FDA关于ANDA强制降解试验的观点-英文版

合集下载

FDA关于ANDA强制降解试验的观点

FDA关于ANDA强制降解试验的观点
ICH Q1B 为光稳定性试验提出了一些建议,在 ICH 稳定性指导原则和验证指南中,没有 可以参考的关于其他降解条件的建议,对于氧化和水解降解研究也仅有有限的信息。
原料药与辅料分析方面的药物专著可以为不同原料药的各降解条件提供参考。
二、仿制药强制降解试验研究存在的问题
仿制药申请时提供的强制降解试验研究数据不完整是申报的一大缺陷。美国仿制药申报 常见缺陷解读(CMC 部分)已经出版,常见的一些例子说明,强制降解试验的缺陷包括以下 几个方面: 1. 原料药在各破坏条件下均不产生降解。请重复破坏试验以获得足够的降解产物,若没有产生降 解,请提供依据。 2. 破坏条件过于剧烈,导致大部分药物均被降解。请用温和的破坏条件或减少样品暴露时间以 产生相关的降解产物。 3. 请注意即使你已经用含量测定的方法对破坏的样品进行了检测,为了验证有关物质的检测方 法具有稳定性指示功能,破坏的样品也应用有关物质的方法进行测定。 4. 请提供所做的验证试验数据,以证明用以检测未破坏样品和破坏样品的方法能够检测出所有 的降解杂质。
对于脂溶性药物,可选择惰性溶剂溶解药物,在选择助溶剂时,应考虑药物分子中存在 的官能团不与助溶剂发生反应。
5. 请对破坏样品中已知和未知的降解产物进行列表总结。 6. 请确定检测软件对峰纯度检测的峰高要求。 7. 请说明破坏样品质量不守衡的理由。 8. 请鉴别原料药与辅料相互作用产生的降解产物。 9. 光稳定性研究表明药品对光敏感,请解释这一点是如何在分析方法,生产工艺以及药品运输
过程反映出来的。 在仿制药申请中,尽量减少பைடு நூலகம்上缺陷,通常会建议在申请过程中报告强制降解试验的相关信
对全部高中资料试卷电气设备,在安装过程中以及安装结束后进行高中资料试卷调整试验;通电检查所有设备高中资料电试力卷保相护互装作置用调与试相技互术关,系电,力根保通据护过生高管产中线工资敷艺料设高试技中卷术资配0料不置试仅技卷可术要以是求解指,决机对吊组电顶在气层进设配行备置继进不电行规保空范护载高高与中中带资资负料料荷试试下卷卷高问总中题体资,配料而置试且时卷可,调保需控障要试各在验类最;管大对路限设习度备题内进到来行位确调。保整在机使管组其路高在敷中正设资常过料工程试况中卷下,安与要全过加,度强并工看且作护尽下关可都于能可管地以路缩正高小常中故工资障作料高;试中对卷资于连料继接试电管卷保口破护处坏进理范行高围整中,核资或对料者定试对值卷某,弯些审扁异核度常与固高校定中对盒资图位料纸置试,.卷保编工护写况层复进防杂行腐设自跨备动接与处地装理线置,弯高尤曲中其半资要径料避标试免高卷错等调误,试高要方中求案资技,料术编试交写5、卷底重电保。要气护管设设装线备备置敷4高、调动设中电试作技资气高,术料课中并中3试、件资且包卷管中料拒含试路调试绝线验敷试卷动槽方设技作、案技术,管以术来架及避等系免多统不项启必方动要式方高,案中为;资解对料决整试高套卷中启突语动然文过停电程机气中。课高因件中此中资,管料电壁试力薄卷高、电中接气资口设料不备试严进卷等行保问调护题试装,工置合作调理并试利且技用进术管行,线过要敷关求设运电技行力术高保。中护线资装缆料置敷试做设卷到原技准则术确:指灵在导活分。。线对对盒于于处调差,试动当过保不程护同中装电高置压中高回资中路料资交试料叉卷试时技卷,术调应问试采题技用,术金作是属为指隔调发板试电进人机行员一隔,变开需压处要器理在组;事在同前发一掌生线握内槽图部内纸故,资障强料时电、,回设需路备要须制进同造行时厂外切家部断出电习具源题高高电中中源资资,料料线试试缆卷卷敷试切设验除完报从毕告而,与采要相用进关高行技中检术资查资料和料试检,卷测并主处且要理了保。解护现装场置设。备高中资料试卷布置情况与有关高中资料试卷电气系统接线等情况,然后根据规范与规程规定,制定设备调试高中资料试卷方案。

FDA发布行业指南草案《ANDA提交质量管理规范》

FDA发布行业指南草案《ANDA提交质量管理规范》

FDA发布行业指南草案《ANDA提交质量管理规范》2018年伊始,FDA 即一并发布了两份文件,旨在简化和改善仿制药申请(ANDA)的提交和审评。

第一份文件是行业指南草案《ANDA 提交质量管理规范》(Good ANDA Submission Practices),强调了我们在仿制药申请中看到的可能导致审批延迟的常见缺陷。

第二份文件是《ANDA 评估质量管理规范》(Good ANDA Assessment Practices),其中概述了FDA 工作人员的ANDA 评估实践。

其中,行业指南草案《ANDA 提交质量管理规范》(Good ANDA Submission Practices)中,从专利和排他性缺陷、标签缺陷、产品质量缺陷和生物等效性缺陷四个角度对ANDA提交中的常见缺陷进行了系统的整理,用于指导制药公司及其代理人的ANDA提交工作,其中大体内容如下:Ⅰ专利和排他性缺陷A. 法律诉讼的文件和通知B. 解决或提起法律诉讼C. P IV证明通知D. 橙皮书中新增或修订的信息E. 对未经批准的ANDA进行的修改F. 商业营销通知Ⅱ标签缺陷A. 容器标签和纸盒标签样稿B. 容器标签和纸盒标签的颜色差别C. 标签格式D. 注射剂1. 包装类型2. 产品规格3. 套圈和瓶盖顶封Ⅲ产品质量缺陷A. 原料药1. 活性药物成分起始原料2. API 生产工艺3. 杂质a API表征信息b 实际和潜在杂质的致突变能力的安全性评估4. 可分离中间体的质量标准5. 特定关键质量属性的检测B. 制剂1. 建立关键质量属性2. 杂质的鉴别、控制和质量a 鉴别和控制杂质b 原料药或制剂中杂质超过相关限度阈值的安全限定3. 非活性成分a 引用非活性成分数据库进行说明b 仿制药制剂中非活性成分超出IID最大值的安全性说明4. 分析方法的验证C. 体外溶出度(生物药剂学)1. 当溶出测试不能标准化时,开发和验证内部溶出度测试方法2. 溶出度接受标准D. 设施1. 生产设备的标识2. 核查的准备3. 合同生产设备和CGMP的选择E. 商业生产工艺F. 微生物学考虑1. 过程中的生物负载测试和验收标准2. 细菌内毒素测试方法的描述和验证3. 支持延长存储时间的微生物数据Ⅳ生物等效性缺陷A. 生物分析研究数据B. 临床概要C. 特定产品指导原则的偏离D. 有关BE的信息和体内BE研究的安全性E. 配方和非活性成分的差异F. 依据21 CFR 314.99(b)的豁免要求本文转自:制剂汇网,转发仅为学习交流,如有侵权,请联系小编删除。

