Drug RA丨第12章 活性物质注册的全球监管程序

职场   2024-11-02 17:43   江苏  


此次医博会由同写意策划,以“发展新质生产力,共享健康新未来”为主题,用全新的视角瞄准国内外医药及大健康产业发展前沿。诚邀海内外医药及大健康各界嘉宾齐聚中国医药城,共赴时代之约。展位/报告火热征集中!



引进新药有两个关键目标。其一是在药品生命周期内成功申请,获得上市许可。申请过程包括上市许可申请(MAA)、新药申请(NDA)或生物许可申请(BLA),以及活性物质注册,其中详细说明了产品的生产和控制方式,并总结了多年的测试结果。其次是通过在药品销售所在国开发并获得知识产权来创造独特价值。


活性药物成分(active pharmaceutical ingredient,API)是开发医药产品的核心,是能够带来预期健康益处的药物物质。无论是化学药物还是生物制品,原料药开发商和生产商都要投入大量时间和高昂成本来确定原料药的特性和生产工艺。活性物质的注册提供了有关药物的全面信息,使监管机构和医疗从业人员能够确保治疗疗效大于风险(therapeutic benefits outweigh the risks)

监管机构会对质量、安全性和疗效进行评估,并对未来上市许可持有人确保和监控可持续效益风险比的能力进行评估(evaluate the ability of the future marketing license holder to ensure and monitor a sustainable benefit-risk ratio)。除了总结质量、非临床和临床数据外,上市申请文件还揭示了安全报告系统如何从开发阶段过渡到上市后批准阶段。

公司在寻求制定全球监管战略时,需要确定在哪些国家或经济区注册拟议的活性物质。2020年,有10个国家被确定为顶级医药市场,如图11-1所示。

如今,全球市场已被划分为不同的经济区块。因此,顶级制药国可划分为以下区块(图11-2)


北美:美国
欧盟:法国、德国、意大利和西班牙;
金砖四国(BRIC):巴西和中国;
跨太平洋伙伴关系:日本;以及
英联邦:加拿大和英国。


用于生产原料药的原材料和原料药生产过程本身都应受到严格控制,并受到包括现行良好生产规范(current good manufacturing practices,cGMP) 在内的全球法规的约束,以便生产出可配制成医药产品并安全使用的优质原料药。为支持临床研究或上市而在注册资料中描述的CMC必须全面描述原料药,以及第11、12、13和14章(后续推文将推出)中描述的其他基本信息。

各国对原料药生产商提交药品主文件(Drug master file,DMF)的监管要求和获得批准的审查程序可能有所不同。表11-1列出了美国、欧盟、日本和中国等国家的要求。本章信息反映的是美国、加拿大、英国、巴西、中国和日本的情况。





全球监管要求


美国


关于提交DMF的规定已编入《联邦法规汇编》第21篇第314.420部分。2019年 10月,美国食品和药品管理局(FDA)发布了《Drug Master Files Guidance for Industry》 修订版1.0,对该机构1989年9月的指导文件进行了更新。该指南说明了FDA目前对DMF的想法,但并未制定可依法强制执行的要求(This guidance describes the FDA’s current thinking on DMFs and does not establish legally enforceable requirements.)

该指南将DMF定义为"向FDA提交的文件,可用于提供有关用于生产、加工、包装和储存人用药品的设施、工艺或物品的机密详细信息"(submissions to FDA that may be used to provide confidential, detailed information about facilities, processes or articles used in the manufacture,processing, packaging, and storing of human drug products.)。DMF通常不用于提交非专有信息。

FDA指南中定义的DMF有四种类型:

  1. II型包括原料药、原料药中间体及其生产中所用材料的信息;
  2. III型用于包装材料;
  3. IV型包括辅料、着色剂、香料、香精或用于制备的材料;以及
  4. V型用于FDA认可的参考信息。

DMF必须是准确和完整的英文译本;不要求认证译本。一般来说,FDA希望DMF 持有者是DMF所涵盖材料的生产商。如果DMF持有人不是生产商,DMF应包括 DMF持有人对材料生产承担全部责任的声明。任何材料的生产商都可以决定将审查所需的信息直接提交给客户,以便提交研究性新药(IND)、NDA、仿制药申请 (ANDA)、另一份DMF以及监管申请的修订(amendments)和补充(supplement)。除了模块1、2.1、模块4.1和模块6外,FDA允许交叉引用 DMF或模块32S的大部分内容。此外,无论通过DMF提交还是直接纳入市场申请,数据要求都是一样的。

