Guidance for Industry

合集下载

FDA 行业指南 中英对照 待完成

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 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。

guidance for industry 指导原则

guidance for industry 指导原则

"Guidance for Industry" 通常指的是由政府监管机构、行业协会或标准化组织发布的一系列建议、指导原则或最佳实践,旨在为特定行业或领域内的企业、从业人员和利益相关者提供方向、解释和规范性建议。

这些指导原则可能涉及产品质量、安全性、合规性、制造工艺、环境保护、标签和包装要求等方面。

发布这些指导原则的目的通常包括:
确保安全和合规:通过提供明确的期望和标准,帮助行业参与者遵守相关法律法规,从而确保产品、服务和运营的安全性和合规性。

促进一致性:在行业内推广统一的标准和做法,有助于减少差异,提高产品和服务的质量和可靠性。

提高效率和创新:通过提供最佳实践和推荐的技术解决方案,帮助企业改进流程、减少浪费、提升效率,并鼓励技术创新和持续改进。

保护消费者权益:确保消费者能够获得准确的信息,并在使用产品或接受服务时享有适当的保护。

促进国际贸易:在全球范围内统一或协调标准和指导原则,有助于减少贸易壁垒,促进国际贸易和合作。

环境保护和可持续发展:提供关于环境保护、资源利用和废物管理的指导,支持行业的可持续发展。

这些指导原则通常不是强制性的法律要求,但它们往往反映了监管机构的期望和行业标准,因此遵守这些指导原则通常被视为良好的业务实践。

在某些情况下,不遵守这些指导原则可能会导致监管机构的审查、处罚或声誉损害。

因此,行业内的企业和个人通常会认真考虑并遵循这些指导原则。

Guidance for Industry(双语)

Guidance for Industry(双语)

Guidance for IndustryQuality Considerations in Demonstrating Biosimilarity to a Reference Protein Product工业指南质量考虑证实与参考蛋白产品生物相似性DRAFT GUIDANCEThis guidance document is being distributed for comment purposes only.此指南发布征求意见的目的只有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.关于此草案的意见和建议应于联邦公报上宣布的指南草案的征求意见稿有效性的通知60天内提交。

For questions regarding this draft document contact (CDER) Sandra Benton at301-796-2500.U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluationand Research (CBER)February 2012BiosimilarityGuidance forIndustry Quality Considerations in DemonstratingBiosimilarity to a Reference Protein ProductAdditional copies are available from: Office of Communications Division of Drug Information, WO51, Room 2201 Center for Drug Evaluation and Research Food and Drug Administration 10903 New Hampshire Ave., Silver Spring, MD 20993 Phone: 301-796-3400; Fax: 301-847-8714druginfo@/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htmand/orOffice of Communication, Outreach, and Development, HFM-40 Center for Biologics Evaluation and Research Food and Drug Administration 1401 Rockville Pike, Rockville, MD 20852-1448 (Tel)800-835-4709 or 301-827-1800/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/default.htmU.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluationand Research (CBER)February 2012 Biosimilarity Contains Nonbinding RecommendationsDraft — Not for ImplementationTABLE OF CONTENTSI. INTRODUCTION (1)II.BACKGROUND (2)III.SCOPE (4)IV.DEFINITIONS (5)V. GENERALPRINCIPLES (6)VI. FACTORS FOR CONSIDERATION IN ASSESSING WHETHER PRODUCTS ARE HIGHLYSIMILAR (9)A. ExpressionSystem (9)B. ManufacturingProcess (10)C. Assessment of PhysicochemicalProperties (10)D. FunctionalActivities (11)E. Receptor Binding and ImmunochemicalProperties (12)F.Impurities ............................................................................................................................... (12)G. Reference Product and ReferenceStandards (13)H. Finished DrugProduct (14)I.Stability.................................................................................................................................... (15)VII.CONCLUSION (15)VIII. RELEVANT GUIDANCES (16)5 10 15 20 25 30 35 40 Contains Nonbinding RecommendationsDraft — Not for ImplementationGuidance for Industry1Quality Considerations in Demonstrating Biosimilarity to aReference Protein ProductThis draft guidance, when finalized, will represent 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 .该指南草案,一旦定稿,将代表美国食品和药物管理局(FDA)的电流思考这个话题。