准备FDA认证前检查中英文对照

准备FDA认证前检查中英文对照
然而,DMF持有人仍应通过年度报告和DMF修订将更新提交给FDA
This way, the DMF will be up-to-date when reviewed这样, DMF被审查时将是最新的
2021/8/6
4
The Pre-Approval Inspection (PAI) 认证前检查(PAI)
2021/8/6
2
Purpose of a pre-Approval Inspection 认证前检验的目的
Ensure that data submitted in the ANDA or DMF submission is supported by raw data at the facility
Preparation for two people generally means:
配备两个人通常是指
Two Interpreters (provided by the firm, at present) 两名翻译员(目前由公司提供的) The interpreters are extremely important to the smooth running of the inspection 翻译员对于检查顺利进行是非常重要的
When is the ANDA reviewed by FDA?
FDA什么时候审查ANDA
After the ANDA holder files the ANDA ANDA持有人将ANDA归档后
Because of backlogs, expect at least 6 month delay before reviewing 由于积压,预计至少延迟6个月才能被审查
Two adjacent conference rooms 两个相邻的会议室 Investigator 调查人员 Chemist化验师

FDA阐述ANDA申请人如何推行质量源于设计的执行的实例药物开发报告——中文版

FDA阐述ANDA申请人如何推行质量源于设计的执行的实例药物开发报告——中文版

简明新药申请质量源于设计:实例修饰释放制剂实例简介这是阐释ANDA申请人如何推行质量源于设计的执行的实例药物开发报告。

此实例的目的是阐释ANDA申请人在他们的开发过程中执行质量源于设计时可能用到的药物开发试验的类型,以及推进有关仿制药办公室如何使用该信息进行审核的讨论。

尽管我们已尽力使此实例尽可能真实,但一个真正产品的开发可能不同于此实例。

该实例出于阐释性的目的,并依赖于申请人的经验和学识,特定产品的实验程度可能不同。

在申报文件中,应全面阐释经验和知识的影响。

风险评估过程是此阐释的一个途径,在此实例中的许多地方,使用替代性的药物开发方法也是合适的。

读者注释在全文中以斜体标出,问题和意见可发送至GenericDrugs@.药物开发报告修饰释放仿制药品的实例质量源于设计目录1.1 摘要 (4)1.2 参照药品的分析 (5)1.2.1 临床 (5)1.2.2 药代动力学 (6)1.2.3 药物释放 (6)1.2.4 理化特性 (7)1.2.5 组成 (8)1.3 简明新药申请产品的质量目标产品属性 (8)1.4 溶出方法的开发和生物等效性试验 (11)1.4.1 前瞻性溶出方法的开发 (12)1.4.2体外-体内关系的建立 (14)1.4.3关键生物等效性试验 (18)2.1药品的成分 (19)2.1.1原料药 (19)2.1.1.1物理性质 (19)2.1.1.2化学性质 (20)2.1.1.3生物性质 (21)2.1.1.4原料药属性的风险评估 (21)2.1.2辅料 (23)2.1.2.1 速释颗粒中的辅料 (23)2.1.2.2缓释包衣微丸中的辅料 (25)2.1.2.3实例修饰释放片剂10mg中的辅料 (26)2.2制剂产品 (27)2.2.1处方开发 (27)2.2.1.1处方成分的初始风险评估 (28)2.2.1.2速释颗粒处方开发 (30)2.2.1.3缓释微丸处方开发 (31)2.2.1.4模型片剂的处方开发 (47)2.2.1.5 模型片剂处方和生物等效性预实验 (54)2.2.1.6处方成分的更新后风险评估 (57)2.2.2 过量投料 (59)2.2.3理化和生物性质 (59)2.3生产工艺开发 (59)2.3.1制剂生产工艺的初始风险评估 (61)2.3.2 速释颗粒工艺开发 (64)2.3.3 缓释微丸的工艺开发 (65)2.3.3.1药物包层工艺开发 (65)2.3.3.2缓释聚合物包衣工艺的开发 (77)2.3.4 总混和润滑工艺的开发 (93)2.3.5 压片工艺开发 (99)2.3.6 展示批 (108)2.3.7放大至商业化规模 (109)2.3.7.1 速释制粒的放大 (109)2.3.7.2 缓释微丸药物包层和聚合物包衣的放大 (110)2.3.7.3 总混和压缩的放大 (114)2.3.8 制剂生产工艺的更新后风险评估 (115)2.4 容器密封系统 (117)2.5 微生物属性 (118)2.6 相容性 (118)2.7 控制策略 (118)2.7.1 速释颗粒的控制策略 (118)2.7.2 缓释包衣微丸的控制策略 (120)2.7.3实例修饰释放片10mg的控制策略 (122)2.7.4 产品生命周期管理和持续改进 (123)A.1 附件I (125)缩略语表 (126)1.1 摘要以下药物开发报告总结了某例修饰释放片剂仿制药10mg的开发情况,其参照药为具有治疗性缓解作用的品牌修饰释放片剂10mg。

美国FDA指导原则CPGSEC585700根腐烂的薯片英文原版

美国FDA指导原则CPGSEC585700根腐烂的薯片英文原版

美国FDA指导原则CPGSEC585700根腐烂的薯片英文原版Guidance for Industry: Control of Salmonella in Low-Moisture Food Establishments1. Introduction2. Background3. Good Agricultural PracticesLow-moisture food establishments should work closely with their potato suppliers to ensure that proper Good Agricultural Practices (GAPs) are followed. This includes proper storage, handling, and transportation of potatoes to minimize the risk of bacterial contamination. It is also essential to maintain proper documentation and traceability to identify potential sources of contamination.4. Hygiene and Sanitation PracticesEstablishments should have robust hygiene and sanitation practices in place to prevent cross-contamination and the spread of Salmonella. This includes thorough cleaning and sanitizing of equipment, surfaces, and utensils. Employees should follow good personal hygiene practices, such as proper handwashing and wearing protective clothing, to minimize the risk of introducing bacteria into the production area.5. Environmental MonitoringRoutine environmental monitoring should be conducted in low-moisture food establishments to detect the presence of Salmonella. This can be done through swabbing of surfaces, equipment, and the production environment. Regular sampling and testing of finished products should also be conducted to ensure the absence of Salmonella.6. Employee TrainingAll employees should receive proper training on food safety practices and be aware of the potential risks associated with Salmonella contamination. Training should include information on proper hygiene practices, sanitation procedures, and the importance of following established protocols to prevent contamination.7. Hazard Analysis and Critical Control Points (HACCP)Implementing a HACCP system can help low-moisture food establishments identify potential hazards and implement preventive measures. The seven principles of HACCP should be followed, including conducting a hazard analysis, determining critical control points, establishing critical limits, monitoring procedures, corrective actions, verification, and record-keeping.8. Supplier VerificationEstablishments should have a system in place to verify the safety of their potato suppliers. This includes conducting regular audits, requesting certificates of analysis, and verifying that suppliers have adequate food safety systems in place to minimize the risk of Salmonella contamination.9. Recall PlanLow-moisture food establishments should have a recall plan in place to respond quickly and effectively in the event of a product recall. The plan should outline the steps to be taken, including notifying regulatory authorities, conducting a thorough investigation, and issuing consumer notifications to ensure the safety of the public.10. Conclusion。