FDA的指南提供了准备和提交DMF的详细信息、DMF中应包含的信息以及该机构的审查程序。其他信息可在FDA的DMF网页上查阅。该机构每季度更新DMF列表。截至2022年6月,共有35,000多份DMF在列。该清单包括少量(490个)I 型DMF;不过,I型DMF已于2000年停止使用,但其他DMF类型的编号没有变化。在FDA清单上的DMF中,数量最多的是II型DMF(约24,800个)。其次是III型DMF,约有6,500个。II型DMF是使用最广泛的DMF类型。在考虑为您的原料药采用III型DMF,或作为监管申请的发起人/申请人引用III型DMF时,指南中的以下内容非常重要。

II型DMF必须按照以电子格式提供监管提交的行业指南-某些人用药品申请和使用eCTD规范的相关提交(Rev.6),以标准电子通用技术文件(eCTD)格式提交。FDA的eCTD提交标准以及eCTD部分标题的完整列表可在该机构的网站上提供此外,还提供了CTD的其他信息和ICH指南。10GB或更小的DMF必须通过FDA的电子提交网关(ESG)提交。大多数DMF都属于这一类。超过10G的作品也可以通过ESG提交,也可以通过CD或u盘等物理介质递交。FDA于2019年6月宣布了一项与生物制品许可申请(BLA)和主文件相关的拟议规则(84 FR 30968)。拟议规则最终确定后,将编纂三个具体方面:

允许最初根据联邦食品、药品和化妆品(FD&C)法案在新药申请(NDA)中批准的某些生物制品,在NDA被视为公共卫生服务(PHS)法案下生物制品的许可后,继续依赖DMF获取原料药(DS,drug substance)、原料药中间体(drug substance intermediate,DSI)或药品(drug product,DP)的信息。

根据PHS法案的BLA可以依赖主文件,但DS信息除外。DSI定义为原料药生产过程中生产的材料,在成为原料药或DP(成品剂型,如片剂、胶囊)后,经过进一步的分子变化或纯化。PHS法案还编纂了FDA允许研究性新药申请的实践,通过参考包含在许可产品的主文件中的任何信息。该局通常希望在申请的相关部分向BLA提交原料药信息。

由于DMF不会被批准或否决,因此DMF中不需要提交环境评估(eCTD第 1.12.14节)。然而,DMF持有者应在DMF中承诺按照适用的环境法律运行其设施。该声明也可作为附录A.2.1的一部分,纳入申请人提交的监管材料中。

在以eCTD格式提交II型DMF原件之前,DMF持有者(DMF holder)必须向FDA 的药物评价与研究中心(CDER)申请一个预先分配的编号。对于II型DMF,模块3是最相关的部分,模块3的信息汇总在模块2"质量总体摘要 "中。模块4"非临床 "通常不是II型DMF所必需的,除非进行了支持安全性的非临床评估,如杂质评估。

在封面信(cover letter)中,必须包括DMF持有人签署的承诺声明,即DMF是最新的,持有人将遵守DMF中的声明。应提供联系人和负责官员(如果与联系人不同)的详细信息,包括姓名、职称、电话号码、传真号码(如果有)、安全电子邮件地址以及具体职责说明。FDA建议使用CDER网页上的cover letter模板,用于原始和后续提交。

为方便与FDA沟通,境外DMF持有者必须指定一名熟悉机构法规、指南和程序的代理,最好是在美国。代理人不负责DMF的内容;这一责任不能也不应委托给代理人。代理人可代表DMF持有人提交DMF,并可在DMF提交材料上签字,但以下情况除外:


代理人委任书(Agent appointment letters)

• 承诺声明(Statement of commitment)

• 名称变更(Name changes)

• 持有人转让(Holder transfers)

• 新持有人接受函(New holder acceptance letters)

• DMF关闭请求(DMF closure requests)