FDA药物临床前指南-Guidance for Industry Unmarketed Investigational Drugs

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.。

Guidance for Industry Changes to an Approved NDA or ANDA Q&A 中英对照

Guidance for Industry Changes to an Approved NDA or ANDA Q&A 中英对照

Guidance forIndustry Changes to an Approved NDA or ANDA Questions and Answers目录REPORTING CATEGORIES报告类别 (1)GENERAL REQUIREMENTS一般要求 (2)MANUFACTURING SITES生产地址 (3)MANUFACTURING PROCESS生产工艺 (8)SPECIFICATIONS规程 (12)PACKAGE包装 (13)MISCELLANEOUS CHANGES多重变更 (15)Guidance for Industry1Changes to an Approved NDA or ANDAQuestions and AnswersThis document provides questions and answers relating to the guidance on Changes to an Approved NDA or ANDA (the guidance).2 The questions are based on those posed to CDER by applicants. The questions and answers are presented using subject headings that correspond to the table of contents in the guidance.本文提供指南文件的常见问题及解答。

常见问题来源于申请人向CDER提出的问题。

常见问题及解答按照指南文件的目录顺序列表。

REPORTING CATEGORIES报告类别Q1: For a change that is reported in a Supplement — Changes Being Effected in 30 Days, will CDER complete the review of the supplement within 30 Days?问1:对于按照30日内生效变更申请提交的报告,CDER是否能在30日内将增补文件审核完毕?A1: Within 30 days CDER will notify the applicant that prior approval isrequired for the change (i.e., CDER has designated the supplement a priorapproval supplement) or that the FDA has determined appropriate information is missing, including information that should have been developed by theapplicant in assessing the effects of the change. Supplement reviews will beperformed consistent with standard procedures. It is unlikely that a substantive review and action letter will be completed within 30 days.答1:30日内,CDER确定增补应该按照批准前增补提交,将通知申请人该项事务;或者FDA判断应有的信息存在缺陷,包括申请人应当开展的变更影响评估报告,也会通知申请人。

粉体混合均匀性指导原则的历史变迁

粉体混合均匀性指导原则的历史变迁

摘要自1998年第一份混合均匀性指导原则开始,关于药物混合均匀性取样方法以及接受标准的讨论就从未停止过,随着讨论的不断深入工业界对产品的质量控制亦走向越来越科学的方向。

FDA初代指导原则1999年8月,美国食品药品监督管理局 (FDA) 发布了一份名为“Guidance for Industry, ANDAs: Blend Uniformity Analysis”的指导原则,专门用于指导ANDA 产品的开发,其中详细的列出了混合均匀度抽样和验收标准,然NDA的亦同样用该方法进行混合均匀度的评价。

该方法是独立于USP成品放行之外的过程检测方法,并在其中提到只要产品不符合混合均匀性要求无论最终产品是否符合含量均匀度要求该批产品最终都不得放行。

产生上述矛盾的根本问题在于混合取样问题(取样器相关的混合采样错误问题)和分析方法不足(混合样品分析过程中的称重误差),然该指导原则中提到的混合均匀度限度依然被国内众多药企所奉行。

PQRI脑洞大开通常而言,最终制剂的含量均匀性很大程度上取决于混合机内粉体的均匀性,然对于存在潜在分层风险的物料而言混合器内的均匀性则很难确保最终制剂的含量均匀性。

基于99指导原则中过程检测和放行测试矛盾问题,2000年产品质量研究院(PQRI混合均匀性工作组(BUWG)) 就ANDA和NDA向FDA提交了一份提案,建议对采用最终混合取样和过程中剂量单位使用分层抽样相结合的方法来评价混合均匀性,99指导原则与2002年被FDA召回。