201507FDA行业指南:分析方法验证(中英文)(中)

201507FDA行业指南:分析方法验证(中英文)(中)

201507FDA行业指南:分析方法验证(中英文)(中)A. Principle/Scope 原理/范围A description of the basic principles of the analytical test/technology (i.e., separation, detection); target analyte(s) and sample(s) type (e.g., drug substance, drug product, impurities or compounds in biological fluids).分析测试/技术(即分离、检测)基本原因的描述;目标分析物和样品类型(例如,原料药、制剂、杂质或生物流体中的化合物)。

B. Apparatus/Equipment 仪器/设备All required qualified equipment and components (e.g., instrument type, detector, column type, dimensions, and alternative column, filter type).所有需要的确认过的仪器和组件(例如,仪器类型、检测器、柱子类型、尺寸和可替代的柱子、过滤器类型)。

C. Operating Parameters 运行参数Qualified optimal settings and ranges (include allowed adjustments supported by compendial sources or development and/or validation studies) critical to the analysis (e.g., flow rate, components temperatures, run time, detector settings, gradient, head space sampler). A drawing with experimental configuration and integration parameters may be used, as applicable.确认过的优化的设置和范围(包括来自药典或研发和/或验证研究的允许调整),对于分析过程非常关键(例如,流速、部件温度、运行时间、检测器设置、梯度、顶空进样器)。

FDA关于ANDA强制降解试验的观点

FDA关于ANDA强制降解试验的观点

FDA关于ANDA强制降解试验的观点2016-02-15HPC药闻药事风险管理谢大侠强制降解试验为方法学验证中的重要内容,为了解国外对强制降解试验的要求,根据Pharmaceutical Technology 第36卷5期中“FDA Perspectives: Scientific Considerations of Forced Degradation Studies in ANDA Submissions”一文(发布时间为2012年5月2日,作者为Ragine Maheswaran),对FDA关于强制降解试验的相关要求进行了翻译整理,具体内容如下:一、强制降解试验简介强制降解试验也称破坏性试验,其试验目的明确。

强制降解试验可预测原料药的稳定性或影响制剂的纯度、有效性和安全性的因素。

了解不同破坏条件下药物的降解产物和降解途径是非常必要的。

强制降解试验可以为分析方法的建立、说明书的制定和处方设计的确定等提供有益的参考。

样品破坏的程度取决于药物本身的性质和产品的剂型。

ICH Q1B为光稳定性试验提出了一些建议,在ICH稳定性指导原则和验证指南中,没有可以参考的关于其他降解条件的建议,对于氧化和水解降解研究也仅有有限的信息。

原料药与辅料分析方面的药物专著可以为不同原料药的各降解条件提供参考。

二、仿制药强制降解试验研究存在的问题仿制药申请时提供的强制降解试验研究数据不完整是申报的一大缺陷。

美国仿制药申报常见缺陷解读(CMC部分)已经出版,常见的一些例子说明,强制降解试验的缺陷包括以下几个方面:1.原料药在各破坏条件下均不产生降解。

请重复破坏试验以获得足够的降解产物,若没有产生降解,请提供依据。

2.破坏条件过于剧烈,导致大部分药物均被降解。

请用温和的破坏条件或减少样品暴露时间以产生相关的降解产物。

3.请注意即使你已经用含量测定的方法对破坏的样品进行了检测,为了验证有关物质的检测方法具有稳定性指示功能,破坏的样品也应用有关物质的方法进行测定。

FDA关于破坏实验的一些最新看法和要求(中文版本)

FDA关于破坏实验的一些最新看法和要求(中文版本)

FDA关于ANDA强制降解试验的观点:强制降解试验为方法学验证中的重要内容,为了解国外对强制降解试验的要求,根据Pharmaceutical Technology第36卷5期中“FDAPerspectives: Scientific Considerations of Forced DegradationStudies in ANDA Submissions”一文(发布时间为2012年5月2日,作者为Ragine Maheswaran),对FDA关于强制降解试验的相关要求进行了翻译整理,具体内容如下:一、强制降解试验简介强制降解试验也称破坏性试验,其试验目的明确。

强制降解试验可预测原料药的稳定性或影响制剂的纯度、有效性和安全性的因素。

了解不同破坏条件下药物的降解产物和降解途径是非常必要的。

强制降解试验可以为分析方法的建立、说明书的制定和处方设计的确定等提供有益的参考。

样品破坏的程度取决于药物本身的性质和产品的剂型。

ICHQ1B为光稳定性试验提出了一些建议,在ICH稳定性指导原则和验证指南中,没有可以参考的关于其他降解条件的建议,对于氧化和水解降解研究也仅有有限的信息。

原料药与辅料分析方面的药物专著可以为不同原料药的各降解条件提供参考。

二、仿制药强制降解试验研究存在的问题仿制药申请时提供的强制降解试验研究数据不完整是申报的一大缺陷。

美国仿制药申报常见缺陷解读(CMC部分)已经出版,常见的一些例子说明,强制降解试验的缺陷包括以下几个方面:原料药在各破坏条件下均不产生降解。

请重复破坏试验以获得足够的降解产物,若没有产生降解,请提供依据。

破坏条件过于剧烈,导致大部分药物均被降解。

请用温和的破坏条件或减少样品暴露时间以产生相关的降解产物。

请注意即使你已经用含量测定的方法对破坏的样品进行了检测,为了验证有关物质的检测方法具有稳定性指示功能,破坏的样品也应用有关物质的方法进行测定。

请提供所做的验证试验数据,以证明用以检测未破坏样品和破坏样品的方法能够检测出所有的降解杂质。

美国ANDA片剂(IR)开发模板-1(中英文对照)

美国ANDA片剂(IR)开发模板-1(中英文对照)

Quality by Design for ANDAs: An Example for Immediate-Release Dosage FormsANDAs的质量源于设计:速释制剂的实例Introduction to the Example实例简介This is an example pharmaceutical development report illustrating how ANDA applicants can move toward implementation of Quality by Design (QbD). The purpose of the example is to illustrate the types of pharmaceutical development studies ANDA applicants may use as they implement QbD in their generic product development and to promote discussion on how OGD would use this information in review.这是一个有关药物开发报告的实例,用以说明ANDA申请人如何实施质量源于设计(QbD)。