该指南进一步澄清,DMF的代理人与药品上市和注册系统(DRLS)的代理人不同;除非此人同时也是DMF和DRLS的代理人,否则不应将其列入DMF的注册名单。DMF持有人可以为不同的DMF指定不同的代理人。

早些年,DMF是在申办者/申请人申请中引用之前提交给FDA的。最近,DMF的提交更接近或与申请同时提交,以支持临床评估或产品上市审批。提交DMF后,FDA将审查文件中的管理信息,并向DMF持有人(和代理人,如适用)发送确认函(acknowledgement letter),列出DMF编号、主题(标题)、类型(I、II、III或IV)和持有人姓名,如DMF封面函中所述。如果管理信息不完整,FDA会联系持有人(和代理人,如果适用),要求提供缺失的信息。

在所有行政信息备案问题得到充分解决,并且DMF在申请或另一个DMF中被引用之前,DMF不能用于技术审查。行政信息中的一份重要文件是取消资格证明。DMF持有人被列入《食品与药物管制法》第306(k)(1)条要求的 "以任何身份在申请中使用其服务的人员"类别。DMF持有人可在eCTD第1.3.3中提交自己的取消资格证明。关于取消资格证明的更多信息,请参阅FDA的《提交取消资格证明声明的行业指南》草案。

成功使用DMF的最关键文件是授权书(Letter of Authorization,LoA)。它是完整审查DMF中参考技术信息的关键。在将LoA提交给DMF用于特定申请或其他DMF(21 CFR Part 314.420[d])之前,FDA不会审查DMF中的技术信息。LoA允许FDA审查DMF,并允许授权方(即提出申请的公司或个人,或其他DMF)通过引用将信息纳入申请或其他DMF(eCTD第1.4.1节)。审查员可能会发现需要更多信息才能继续审查,或者DMF不能用于支持ANDA或NDA的批准,或者在IND的情况下,不能用于允许临床试验的进行。

在这些情况下,FDA将就有关问题联系DMF持有人和代理人(如适用)。如果DMF持有人不同意请求或意见,这种情况可能会给申请人带来挑战。与DMF持有者签订的质量协议必须包括有关DMF审查中监管机构提出的意见和要求的公开和具体的沟通时限,以便申请人能够处理、减轻和在内部沟通对批准或临床试验启动的任何影响或延误。此外,申请人还必须了解DMF持有人做出的任何可能影响医药产品的承诺。

DMF持有人可授权一个或多个申请人以引用方式纳入DMF中的信息。即使授权方和DMF持有者是同一家公司,也要提交LoA。不过,申请人在引用自己的材料时无需创建新的DMF,可以直接将信息纳入自己的申请中。


DMF持有者向被授权方发送LoA的副本,被授权方必须在申请(21 CFR Part 314.50[a][1])或DMF(eCTD第1.4.2节)中包含LoA的副本。在eCTD第1.4.3节中,必须列出目前被授权以引用方式纳入DMF中任何信息的各方的名单,并在其他各方被授权或撤销时进行修订和提交。

  • 上述信息适用于所有四类DMF。对于II型DMF,特别是DS、DSI及其制备和储存中使用的材料,应在关于标签的eCTD第1.149节中提供一份运往现场或现场进行远期加工的运输标签副本。

  • 每份II型DMF应针对单一药物物质中间体及其制备过程中使用的材料类型提交,并包括一份声明,说明DMF所涵盖的材料是按照cGMP生产的。使用不同工艺生产的药物物质也应提交单独的DMF。

  • 关于指定起始原料和中间体的定义和标准,ICH行业指导原则Q7《活性药物成分良好生产规范指导原则》(修订版1)15和Q11《药物物质的开发与生产》以及ICH Q7和Q11相应的《问题与解答指导原则》均有论述。药物制剂生产商应根据其稳定性规程收集稳定性数据,并应继续在DMF的质量/稳定性修正案中提交正在进行的研究数据,参见ICH(国际人用药品技术要求协调理事会)的行业指南Q1A(R2)《新药物质和产品的稳定性测试》。