次年FDA发布了“Guidance for Industry :Powder Blendsand Finished Dosage Units—Stratified In-Process Dosage Unit Sampling and Assessment”由此开启了以过程分层取样来评价混合均匀性的重要跨越。

分层取样评估混合均匀性优势主要体现在如下几个方面:1.该方法可以对产品的同质性进行精确的度量。

FDA化验室OOS指南中英文_Guidance_for_Industry_OOS_CHEN

FDA化验室OOS指南中英文_Guidance_for_Industry_OOS_CHEN

Guidance for Industry Investigating Out-of-Specification (OOS)Test Results forPharmaceutical ProductionU.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)October 2006Pharmaceutical CGMPs Contains Nonbinding RecommendationsGuidance for Industry Investigating Out-of-Specification (OOS)Test Results forPharmaceutical ProductionAdditional copies are available from: 本文件可自以下途径得到Office of Training and Communication 培训和交流办公室Division of Drug Information HFD-240 药品信息分部Center for Drug Evaluation and Research 药品审评中心Food and Drug Administration 食品药品管理局5600 Fishers LaneRockville, MD 20857(Tel) 301-827-4573/cder/guidance/index.htmU.S. Department of Health and Human Services 美国卫生和福利部Food and Drug Administration 食品药品管理局Center for Drug Evaluation and Research (CDER) 药品审评中心October 2006Pharmaceutical CGMPs Contains Nonbinding RecommendationsTABLE OF CONTENTSInvestigating Out-of-Specification (OOS) Test Results for Pharmaceutical Production (4)I. INTRODUCTION 介绍 (4)II. BACKGROUND 背景 (5)III. IDENTIFYING AND ASSESSING OOS TEST RESULTS界定和评价OOS检验结果— PHASE I: LABORATORY INVESTIGATION第一步:化验室调查 (6)A. Responsibility of the Analyst 化验员职责 (7)B. Responsibilities of the Laboratory Supervisor化验室主管职责 (8)IV. INVESTIGATING OOS TEST RESULTS 对OOS结果的调查— PHASE II: FULL-SCALE OOS INVESTIGATION 第二步:全面OOS调查 (10)A. Review of Production 生产情况审核 (10)B. Additional Laboratory Testing 附加化验室测试 (11)C. Reporting Testing Results 报告测试结果 (14)V. CONCLUDING THE INVESTIGATION 调查结论 (17)A. Interpretation of Investigation Results 调查结果解释 (18)B. Cautions 注意事项 (19)C. Field Alert Reports (20)GUIDANCE FOR INDUSTRY1行业指南Investigating Out-of-Specification (OOS) Test Results forPharmaceutical Production药物生产中不合格结果的调查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. 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.本指南代表FDA对本专题现行的想法。

中英文对照Guidance for Industry Changes to an Approved NDA or ANDA

中英文对照Guidance for Industry Changes to an Approved NDA or ANDA

Guidance for Industry Changes to anApprovedNDA or ANDA已批准申请的新药变更指南U.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)April 2004CMCRevision 1I. INTRODUCTION AND BACKGROUNDThis guidance provides recommendations to holders of new drug applications (NDAs) and abbreviated new drug applications (ANDAs) who intend to make post approval changes in accordance with section 506A of the Federal Food, Drug, and Cosmetic Act (the Act) and § 314.70 (21 CFR 314.70). The guidance covers recommended reporting categories for postapproval changes for drugs other than specified biotechnology and specified synthetic biological products. It supersedes the guidance of the same title published November 1999. Recommendations are provided for postapproval changes in (1) components and composition,(2) manufacturing sites, (3) manufacturing process, (4) specifications, (5) container closure system, and (6) labeling, as well as (7) miscellaneous changes and (8) multiple related changes. 本指南给打算将已批准变更的新药上市申请和新药报审简表申请的持有者提供建议,使其按照联邦食品、药品、化妆品法案的506A部分和§ 314.70 (21 CFR 314.70)。

  1. 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. 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. 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)。