该实例的目的是说明ANDA申请人在其仿制药开发过程中实施QbD时,可使用的药物开发研究的类型,同时促进探讨OGD在审评中如何使用该信息。

Although we have tried to make this example as realistic as possible, the development of a real product may differ from this example. The example is for illustrative purposes and, depending on applicants’ experience and knowledge, the degree of experimentation for a particular product may vary. The impact of experience and knowledge should be thoroughly explained in the submission. The risk assessment process is one avenue for this explanation. At many places in this example, alternative pharmaceutical development approaches would also be appropriate.虽然我们已试图让实例尽可能切合实际,但真实产品的开发可能与该实例不同。

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)。

FDA原料药GMP指南中英文对照

FDA原料药GMP指南中英文对照

Q7a(中英文对照)FDA原料药GMP指南Table of Contents 目录1. INTRODUCTION 1. 简介1.1 Objective 1.1目的1.2 Regulatory Applicability 1.2法规的适用性1.3 Scope 1.3范围2. QUALITY MANAGEMENT 2.质量管理2.1 Principles 2.1总则2.2 Responsibilities of the Quality Unit(s) 2.2质量部门的责任2.3 Responsibility for Production Activities 2.3生产作业的职责2.4 Internal Audits (Self Inspection) 2.4内部审计(自检)2.5 Product Quality Review 2.5产品质量审核3. PERSONNEL 3. 人员3.1 Personnel Qualifications 3.人员的资质3.2 Personnel Hygiene 3.2 人员卫生3.3 Consultants 3.3 顾问4. BUILDINGS AND FACILITIES 4. 建筑和设施4.1 Design and Construction 4.1 设计和结构4.2 Utilities 4.2 公用设施4.3 Water 4.3 水4.4 Containment 4.4 限制4.5 Lighting 4.5 照明4.6 Sewage and Refuse 4.6 排污和垃圾4.7 Sanitation and Maintenance 4.7 卫生和保养5. PROCESS EQUIPMENT 5. 工艺设备5.1 Design and Construction 5.1 设计和结构5.2 Equipment Maintenance and Cleaning 5.2 设备保养和清洁5.3 Calibration 5.3 校验5.4 Computerized Systems 5.4 计算机控制系统6. DOCUMENTATION AND RECORDS 6. 文件和记录6.1 Documentation System andSpecifications6.1 文件系统和质量标准6.2 Equipment cleaning and Use Record 6.2 设备的清洁和使用记录6.3 Records of Raw Materials, Intermediates, API Labeling and Packaging Materials 6.3 原料、中间体、原料药的标签和包装材料的记录6.4 Master Production Instructions (MasterProduction and Control Records)6.4 生产工艺规程(主生产和控制记录)6.5 Batch Production Records (BatchProduction and Control Records)6.5 批生产记录(批生产和控制记录)6.6 Laboratory Control Records 6.6 实验室控制记录6.7 Batch Production Record Review 6.7批生产记录审核7. MATERIALS MANAGEMENT 7. 物料管理7.1 General Controls 7.1 控制通则7.2 Receipt and Quarantine 7.2接收和待验7.3 进厂物料的取样与测试7.3 Sampling and Testing of IncomingProduction Materials7.4 Storage 7.4储存7.5 Re-evaluation 7.5复验8. 生产和过程控制8. PRODUCTION AND IN-PROCESSCONTROLS8.1 Production Operations 8.1 生产操作8.2 Time Limits 8.2 时限8.3 In-process Sampling and Controls 8.3 工序取样和控制8.4 中间体或原料药的混批8.4 Blending Batches of Intermediates orAPIs8.5 Contamination Control 8.5 污染控制9. 原料药和中间体的包装和贴签9. PACKAGING AND IDENTIFICATIONLABELING OF APIs ANDINTERMEDIATES9.1 General 9.1 总则9.2 Packaging Materials 9.2 包装材料9.3 Label Issuance and Control 9.3 标签发放与控制9.4 Packaging and Labeling Operations 9.4 包装和贴签操作10. STORAGE AND DISTRIBUTION 10.储存和分发10.1 Warehousing Procedures 10.1 入库程序10.2 Distribution Procedures 10.2 分发程序11. LABORATORY CONTROLS 11.实验室控制11.1 General Controls 11.1 控制通则11.2 Testing of Intermediates and APIs 11.2 中间体和原料药的测试11.3 Validation of Analytical Procedures 11.3 分析方法的验证11.4 Certificates of Analysis 11.4 分析报告单11.5 Stability Monitoring of APIs 11.5 原料药的稳定性监测11.6 Expiry and Retest Dating 11.6 有效期和复验期11.7 Reserve/Retention Samples 11.7 留样12. V ALIDATION 12.验证12.1 Validation Policy 12.1 验证方针12.2 Validation Documentation 12.2 验证文件12.3 Qualification 12.3 确认12.4 Approaches to Process Validation 12.4 工艺验证的方法12.5 Process Validation Program 12.5 工艺验证的程序12.6 Periodic Review of Validated Systems 12.6验证系统的定期审核12.7 Cleaning Validation 12.7 清洗验证12.8 Validation of Analytical Methods 12.8 分析方法的验证13. CHANGE CONTROL 13.变更的控制14. REJECTION AND RE-USE OF14.拒收和物料的再利用MATERIALS14.1 Rejection 14.1 拒收14.2 Reprocessing 14.2 返工14.3 Reworking 14.3 重新加工14.4 Recovery of Materials and Solvents 14.4 物料与溶剂的回收14.5 Returns 14.5 退货15. COMPLAINTS AND RECALLS 15.投诉与召回16. CONTRACT MANUFACTURERS(INCLUDING LABORATORIES)16.协议生产商(包括实验室)17. AGENTS, BROKERS, TRADERS, DISTRIBUTORS, REPACKERS, AND RELABELLERS 17.代理商、经纪人、贸易商、经销商、重新包装者和重新贴签者17.1 Applicability 17.1适用性17.2 Traceability of Distributed APIs andIntermediates17.2已分发的原料药和中间体的可追溯性17.3 Quality Management 17.3质量管理17.4 Repackaging, Relabeling, and Holding of APIs and Intermediates 17.4原料药和中间体的重新包装、重新贴签和待检17.5 Stability 17.5稳定性17.6 Transfer of Information 17.6 信息的传达17.7 Handling of Complaints and Recalls 17.7 投诉和召回的处理17.8 Handling of Returns 17.8 退货的处理18. Specific Guidance for APIs Manufactured by Cell Culture/Fermentation 18. 用细胞繁殖/发酵生产的原料药的特殊指南18.1 General 18.1 总则18.2 Cell Bank Maintenance and RecordKeeping18.2细胞库的维护和记录的保存18.3 Cell Culture/Fermentation 18.3细胞繁殖/发酵18.4 Harvesting, Isolation and Purification 18.4收取、分离和精制18.5 Viral Removal/Inactivation steps 18.5 病毒的去除/灭活步骤19.APIs for Use in Clinical Trials 19.用于临床研究的原料药19.1 General 19.1 总则19.2 Quality 19.2 质量19.3 Equipment and Facilities 19.3 设备和设施19.4 Control of Raw Materials 19.4 原料的控制19.5 Production 19.5 生产19.6 Validation 19.6 验证19.7 Changes 19.7 变更19.8 Laboratory Controls 19.8 实验室控制19.9 Documentation 19.9 文件20. Glossary 20. 术语Q7a GMP Guidance for APIs Q7a原料药的GMP指南1. INTRODUCTION 1. 简介1.1 Objective 1.1目的This document is intended to provide guidance regarding good manufacturing practice (GMP) for the manufacturing of active pharmaceutical ingredients (APIs) under an appropriate system for managing quality. It is also intended to help ensure that APIs meet the quality and purity characteristics that they purport, or are represented, to possess. 本文件旨在为在合适的质量管理体系下制造活性药用成分(以下称原料药)提供有关优良药品生产管理规范(GMP)提供指南。