  • 对于拟用于无菌产品的药物物质的灭菌,应根据CDER DMF网页上的行业指南《人用和兽用药物产品申请中灭菌工艺验证的提交文件》和其他支持文件,提交与无菌药物产品相同的无菌保证信息。如果建筑和设施信息(包括平面图)没有交叉引用V类DMF中的信息,则可以在II型DMF中提交。如果用于制备药物物质或药物物质中间体的材料需要FDA审查CMC信息(如定义的人工细胞生长培养基),则应在II型DMF中提交该信息。

  • 与提交监管文件一样,无论是支持临床评估还是批准上市申请,DMF都要提交年度报告。这些报告有助于向FDA保证承诺声明是最新的。如果不提交年度报告,该机构可能会关闭DMF。此外,该报告不应用来报告DMF的变化。在某些情况下,可能有必要根据eCTD序列号提交DMF修订案和年度报告。

  • 非专利产品的II型DMF要求有三个不同之处。对于在ANDA中引用的II型DMF,根据《2012年仿制药用户费用修正案》(通常称为GDUFA),DMF持有者在首次授权在仿制药申请中引用其DMF时需要支付费用。II型DMF必须接受FDA的完整性评估(CA)。2017年10月修订版1.0指南就DMF中应包含的信息提出了建议,以便于进行GDUFA CA。该指南不适用于用于NDA或其他非仿制药申报的II型DMF,也不适用于任何其他类型的DMF。

  • 另外,对于在ANDA或ANDA的事先批准补充(PAS)中引用的II型DMF,作为重新授权 GDUFA的谈判的一部分,21 FDA同意对计划进行改进,允许DMF持有者在某些情况下要求进行早期审查,即“DMF事先评估”。II型DMF的早期评估将在引用该DMF的ANDA或PAS提交前六个月开始。早期评估不适用于用于支持NDA的II型API DMF、指南中未描述的与ANDA有关的呈文或任何其他类型的DMF。

  • GDUFA规定的另一项仅适用于ANDA的重要计划改进是,ANDA中引用的II型DS DMF的持有者可以请求召开电话会议,以回复第一周期DMF缺陷函。



— 结论 

注册活性物质允许持有人向监管机构披露与用于生产、包装和储存产品的设施、工艺或材料有关的有价值的机密或专有信息和“技术诀窍”,而无需披露申请人/赞助商的机密信息。此外,持有人在注册活性物质时,还必须评估财务影响/价值。因此,活性物质的注册申请完全由持有人自行决定。



声明:原英文原文版权归美国法规事务协会(RAPS)所有,本译文供参考,如有任何建议,请联系我们。



英文版

(上下滑动查看更多)

Chapter 12 The Global Regulatory Process for the Registration of Active Substances


There are two key objectives when introducing a new drug. One is obtaining marketing approval with a successful application in a drug’s lifecycle. The application process

encompasses marketing authorization applications (MAAs), New Drug Applications (NDAs), or Biologic License Applications (BLA), along with the registration of active substances detailing how the product is manufactured and controlled, summarizing years of testing. The second is creating unique value by developing and obtaining intellectual property in the country or countries where the drug will be marketed.


The active pharmaceutical ingredient (API)—central to developing a medicinal product—is the drug substance that delivers the intended health benefit. API developers and manufacturers invest many hours at high costs to characterize the API and define the manufacturing process, whether chemical drugs or biologics. Registration of active substances provides comprehensive information about the drug, enabling regulatory agencies and healthcare practitioners to ensure that therapeutic benefits outweigh the risks. Regulators assess quality, safety, and efficacy, and evaluate the ability of the future marketing license holder to ensure and monitor a sustainable benefit-risk ratio. In addition to summarizing quality, nonclinical and clinical data, marketing application submissions also lay bare how the safety reporting system transitions from development to the postapproval phase.


When companies are looking to establish a global regulatory strategy, they need to define which countries or economic blocks to register the proposed active substances. In 2020, 10 countries were identified as top pharmaceutical markets, as shown in Figure 11-1.2 Nowadays, the global market is grouped into economic blocks. Therefore, the top pharmaceutical countries can be grouped into the following blocks (Figure 11-2):


• North America: US;

• EU: France, Germany, Italy, and Spain;

• BRIC: Brazil and China;

• Trans-Pacific Partnership: Japan; and

• Commonwealth: Canada and UK.