Guidance for Industry Revised Recommendations Regarding Invalidation of Test Results of Licensed and 510(k) Cleared Bloodborne Pathogen Assays Used to Test DonorsAdditional copies are available from:Office of Communication, Training and Manufacturers Assistance (HFM-40)1401 Rockville Pike, Rockville, MD 20852-1448(Tel) 1-800-835-4709 or 301-827-1800/cber/guidelines.htmU.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Biologics Evaluation and Research (CBER)July 2001TABLE OF CONTENTSI.INTRODUCTION (1)II.BACKGROUND (1)III.SPECIFIC RECOMMENDATIONS (3)A.CLIA R EQUIREMENTS FOR C ONTROL R EAGENTS (3)B.I NVALIDATION OF T EST R ESULTS B ASED ON T EST K IT P ACKAGE I NSERT (3)C.I NVALIDATION OF T EST R ESULTS B ASED ON CLIA R EGULATIONS (4)IV.REFERENCES (5)GUIDANCE FOR INDUSTRYRevised Recommendations Regarding Invalidation of Test Results of Licensed and 510(k) Cleared Bloodborne Pathogen Assays Used toTest DonorsThis guidance document represents FDA’s current thinking on invalidating test results based on the Clinical Laboratory Improvement Act of 1988 (CLIA) required control reagents. 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.INTRODUCTIONTo reduce infectious disease transmission by blood and blood products, donor samples from blood donations are tested for markers of pathogenic bloodborne infections, including antibodies, antigens, and nucleic acids that may indicate the presence of etiologic agents such as human immunodeficiency virus (HIV), hepatitis B virus (HBV), hepatitis C virus (HCV), human T-cell lymphotropic virus (HTLV), cytomegalovirus (CMV), syphilis, and others. The validity of screening and supplemental (confirmatory) test assay results is determined by the performance of test kit manufacturer supplied reagents labeled as A controls@, used in accordance with the test kit instructions. Historically, FDA has not required additional assay-specific quality control procedures beyond those documented in the test kit package inserts. FDA now recommends implementation of quality assurance and quality control procedures that involve use of external control reagents beyond those provided by the test kit manufacturer in order to contribute to overall testing accuracy and therefore to blood safety. This guidance finalizes the draft guidance entitled A Guidance for Industry-Revised Recommendations for the Invalidation of Test Results When Using Licensed and 510(k) Cleared Bloodborne Pathogen Assays to Test Donors@ that was announced in the Federal Register of September 1, 1999 (64 FR 47847). This guidance also supercedes the January 3, 1994, guidance document on this topic (Ref. 1). These invalidation procedures should be part of blood establishments= comprehensive quality assurance programs (Ref. 2) and documented in blood establishments= Standard Operating Procedures (SOPs). II.BACKGROUNDOn May 28, 1992, September 25, 1992, September 26, 1996, and December 13, 1996, the Blood Products Advisory Committee (BPAC) discussed in open public meetings circumstances under which test results may be invalidated, including actions based on the use of additional quality control reagents in donor testing. These discussions included general aspects of quality assurance related to bloodborne pathogen testing, invalidation procedures based on test kit product insert requirements, and additionalprocedures to invalidate test results based on criteria developed by the user. FDA issued guidance on this subject on January 3, 1994 (Ref. 1). In an effort to further clarify FDA=s guidance and to help ensure compliance with the Clinical Laboratory Improvement Act of 1988 (CLIA), at the December 13, 1996, BPAC meeting, the FDA, the Centers for Disease Control and Prevention (CDC), and the Health Care Financing Administration (HCFA) discussed elements of CLIA. CDC described the differences between a calibrating reagent and a reagent serving as an assay control. Based on CLIA regulations, CDC stated that if a test kit supplied A control@ reagent is used to calculate the assay cutoff, regardless of its container label title, the reagent serves the function of a calibrator and may not serve simultaneously as a control reagent. In such instances, an additional reagent should be included in the assay run to meet CLIA control reagent requirements.For example, if a test kit includes two reagents labeled as negative control and positive control respectively, and the negative control reagent is used to calculate the assay cutoff, the negative control serves as a calibrator and may not also serve as an assay control reagent. Since CLIA regulations require