201(j) (21 U.S.C. 32(i)).substance and, as appropriate, drug product. The information should include, asappropriate:• A general description (e.g., appearance, color, physical state)•Melting or boiling points•Optical rotation•Solubility profile (aqueous and nonaqueous, as applicable)•Solution pH•Partition coefficients•Dissociation constants•Identification of the physical form (e.g., polymorph, solvate, or hydrate) that will be used in the manufacture of the drug product•Biological activitiesFor a naturally derived protein drug substance, additional information should be included, such as:•Isoelectric point•Extinction coefficient•Any unique spectral characteristicsIf the drug substance can exist in more than one physical form, the information included in S.1.3 should be for the form (or forms) of the drug substance that will be used in themanufacture of the drug product. Detailed information on the characterization (e.g., X-ray powder diffraction data, thermal analysis curves) of these and other physical formsand conditions required to produce one form or another should be provided in S.3.1.(S.2)IV. MANUFACTUREInformation concerning the manufacture of the drug substance, as described below, should be provided in S.2.(S.2.1)A. ManufacturersThe name, address, and manufacturing responsibility should be provided for each firm(including contract manufacturers and testing laboratories) and each site (i.e., facility)that will be involved in the manufacturing or testing of the drug substance. Each siteshould be identified by the street address, city, state, and, when available, the drugestablishment registration number.9 The addresses should be for the location where the relevant manufacturing or testing operation will be performed. Addresses for corporate headquarters or offices need not be provided. Building numbers or other specific9 See 21 CFR part 207 for registration requirements for producers of drugs. The registration number is the seven- digit central file number (CFN) or ten-digit FDA Establishment Identifier (FEI).identifying information should be provided for multifacility campuses. For sitesprocessing sterile drug substances, the sterile processing area (e.g., room) should also be included. Addresses for foreign sites should be provided in comparable detail, and thename, address, and phone number of the U.S. agent for each foreign drug establishment, as required under 21 CFR 207.40(c), should be included.To facilitate preapproval inspection related activities, it is recommended that the name,telephone number, fax number and e-mail address of a contact person be provided foreach site listed in the application. Facilities should be ready for inspection when theapplication is submitted to FDA.B. Description of Manufacturing Process and Process Controls (S.2.2)The description of the drug substance manufacturing process represents the applicant’scommitment for the manufacture of the drug substance. A flow diagram and a complete description of the processes and process controls that will be used to manufacture thedrug substance or derive it from a biological source should be provided in S.2.2. Ifalternative processes are to be used, the information should be provided for eachalternative. If justification for an alternative process is warranted, the information should be included in S.2.2 (e.g., comparative impurity data on intermediates) or can be cross-referenced if provided elsewhere in the application (e.g., S.4.4).Diagram101. FlowA flow diagram that gives the steps of the process and shows where materials enter theprocess should be provided. The entire manufacturing process should be depicted (i.e., starting materials through drug substance release testing). See Attachments 1 and 2 for information on starting materials. The flow diagram can be supplemented withinformation presented in tabular form, if appropriate. The flow diagram should include:•Each manufacturing step with identification of those steps that are critical. These manufacturing steps can include reaction, workup (e.g., extraction), isolation (e.g.,centrifugation, distillation), purification (e.g., chromatography, electrophoresis),processing (e.g., micronization), drug substance release testing.•The name or code number of the material being processed in each manufacturing step, as appropriate•Chemical structure (including stereochemical configuration where applicable) or biological identification of starting materials, intermediates, structurally complexreagents, postsynthesis materials, and the drug substance10 Headings that are not followed by alphanumeric designations (i.e., non-CTD-Q headings) are included in this document for ease of providing recommendations on the information that should be included under a CTD-Q heading (in this instance Description of Manufacturing Process and Process Controls (S.2.2)). An application submitted in CTD-Q format need not include these non-CTD-Q headings. An applicant can physically or electronically separate information under a CTD-Q heading as it chooses. However, once a particular approach is adopted, the same approach should be used throughout the life of the application.•Molecular formula and molecular weight of chemical starting materials, intermediates, postsynthesis materials, and drug substance•Solvents, reagents, and auxiliary materials used in each manufacturing step•Critical process controls and the points at which they are conducted•Operating parameters (e.g., temperature, pH, pressure) for each manufacturing step •An indication of whether intermediates are used in situ or isolated before being used in the next reaction step and which intermediates are considered the finalintermediates•Expected yield (percent) for each reaction stepReagents and other materials should not be identified using only trade (i.e., proprietary) names. If a reaction results in a mixture of products (e.g., two or more isomers), each component of the mixture should be indicated in the flow diagram. However, information on side products and impurities should be provided in S.3.2 (see sectionV.B).2. Description of the Manufacturing Process and Process ControlsA narrative description of the manufacturing process that represents the sequence of manufacturing steps undertaken and the scale of production should be provided. This description should provide more detail than that given in the flow diagram. The description should identify all process controls and the associated numeric ranges, limits, or acceptance criteria. Furthermore, any process controls that are considered critical process controls should be highlighted. See below for additional information on process controls. The detailed description of the manufacturing process and process controls should include:• A detailed description of each manufacturing step•Starting materials or intermediate used in each step, with chemical or biological names and quantities specified•Solvents, reagents, and auxiliary materials used in each step, with chemical or biological names and quantities specified•Type of equipment (e.g., Centrifuge) used, including materials of construction when critical•Identification of the manufacturing steps that are considered critical•All process controls and their associated numeric ranges, limits, or acceptance criteria, with critical process controls highlighted•Type of analytical procedure (e.g., HPLC) used for each process test •Identification of intermediates, postsynthesis materials, and unfinished drug substance that are tested (details should be provided in S.2.4)。