201507fda行业指南:分析方法验证(中英文)(下)

201507fda行业指南:分析方法验证(中英文)(下)

201507 FDA行业指南:分析方法验证(中英文)(下)VII. STATISTICAL ANALYSIS AND MODELS 统计学分析和模型A. Statistics 统计学Statistical analysis of validation data can be used to evaluate validation characteristics against predetermined acceptance criteria. All statistical procedures and parameters used in the analysis of the data should be based on sound principles and appropriate for the intended evaluation. Several statistical methods are useful for assessing validation characteristics, for example, an analysis of variance (ANOVA) to assess regression analysis R (correlation coefficient) and R squared (coefficient of determination) or linear regression to measure linearity. Many statistical methods used for assessing validation characteristics rely on population normality, and it is important to determine whether or not to reject this assumption. There are many techniques, such as histograms, normality tests, and probability plots that can be used to evaluate the observed distribution. It may be appropriate to transform the data to better fit the normal distribution or apply distribution-free (nonparametric) approaches when the observed data are not normally distributed. Appropriateliterature or text should be consulted for information on statistical procedures to use when developing new test methods, evaluating existing test methods or evaluating measurement system performance, as well as other general information on the interpretation and treatment of analytical data[18].The data analysis should be assured either by using appropriately validated software or independent verification for correctness.验证数据的统计学分析可以用于评估验证的属性是否符合预定的可接受标准。

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

美国FDA分析方法验证指南中英文对照I. INTRODUCTIONThis guidance provides recommendations to applicants on submitting analyti cal procedures, validation data, and samples to support the documentation of th e 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 guidance is intended to assist applicants in assembling information, submitting samples, and presenting data to support analytical methodologies. Th e recommendations apply to drug substances and drug products covered in new dru g applications (NDAs), abbreviated new drug applications (ANDAs), biologics lic ense applications (BLAs), product license applications (PLAs), and supplements to these applications.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。

这些建议适用于N DA,ANDA,BLA,PLA及其它们的补充中所涉及的原料药和制剂。

2013.8-FDA指南:ANDA:原料药和制剂稳定性试验问答(中英文)

2013.8-FDA指南:ANDA:原料药和制剂稳定性试验问答(中英文)

201308 FDA指南:ANDA:原料药和制剂稳定性试验问答(中英文)Guidance for Industry 行业指南ANDAs: Stability Testing of Drug Substances and ProductsQuestions and AnswersANDA:原料药和制剂稳定性试验问答DRAFT GUIDANCE指南草案This guidance document is being distributed for comment purposes only.本指南文件发布仅供讨论。

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 electronic commentsto . Submit written 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) Radhika Rajagopalan 240-276-8546.U.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)August 2013GenericsGuidance for IndustryANDAs: Stability Testing of Drug Substances and ProductsQuestions and AnswersANDA:原料药和制剂稳定性试验问答Additional 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.htm U.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)August 2013Generics Contains Nonbinding Recommendations Draft — Not for ImplementationTABLE OF CONTENTSI. INTRODUCTION 介绍II. QUESTIONS AND ANSWERS 问与答A. General 一般问题B. Drug Master File 药物主文件.C. Drug Product Manufacturing and Packaging 药品生产和包装D. Amendments to Pending ANDA Application 未批准ANDA申请的增补E. Stability Studies 稳定性试验.Guidance for Industry[1]ANDAs: Stability Testing of Drug Substances andProductsQuestions and AnswersANDA:原料药和制剂稳定性试验问答This 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 right s 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.本指南草案,如果最终定稿,代表的是FDA目前对这一专题的态度。

解析FDA对于ANDA和NDA制剂部分的技术审核

解析FDA对于ANDA和NDA制剂部分的技术审核

解析FDA对于ANDA和NDA制剂部分的技术审核对于ANDA和NDA制剂部分的技术审核FDA于2005年设计并提出了“仿制药质量评估的理论体系”,即基于问题的审核(Question-based Review,QbR)。

QbR的提出,一方面,旨在提供标准化的审评方法,鼓励评审人员重点审核资料中有重要科学上的问题的同时,可以快速地审评低风险药物,深入审评复杂剂型,最优地利用现有的资料。

另一方面,QbR 审评有助于申请厂家了解FDA的产品质量审评逻辑,提高申请文件的质量。

本篇将介绍FDA在审阅ANDA申请或 NDA申请的制剂部分时,所关注的问题有哪些?1. 是否有关于商业化生产的产品描述?按照单位剂量和体重百分比包装和给药的成品制剂,其组份和配比是什么?每一个辅料的作用是什么?2. 各辅料的使用量是否超过了FDA IID 辅料数据库中所规定的对应给药途径的日最大摄入量?如果有,请提供合理化解释。

3. 产品处方若与RLD有不同的地方,需说明。

4. 对于505(b)(1)的申请,说明所选给药方案的合理化依据(rationale for selecting the proposed dosage form for the drugproduct);对于505b(2)和505(j)申请,RLD的特征是什么?用于拟定适应症和病人人群基础上的制剂QTPP(目标质量指标)又是什么?如何对QTPP进行合理化说明的?5. 制剂的质量属性是什么?哪些属性被界定为关键质量属性?对于每一个关键质量属性,其控制目标是什么,制订其目标的依据是什么?6. 实现产品临床表现相关的关键质量属性的方式有哪些?在药物开发过程中运用了哪些体外生物特征评价的方法(如溶出度方法)以确保产品的临床表现?7. 原料药的理化、生物特性及机械特性是什么?包括物理性质描述、pKa、手性、晶型、水溶性(pH)、吸湿性、熔点、分配系数及BCS分类(若适用)等。