use of a minimum of two controls (one negative and one positive) each day of testing, a negative control from an external source should be included in the run to meet CLIA requirements. Such external controls may be developed in-house or procured, and the blood establishment must develop acceptance/rejection criteria for their use. For further information, blood establishments should refer to CLIA regulations under Title 42, Code of Federal Regulations (CFR), Part 493, or contact the State Survey Agency in the state where the laboratory is located or the accrediting organization with which the laboratory is affiliated.It is important to note that the test kit package insert instructions must continue to be followed in order for runs to be considered valid for donor testing. 21CFR 606.65(e). Accordingly, externally supplied reagents should not be substituted for test kit supplied reagents to calculate the assay cutoff, but may be used additionally to satisfy CLIA requirements.As a result of discussions held during BPAC meetings, and additional discussions among CDC, HCFA, and FDA, FDA has developed revised recommendations for blood establishments, integrating current CLIA provisions for invalidating test results based on CLIA designated control reagents.Because blood safety relies significantly on the accuracy of testing, user-developed acceptance/rejection rules should be designed only with the objective of enhancing blood safety. Accordingly, rules developed by blood establishments to meet either CLIA requirements or other in-house developed requirements should only be used to invalidate nonreactive results. Reactive results obtained from a screening test or confirmatory/supplemental test run that satisfies the test kit=s criteria for acceptance should not be invalidated. Such reactive results remain as the initial test of record, and, in the case of a screening test, these specimens should be retested in duplicate as specified in the test kit manufacturer=s instructions. The duplicate retest of a reactive specimen (duplicate rescreen) is designed to reduce the probability of obtaining a false positive result for a truly negative, but non-specifically reactive specimen. A donor sample with an invalidated nonreactive result may be retested singly.III.SPECIFIC RECOMMENDATIONSThese recommendations for procedures to invalidate test results, based on: 1) test kit package insert rejection criteria; and 2) CLIA provisions, are applicable to all blood establishments and should be included in their SOPs.A.CLIA Requirements for Control ReagentsCLIA regulations require control reagents to be used according to 42 CFR 493. If abloodborne pathogen test kit uses any of its manufacturer supplied reagents to serve as acalibrator function, i.e., either or both of the test kit controls (negative or positive) are used tocalculate the assay cutoff, then CLIA regulations require that (an) additional A control@ reagent(s) be included in each run (see table below). Such reagents may be procured or developed in-house. In any case, prior to placing the additional controls in routine use, each lot of suchreagents should have: 1) a known dating period, i.e., established stability (supplied by a control reagent manufacturer or established by the user on in-house developed control reagent); and 2) known performance parameters, i.e., specifications for acceptance. Prior to implementation,additional control reagents should be qualified, i.e., evaluated for suitability by the user applying generally accepted laboratory quality control procedures to establish acceptance/rejectioncriteria, to minimize possible incompatibilities that may exist with particular test kits beforeputting control reagents into use.TEST KIT REAGENT(S)USED IN CALCULATION OFTHE CUTOFF?ADDITIONAL CONTROLREAGENT REQUIREDNegative Control Yes Yes (Negative Control) Negative Control No NoPositive Control Yes Yes (Positive Control)Positive Control No NoPositive and NegativeControlYes Yes (Positive and Negative Controls) B.Invalidation of Test Results Based on Test Kit Package InsertAll results, both reactive and non-reactive, must be considered invalid when it has beendetermined that the assay, or part of the assay, has not been performed according to the test kit package insert instructions, (e.g., improper procedure, compromised reagent, faulty equipment) or that the test kit package insert defined run acceptance criteria have not been met, (e.g.,controls out of specified range) [21 CFR 606.65(e)]. Whenever such situations occur, testresults must be invalidated and the subsequent valid assay becomes