Raw materials used to manufacture APIs and the API manufacturing process itself are subject to tight controls, and global regulations including current good manufacturing practices (cGMPs) to manufacture a quality ingredient that can be formulated into a medicinal product and used safely. The Chemistry, Manufacturing and Controls (CMC) described in a regulatory dossier to support clinical investigation or marketing authorization must provide a full description of the API, among other essential information described in Chapters 11, 12, 13, and 14.


The regulatory requirement for filing Drug Master File (DMF) and review process for API producers to get approved can vary from country to country. Table 11-1 shows the requirements for countries such as the US, EU, Japan, and China. Information in this chapter reflects the situation in the US, Canada, the UK, Brazil, China, and Japan.


Global Regulatory Requirements 


US


The provision for the submission of a DMF is codified in 21 CFR Part 314.420. In October 2019 the Food and Drug Administration (FDA) issued Revision 1 of the Drug Master Files Guidance for Industry updated from the agency’s September 1989 guidance document.3 This guidance describes the FDA’s current thinking on DMFs and does not establish legally enforceable requirements.


The guidance defines DMFs as “submissions to FDA that may be used to provide confidential, detailed information about facilities, processes or articles used in the manufacture, processing, packaging, and storing of human drug products.” DMFs are not typically submitted for nonproprietary information.


There are four types of DMFs defined in the guidance:


1.Type II includes information for the drug substance, drug substance intermediate, and materials used in their manufacture;

2.Type III for packaging material;

3.Type IV for excipient, colorant, flavor, essence, or material used in their preparation; and

4.Type V for FDA-accepted reference information. 


DMFs are required to be an accurate and complete English translation; a certified translation is not required. In general, the FDA expects the DMF holder to be the manufacturer of the material covered by the DMF. If the DMF holder is not the manufacturer, the DMF should include a statement that the DMF holder assumes full responsibility for the manufacturing of the material. The manufacturer of any material can decide to submit the information necessary for review directly to a customer for submission the investigational new drug (IND), the NDA, the Abbreviated New Drug Application (ANDA), another DMF, and amendments and supplements to regulatory applications. The FDA allows for cross-reference to the DMF or most of Module 32S, except forModules 1, 2.1, Module 4.1, and Module 6.4 Also, data require- ments are the same regardless of submission via DMF or direct inclusion in the market application.


The FDA’s guidance provides detailed information to prepare and submit DMFs, the information expected in DMFs, and the agency’s review process. Additional information is provided on the FDA’s DMF webpage.5 The agency updates its DMF list on a quarterly basis. There were more than 35,000 DMFs listed as of June 2022. The list includes a small number (490) of Type I DMFs; however, Type I’s were discontinued in 2000 but the numbering of the other DMF types did not change. Of the number of DMFs on the FDA’s list, the largest number of DMFs are Type II (roughly 24,800). The next largest number of DMFs are Type III with approximately 6,500. Type II DMFs are the most widely used DMF type. When considering a Type III DMF for your API or referencing a Type III DMF as the sponsor/applicant of your regulatory submission, the following are important elements from the guidance.


A Type II DMF must be submitted in the standard electronic common technical document (eCTD) format in accordance with the Guidance for Industry Providing Regulatory Submissions in Electronic Format-Certain Human Pharmaceutical Product Applications and Related Submissions using the eCTD Specifications (Rev. 6). The FDA’s eCTD submission standards with a complete list of eCTD section headings is provided on the agency’s website.6 Additional information and ICH guidelines for

the CTD are also available. A DMF of 10 GB or smaller must be submitted through the FDA’s Electronic Submission Gateway (ESG). The majority of DMFs fall into this category. Submissions of more than 10 gigabytes can also be submitted through the ESG or can be delivered prepaid on physical media, such as a CD or thumb drive. The FDA announced in June 2019 a proposed rule (84 FR 30968) related to Biologics License Applications (BLA) and Master Files.7 The proposed rule, when final, codifies three specific aspects:


It allows certain biological products, originally approved in a New Drug Application (NDA) under the Federal Food, Drug, and Cosmetic (FD&C) Act, to continue relying on a DMF for information on a drug substance (DS), drug substance intermediate (DSI), or drug product (DP) after the NDA is deemed to be a license for a biological product under the Public Health Service (PHS) Act.8 A BLA under the PHS Act may rely on a Master

File except for information on DS, and DSI defined as material produced during the steps of API manufacturing that undergo further molecular change or purification after it becomes an API or DP (finished dosage form, e.g., tablet, capsule). The PHS Act also codified the FDA’s practice of permitting investigational new drug applications to incorporate by reference any information contained in a Master File for products subject to licensure. The agency generally expects drug substance information to be submitted to the BLA in the relevant sections of the application.