FDA关于ANDA强制降解试验观点

FDA关于ANDA强制降解试验观点

FDA关于ANDA强制降解试验的观点强制降解试验为方法学验证中的重要内容,为了解国外对强制降解试验的要求,本公司根据Pharmaceutical Technology 第36卷5期中“FDA Perspectives: Scientific Considerations of Forced Degradation Studies in ANDA Submissions”一文(发布时间为2012年5月2日,作者为Ragine Maheswaran),对FDA关于强制降解试验的相关要求进行了翻译整理,具体内容如下:一、强制降解试验简介强制降解试验也称破坏性试验,其试验目的明确。

强制降解试验可预测原料药的稳定性或影响制剂的纯度、有效性和安全性的因素。

了解不同破坏条件下药物的降解产物和降解途径是非常必要的。

强制降解试验可以为分析方法的建立、说明书的制定和处方设计的确定等提供有益的参考。

样品破坏的程度取决于药物本身的性质和产品的剂型。

ICH Q1B为光稳定性试验提出了一些建议,在ICH稳定性指导原则和验证指南中,没有可以参考的关于其他降解条件的建议,对于氧化和水解降解研究也仅有有限的信息。

原料药与辅料分析方面的药物专著可以为不同原料药的各降解条件提供参考。

二、仿制药强制降解试验研究存在的问题仿制药申请时提供的强制降解试验研究数据不完整是申报的一大缺陷。

美国仿制药申报常见缺陷解读(CMC部分)已经出版,常见的一些例子说明,强制降解试验的缺陷包括以下几个方面:原料药在各破坏条件下均不产生降解。

请重复破坏试验以获得足够的降解产物,若没有产生降解,请提供依据。

破坏条件过于剧烈,导致大部分药物均被降解。

请用温和的破坏条件或减少样品暴露时间以产生相关的降解产物。

请注意即使你已经用含量测定的方法对破坏的样品进行了检测,为了验证有关物质的检测方法具有稳定性指示功能,破坏的样品也应用有关物质的方法进行测定。

请提供所做的验证试验数据,以证明用以检测未破坏样品和破坏样品的方法能够检测出所有的降解杂质。

  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)。

Scientific Considerations of Forced degradation Studies in aNda SubmissionsaBStraCta well-designed stress study can provide insight in choos-ing the appropriate formulation for a proposed product prior to intensive formulation development studies. it can prevent re-development or re-validation of a stabil-ity indicating analytical method. this paper outlines the scientific aspects of forced degradation studies that should be considered in relation to aNda submissions. iNtroduCtioNForced degradation is synonymous with stress test-ing and purposeful degradation. Purposeful degra-dation can be a useful tool to predict the stability of a drug substance or a drug product with effects on purity, potency, and safety. it is imperative to know the impurity profile and behavior of a drug substance under various stress conditions. Forced degradation also plays an important role in the development of analytical methods, setting specifications, and design of formulations under the quality-by-design (Qbd) paradigm. the nature of the stress testing depends on the individual drug substance and the type of drug product (e.g., solid oral dosage, lyophilized powders, and liquid formulations) involved (1).the international Conference on Harmonisation (iCH) Q1B guideline provides guidance for perform-ing photostability stress testing; however, there are no additional stress study recommendations in the iCH sta-bility or validation guidelines (2). there is also limited information on the details about the study of oxidation and hydrolysis. the drug substance monographs of analytical Profiles of drug Substances and excipients provide some information with respect to different stress conditions of various drug substances (3).the forced degradation information provided in the abbreviated new drug application (aNda) submissions is often incomplete and in those cases deficiencies are cited. an overview of common deficiencies cited through-out the chemistry, manufacturing, and controls (CMC) section of the aNdas has been published (4-6). Some examples of commonly cited deficiencies related to forced degradation studies include the following:• y our d rug s ubstance d oes n ot s how a ny d egrada-tion under any of the stress conditions. Pleaserepeat stress studies to obtain adequate degra-dation. if degradation is not achievable, pleaseprovide your rationale.• P lease note that the conditions employed forstress study are too harsh and that most of yourdrug s ubstance h as d egraded. P lease r epeat y ourstress s tudies u sing m ilder c onditions o r s horterexposure t ime t o g enerate r elevant d egradationproducts.• i t is noted that you have analyzed your stressedsamples as per the assay method conditions.For the related substances method to be sta-bility indicating, the stressed samples shouldbe analyzed using related substances methodconditions.• P lease state the attempts you have made toensure that all the impurities including thedegradation p roducts o f t he u nstressed a nd t hestressed samples are captured by your analyti-cal method.ragine MaheswaranaBout tHe autHorragine Maheswaran, Ph.d., is a CMC reviewer at the office of generic drugs within the office of Pharmaceutical Science, under the uS Food and drug administration’s Center for drug evaluation and research and may be reached by e-mail at ragine.Maheswaran@.[For more author information, go to /bios• P lease provide a list summarizing the amountof d egradation p roducts (known a nd u nknown)in your stressed samples.• P lease verify the peak height requirement ofyour s oftware f or t he p eak p urity d etermination.• P lease e xplain t he m ass i mbalance o f t he s tressedsamples.• P lease identify the degradation products thatare formed due to drug-excipient interactions.• y our photostability study shows that the drugproduct is very sensitive to light. Please explainhow this is reflected in the analytical method,manufacturing process, product handling, etc.in an attempt to minimize deficiencies in the aNda submissions, some general recommendations to conduct forced degradation studies, to report relevant information in the submission, and to utilize the knowledge of forced degradation in developing stability indicating analytical methods, manufacturing process, product handling, and storage are provided in this article.StreSS CoNditioNStypical stress tests include four main degradation mecha-nisms: heat, hydrolytic, oxidative, and photolytic degrada-tion. Selecting suitable reagents such as the concentration of acid, base, or oxidizing agent and varying the conditions (e.g., temperature) and length of exposure can achieve the preferred level of degradation. over-stressing a sample may lead to the formation of secondary degradants that would not be seen in formal shelf-life stability studies and under-stressing may not serve the purpose of stress test-ing. therefore, it is necessary to control the degradation to a desired level. a generic approach for stress testing has been proposed to achieve purposeful degradation that is predictive of long-term and accelerated storage condi-tions (7). the generally recommended degradation varies between 5-20% degradation (7-10). this range covers the generally permissible 10% degradation for small molecule pharmaceutical drug products, for which the stability limit is 90%-110% of the label claim. although there are refer-ences in the literature that mention a wider recommended range (e.g., 10-30%), the more extreme stress conditions often provide data that are confounded with secondary degradation products.PhotostabilityPhotostability testing should be an integral part of stress testing, especially for photo-labile compounds. Some recommended conditions for photostability testing are described in iCH Q1B Photostability testing of New drug Substances and Products (2). Samples of drug substance, and solid/liquid drug product, should be exposed to a minimum of 1.2 million lux hours and 200 watt hours per square meter light. the same samples should be exposed to both white and uv light. to minimize the effect of temperature changes during exposure, tempera-ture control may be necessary. the light-exposed samples should be analyzed for any changes in physical proper-ties such as appearance, clarity, color of solution, and for assay and degradants. the decision tree outlined in the iCH Q1B can be used to determine the photo stability testing conditions for drug products. the product label-ing should reflect the appropriate storage conditions. it is also important to note that the labeling for generic drug products should be concordant with that of the reference listed drug (rld) and with united States Pharmacopeia (uSP) monograph recommendations, as applicable. Heatthermal stress testing (e.g., dry heat and wet heat) should be more strenuous than recommended iCH Q1a accel-erated testing conditions. Samples of solid-state drug substances and drug products should be exposed to dry and wet heat, whereas liquid drug products can be exposed to dry heat. it is recommended that the effect of temperature be studied in 10ºC