the initial test of record.The determination of nonadherence to the test kit package insert requirements may be a resultof an investigation into an unexplained discrepancy, a routine review of the assay data, orconcerns raised by a blood establishment's quality control procedures. For example, aninvestigation of procedures may be performed in response to unexpected external controlreagent values or failure to meet the blood establishment's additional statistical acceptancecriteria for rate of reactivity. Documentation of each incident of invalidation should include thebasis for invalidation, the details of an investigation, including records of supervisory oversight, the outcome of the investigation, and, if indicated, any corrective action taken. All of theseactions should be taken prior to repeat testing of donor samples.C.Invalidation of Test Results Based on CLIA RegulationsWhen the test kit package insert instructions have been met, but CLIA control requirementshave not been met: 1)any reactive results must not be considered invalid and 2) non-reactive results should be invalidated. All specimens giving initially reactive results shouldbe re-tested in duplicate, or as the package insert directs [21 CFR 606.65(e)]. If either of the repeated duplicates is reactive, the unit should be classified as repeatedly reactive and no further repeat testing should be performed. Specimens giving repeatedly reactive test results thenshould be further assayed and reported as recommended in FDA Recommendations to Blood Establishments (Ref. 3, 4, 5, 6, 7, 8, 9) regarding viral marker testing of donations. Invalidated non-reactive specimens should be re-tested singly and those results, if valid, should become the test of record.Invalidation criteria pursuant to CLIA requirements should be based on site-user developedrules, e.g., population reactive prevalence data, or standard deviation based limits, which may be derived from generally accepted clinical laboratory quality control algorithms.Documentation for all incidents of invalidation should include the basis for invalidation, thedetails of an investigation, including records of supervisory oversight, the outcome of theinvestigation, and, if indicated, any corrective action taken. All of these actions should be taken prior to repeat testing of donor samples.Reactive results should only be invalidated when an assay run either fails to meet test kitpackage insert acceptance criteria or the assay was not performed in accordance with the test kit package insert. Accordingly, invalidation of reactive test results should not be based solely on either CLIA required quality control reagent performance or site supplemental quality control rules. Examples of circumstances when reactive results should not be invalidated include CLIA required control reagent values exceeding site established quality control acceptance limits, orreactive rates exceeding site defined upper limits.IV.REFERENCES1.FDA Recommendations for the Invalidation of Test Results When Using Licensed Viral MarkerAssays to Screen Donors, 3 January 1994, Center for Biologics Evaluation and Research,FDA.2.Guideline for Quality Assurance in Blood Establishments, 11 July 1995, Center for BiologicsEvaluation and Research, FDA.3.Revised Recommendations for Testing Whole Blood, Blood Components, Source Plasma andSource Leukocytes for Antibody to Hepatitis C Virus Encoded Antigen (Anti-HCV), 23 April 1992, Center for Biologics Evaluation and Research, FDA.4.Revised Recommendations for Testing Whole Blood, Blood Components, Source Plasma andSource Leukocytes for Antibody to Hepatitis C Virus Encoded Antigen (Anti-HCV), 5 August 1993, Center for Biologics Evaluation and Research, FDA.5.Additional Recommendations for Testing Whole Blood, Blood Components, Source Plasmaand Source Leukocytes for Antibody to Hepatitis C Virus Encoded Antigen (Anti-HCV), 16May 1996, Center for Biologics Evaluation and Research, FDA.6.Recommendations for the Management of Donors and Units that are Initially Reactive forHepatitis B Surface Antigen (HBsAg), 2 December 1987, Center for Biologics Evaluation and Research, FDA.7.Revised Recommendations for the Prevention of Human Immunodeficiency Virus (HIV)Transmission by Blood and Blood Products, 23 April 1992, Center for Biologics Evaluationand Research, FDA.8.FDA Recommendations Concerning Testing for Antibody to Hepatitis B Core Antigen (Anti-HBc), 10 September 1992, Center for Biologics Evaluation and Research, FDA.9.HTLV-I Antibody Testing, 29 September 1989, Center for Biologics Evaluation and Research,FDA.。

相关文档
最新文档