Because DMFs are not approved or disapproved, an environmental assessment is not required to be submitted in a DMF (eCTD Section 1.12.14).3 However, DMF holders should include a commitment in the DMF to operate their facilities in compliance with applicable environmental laws. This statement can also be included in the applicants/sponsors regulatory submission as part of Appendix A.2.1.


Before submitting the original Type II DMF in eCTD format, the DMF holder must request a pre-assigned number from the FDA’s Center for Drug Evaluation and Research (CDER).10 Original submissions must include a cover letter and complete administrative and technical information in the appropriate eCTD modules. For a Type II DMF, Module 3 is the most relevant section, with Module 3 information summarized in Module 2,Quality Overall Summary. Module 4, Nonclinical, is not typically necessary for a Type II DMF unless nonclinical evaluations to support safety were conducted, such as for impurities.


In the cover letter, it is critical to include a statement of commitment signed by the DMF holder that the DMF is current, and the holder will comply with the statements made in the DMF.Only one company should be listed as the DMF holder; joint submissions are not accepted. Detailed information of the contact person and responsible official, if different from the contact person, should be provided including a name, title, phone numbers, fax numbers (if available), a secure email address, and a description of specific responsibilities. The FDA recommends using the cover letter template found on the CDER’s webpage for original and subsequent submissions.3 To facilitate communication with the FDA, it is important for foreign DMF holders to appoint an agent, preferably in the US, who is familiar with agency regulations, guidance, and procedures.


Agents are not responsible for the contents of the DMF; this responsibility cannot and should not be delegated to the agent. The agent can submit on behalf of the DMF holder and can sign DMF submissions with the following exceptions:


• Agent appointment letters

• Statement of commitment

• Name changes

• Holder transfers

• New holder acceptance letters

• DMF closure requests


The guidance further clarifies that the agent for a DMF is not the same as the agent for the purposes of the Drug Listing and Registration System (DRLS); this person should not be listing for registration purposes in the DMF unless they are also the agent for the DMF and DRLS. DMF holders can have different agents for different DMFs.


In earlier years, DMFs were submitted to the FDA in advance of being referenced in a sponsor/applicant application.


More recently, DMFs are submitted closer to or in parallel with applications to support clinical evaluation or product approvals. When a DMF is submitted, the FDA will review the file for administrative information and send an acknowledgement letter to the DMF holder (and agent, if applicable), listing the DMF number, subject (title), type (I, II, III, or IV), and holder’s name, as specified in the DMF cover letter. If administrative information is incomplete, the FDA contacts the holder (and agent, if applicable) to request the missing info. Until all administrative information filing issues are adequately addressed and the DMF is referenced in an application or another DMF, the DMF is not available for technical review. A key document in the administrative information is the debarment certification. DMF holders are included in the category of “Persons whose services were used in any capacity in connection with the application” required under Section 306(k)(1) of the FD&C Act. DMF holders can submit their own debarment certifications in eCTD Section 1.3.3.9 More information on debarment certifications is included in the FDA’s draft Guidance for Industry Submitting Debarment Certification Statements.11


The most critical document to the successful use of the DMF is the Letter of Authorization (LoA). It is key to a complete review of the referenced technical information in the DMF. Until an LoA is submitted to the DMF for a specific application or other DMF (21 CFR Part 314.420[d]),12 the FDA will not review the DMF for technical information. The LoA permits the FDA to review the DMF and permits the authorized party (i.e., the company or individual who applies, or another DMF)to incorporate information into an application or another DMF by reference (eCTD Section 1.4.1). Reviewers may find that additional information is needed to continue a review, or that the DMF cannot be used to support approval of an ANDA or NDA, or in the case of an IND, allow clinical trials to proceed.