increments above that for routine accelerated testing, and humidity at 75% rela-tive humidity or greater (1). Studies may be conducted at higher temperatures for a shorter period (10). testing at multiple time points could provide information on the rate of degradation and primary and secondary degrada-tion products. in the event that the stress conditions pro-duce little or no degradation due to the stability of a drug molecule, one should ensure that the stress applied is in excess of the energy applied by accelerated conditions (40º for 6 months) before terminating the stress study. acid and Base Hydrolysisacid and base hydrolytic stress testing can be carried out for drug substances and drug products in solution at ambient temperature or at elevated t emperatures. the selection of the type and concentrations of an acid or a base depends on the stability of the drug substance.a strategy for generating relevant stressed samples for hydrolysis is stated as subjecting the drug substance solution to various pHs (e.g., 2, 7, 10-12) at room tem-perature for two weeks or up to a maximum of 15% degradation (7). Hydrochloric acid or sulfuric acid (0.1 M to 1 M) for acid hydrolysis and sodium hydroxide or potassium hydroxide (0.1 M to 1 M) for base hydrolysis are suggested as suitable reagents for hydrolysis (10). For lipophilic drugs, inert co-solvents may be used tosolubilize the drug substance. attention should be given to the functional groups present in the drug molecule when selecting a co-solvent. Prior knowledge of a com-pound can be useful in selecting the stress conditions. For instance, if a compound contains ester functionality and is very labile to base hydrolysis, low concentrations of a base can be used. analysis of samples at various intervals can provide information on the progress of degradation and help to distinguish primary degradants from secondary degradants.oxidationoxidative degradation can be complex. although hydro-gen peroxide is used predominantly because it mimics possible presence of peroxides in excipients, other oxi-dizing agents such as metal ions, oxygen, and radical initiators (e.g., azobisisobutyronitrile, aiBN) can also be used. Selection of an oxidizing agent, its concentration, and conditions depends on the drug substance. Solutions of drug substances and solid/liquid drug products can be subjected to oxidative degradation. it is reported that subjecting the solutions to 0.1%-3% hydrogen peroxide at neutral pH and room temperature for seven days or up to a maximum 20% degradation could potentially generate relevant degradation products (10). Samples can be analyzed at different time intervals to determine the desired level of degradation.different stress conditions may generate the same or different degradants. the type and extent of degradation depend on the functional groups of the drug molecule and the stress conditions.aNalySiS MetHodthe preferred method of analysis for a stability indicating assay is reverse-phase high-performance liquid chroma-tography (HPlC). reverse-phase HPlC is preferred for several reasons, such as its compatibility with aqueous and organic solutions, high precision, sensitivity, and ability to detect polar compounds. Separation of peaks can be carried out by selecting appropriate column type, column temperature, and making adjustment to mobile phase pH. Poorly-retained, highly polar impurities should be resolved from the solvent front. as part of method development, a gradient elution method with varying mobile phase composition (very low organic composi-tion to high organic composition) may be carried out to capture early eluting highly polar compounds and highly retained nonpolar compounds. Stressed samples can also be screened with the gradient method to assess poten-tial elution pattern. Sample solvent and mobile phase should be selected to afford compatibility with the drug substance, potential impurities, and degradants. Stress sample preparation should mimic the sample preparation outlined in the analytical procedure as closely as possible. Neutralization or dilution of samples may be necessary for acid and base hydrolyzed samples. Chromatographic profiles of stressed samples should be compared to those of relevant blanks (containing no active) and unstressed samples to determine the origin of peaks. the blank peaks should be excluded from calculations. the amount of impurities (known and unknown) obtained under each stress condition should be provided along with the chromatograms (full scale and expanded scale show-ing all the peaks) of blanks, unstressed, and stressed samples. additionally, chiral drugs should be analyzed with chiral methods to establish stereochemical purity and stability (11, 12).the analytical method of choice should be sensitive enough to detect impurities at low levels (i.e., 0.05% of the analyte of interest or lower), and the peak responses should fall within the range of detector’s linearity. the analytical method should be capable of capturing all the impurities formed during a formal stability study at or below iCH threshold limits (13, 14). degradation product identifica-tion and characterization are to be performed based on for-mal stability results in accordance with iCH requirements. Conventional methods (e.g., column chromatography) or hyphenated techniques (e.g., lC-MS, lC-NMr) can be used in the identification and characterization of the degradation products. use of these techniques can provide better insight into the structure of the impurities that could add to the knowledge space of potential structural alerts for genotoxicity and the control of such impurities with tighter limits (12-17). it should be noted that structural characterization of degradation products is necessary for those impurities that are formed during formal shelf-life stability studies and are above the qualification threshold limit (13).various detection types can be used to analyze stressed samples such as uv and mass spectroscopy. the detec-tor should contain 3d data capabilities such as diode array detectors or mass spectrometers to be able to detect spectral non-homogeneity. diode array detection also offers the possibility of checking peak profile for multiple wavelengths. the limitation of diode array arises when the uv profiles are similar for analyte peak and impurity or degradant peak and the noise level of the system is high to mask the co-eluting impurities or degradants. Compounds of similar molecular weights and functional groups such as diastereoisomers may exhibit similar uv profiles. in such cases, attempts must be made to modify the chromatographic parameters to achieve necessaryseparation. an optimal wavelength should be selected to detect and quantitate all the potential impurities and degradants. use of more than one wavelength may be necessary, if there is no overlap in the uv profile of an analyte and impurity or degradant peaks. a valuable tool in method development is the overlay of separation signals at different wavelengths to discover dissimilarities in peak profiles.Peak Purity analysisPeak purity is used as an aid in stability indicating meth-od development. the spectral uniqueness of a compound is used to establish peak purity when co-eluting com-pounds are present.Peak purity or peak homogeneity of the peaks of interest of unstressed and stressed samples should be established using spectral information from a diode array detector. when instrument software is used for the determination of spectral purity of a peak, relevant parameters should be set up in accordance with the man-ufacturer’s guidance. attention should be given to the peak height requirement for establishing spectral purity. uv detection becomes non linear at higher absorbance values. thresholds should be set such that co-eluting peaks can be detected. optimum location of reference spectra should also be selected. the ability of the soft-ware to automatically correct spectra for continuously changing solvent background in gradient separations should be ascertained.establishing peak purity is not an absolute proof that the peak is pure and that there is no co-elution with the peak of interest. limitations to peak purity arise when co-eluting peaks are spectrally similar, or below the detec-tion limit, or a peak has no chromophore, or when they are not resolved at all.Mass BalanceMass balance establishes adequacy of a stability indicat-ing method though it is not achievable in all circum-stances. it is performed by adding the assay value and the amounts of impurities and degradants to evaluate the closeness to 100% of the initial value (unstressed assay value) with due consideration of the margin of analytical error (1).Some attempt should be made to establish a mass balance for all stressed samples. Mass imbalance should be explored and an explanation should be provided. varying responses of analyte and impurity peaks due to differences in uv absorption should also be examined by the use of external standards. Potential loss of volatile impurities, formation of non-uv absorbing compounds, formation of early eluants, and potential retention of compounds in the column should be explored. alternate detection techniques such as ri lC/MS may be employed to account for non-uv absorbing degradants. terMiNatioN oF StudyStress testing could be terminated after ensuring adequate exposure to stress conditions. typical a ctivation energy of drug substance molecules varies from 12-24 kcal/mol (18). a compound may not necessarily degrade under every single stress condition, and general guideline on exposure limit is cited in a review article (10). in cir-cumstances where some stable drugs do not show any degradation under any of the stress conditions, specificity of an analytical method can be established by spiking the drug substance or placebo with known impurities and establishing adequate separation.otHer CoNSideratioNSStress testing may not be necessary for drug substances and drug products that have pharmacopeial methods and are used within the limitations outlined in uSP <621>. in the case where a generic drug product uses a different polymorphic form from the rld, the drug substance should be subjected to stress testing to evaluate the physiochemical changes of the polymorphic form because different polymorphic forms may exhibit dif-ferent stability characteristics.ForCed degradatioNiN QBd ParadigMa systematic process of manufacturing quality drug prod-ucts that meet the predefined targets for the critical quality attributes (CQa) necessitates the use of knowledge obtained in forced degradation studies.a well-designed, forced degradation study is indis-pensable for analytical method development in a Qbd paradigm. it helps to establish the specificity of a stability indicating method and to predict potential degradation products that could form during formal stability studies. incorporating all potential impurities in the analytical method and establishing the peak purity of the peaks of interest helps to avoid unnecessary method re-development and revalidation.Knowledge of chemical behavior of drug substances under various stress conditions can also provide useful information regarding the selection of excipients for formu-lation development. excipient compatibility is an integral part of understanding potential formulation interactions during product development and is a key part of product understanding. degradation products due to drug-excipi-ent interaction or drug-drug interaction in combina-tion products can be examined by stressing samples of drug substance, drug product, and placebo separately and comparing the impurity profiles. information obtained regarding drug-related peaks and non-drug-related peaks can be used in the selection and devel-opment of more stable formulations. For instance, if a drug substance is labile to oxidation, addition of an antioxidant may be considered for the formulation. For drug substances that are labile to acid or undergo stereochemical conversion in acidic medium, delayed-release formulations may be necessary. acid/base hydrolysis testing can also provide useful insight in the formulation of drug products that are liquids or suspensions.Knowledge gained in forced degradation studies can facilitate improvements in the manufacturing process. if a photostability study shows a drug substance to be photolabile, caution should be taken during the manufacturing process of the drug product. useful information regarding process development (e.g., wet versus dry processing, temperature selection) can be obtained from thermal stress testing of drug substance and drug product.additionally, increased scientific understanding of degradation products and mechanisms may help to determine the factors that could contribute to stability failures such as ambient temperature, humidity, and light. appropriate selection of packaging materials can be made to protect against such factors. CoNCluSioNan appropriately-designed stress study meshes well with the Qbd approaches currently being promoted in the pharmaceutical industry. a well-designed stress study can provide insight in choosing the appropriate formulation for a proposed product prior to inten-sive formulation development studies. a thorough knowledge of degradation, including mechanistic understanding of potential degradation pathways, is the basis of a Qbd approach for analytical method development and is crucial in setting acceptance criteria for shelf-life monitoring. Stress testing can provide useful insight into the selection of physical form, stereochemical stability of a drug substance, packaging, and storage conditions. it is important to perform stress testing for generic drugs due to allowable qualitative and quantitative differences in formulation with respect to the rld, selection of manufacturing process, processing parameters, and packaging materials.reFereNCeS1. iCH, Q1a(r2) Stability testing of New drug Substances andProducts, geneva, February 2003.2. iCH, Q1B Stability testing: Photostability testing of New drugSubstances and Products, geneva, November 1996.3. H. Brittain, analytical Profiles of drug Substances and excipients,academic Press, london.4. a. Srinivasan and r. iser, Pharm. technol. 34(1), 50-59, 2010.5. a. Srinivasan, r. iser, and d. gill, Pharm. technol. 34(8), 45-51, 2010.6. a. Srinivasan, r. iser, and d. gill, Pharm. technol. 35(2), 58-67, 2011.7. S. Klick, et al., Pharm.technol. 29(2) 48-66, 2005.8. K. M. alsante, l. Martin and S. w. Baertschi, Pharm.technol.27(2) 60-72, 2003.9. d. w. reynolds, K. l. Facchine, J. F. Mullaney, K. M. alsante,t. d. Hatajik, and M. g. Motto, Pharm.technol. 26(2), 48-56, 2002.10. K. M. alsante, a. ando, r. Brown, J. ensing, t. d. Hatajik, w.Kong, and y. tsuda, advanced drug delivery reviews 59, 29-37 (2007).11. Fda, guidance for industry on analytical Procedures and methodsvalidation Chemistry, Manufacturing, and C ontrols documenta-tion (draft), rockville, Md, august 2000.12. iCH, Q6a: Specifications: test Procedures and acceptance Crite-ria for New drug Substances and New drug Products: Chemical Substances, geneva, october 1999.13. iCH, Q3a(r2) impurities in New drug Substances, geneva,october 2006.14. iCH, Q3B(r2) impurities in New drug Products, geneva, June2006.15. Fda, guidance for industry aNdas: impurities in drug Sub-stances (draft), rockville, Md, august 2005.16. Fda, guidance for industry aNdas: impurities in drug Products(draft), rockville, Md, November 2010.17. eMea, guideline on the limits of genotoxic impurities, Com-mittee for Medical Products for Human use (CHMP) (doc. ref eMea/CHMP/QwP/251344/2006), Jan. 1, 2007.18. K. a. Conners et al., Chemical Stability of Pharmaceuticals,wiley and Sons, New york, New york, 2nd ed., 1986) p.19.JvtaCKNowledgMeNtS aNd diSClaiMerthe author would like to thank Bob iser, Naiqi ya, dave Skanchy, Bing wu, and ashley Jung for their scientific input and support.disclaimer: the views and opinions in this articleare only those of the author and do not necessarily reflect the views or policies of the uS Food and drug administration.。

相关文档
最新文档