In these cases, the FDA will contact the DMF holder and the agent, if applicable, regarding the concerns. This situation may pose a challenge to applicants/sponsors if the DMF holder does not share the request or comments. It is critical that the Quality Agreement with the DMF holder include open and specified communication timelines regarding comments and requests from regulatory authorities from a DMF review so any impact or delays to approval or clinical trial start can be addressed, mitigated,and communicated internally by the applicant/sponsor. Further, it is important for the applicant/sponsor to be aware of any commitments made by the DMF holder that may impact the medicinal product.


DMF holders can authorize one or more applicants/sponsors to incorporate by reference the information contained in the DMF. An LOA is submitted even if the authorized party and the DMF holder are the same company. However, an applicant does not need to create a new DMF when referencing its own material and can include the information directly in its own application.


The DMF holder sends a copy of the LoA to the authorized party, and the authorized party must include a copy of the LoA in the application (21 CFR Part 314.50[a][1])13 or DMF (eCTD Section 1.4.2).9 The FDA recommends the use of its LoA template on the CDER’s webpage.14 Importantly, an LoA does not give an authorized party permission to view, review, or access a DMF. In eCTD Section 1.4.3,9 a list of parties currently authorized to incorporate by reference any information in the DMF must be included, and revised and submitted when additional parties are authorized or withdrawn.

The above information is applicable to all four DMF types. For Type II DMFs, specifically DS, DSIs, and materials used in their preparation and storage, a copy of the shipping label to the site or sites(s) for forward processing should be provided in the eCTD Section 1.149 on labeling.


Each Type II DMF should be submitted for a single drug substance, drug substance intermediate, and type of material used in their preparation, and include a statement that the material covered by the DMF is manufactured under cGMPs. Separate DMFs should also be submitted for drug substances manufactured using different processes.


The definition and criteria for designating starting materials and intermediates are discussed in ICH guidance for industry Q7 Good Manufacturing Practice Guidance for Active Pharmaceutical Ingredients (Rev. 1)15 and Q11 Development and Manufacture of Drug Substances16 and ICH Q7 and Q11’s corresponding Questions and Answers guidance.17,18 Drug substance manufacturers should collect stability data according to their stability protocol and should continue to submit data from ongoing studies in a quality/stability amendment to the DMF, see guidance for industry, Q1A(R2) Stability Testing of New Drug Substances and Products,19 from ICH, the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use.


For sterilization of drug substances to be used in sterile products, the same sterility assurance information should be submitted as for sterile drug products in accordance with guidance for industry, Submission Documentation for Sterilization Process Validation in Applications for Human and Veterinary Drug Products,20 and other supporting documents included on the CDER’s DMF webpage. Building and facility information, including floor plans, can be submitted in a Type II DMF if a Type V DMF is not cross-referenced for the information. If material used in the preparation of a drug substance or drug substance intermediate requires FDA review of CMC information (e.g., defined artificial cell growth media), the information should be submitted in the Type II DMF.


As with a regulatory submission, whether supporting clinical evaluation or an approved market application, annual reports are submitted for DMFs. These reports help assure the FDA that the statement of commitment is current. Failure to submit the report annually may result in closure of the DMF by the agency. Further, this report should not be used to report changes to the DMF. In some cases, it may be necessary to submit an amendment to the DMF and an annual report under eCTD sequence numbers.


There are three differences in Type II DMF requirements for generic products that do not apply to nongeneric products. For Type II DMFs referenced in ANDAs, under the Generic Drug User Fee Amendments of 2012,21 commonly referred to as GDUFA, DMF holders are required to pay a fee when first authorizing the reference of their DMF in a generic application. Type II Drug Substance DMFs must undergo an FDA completeness assessment (CA). The October 2017 Rev 1 guidance makes recommendations about the information that should be included in the DMF to facilitate a GDUFA CA. The guidance does not apply to Type II Drug Substance DMFs used for NDAs or other submissions that are not generic drug submissions, or any other types of DMFs.


Also, for Type II DMFs referenced in ANDAs or a prior approval supplement (PAS) to an ANDA, as part of negotiations to reauthorize GDUFA,21 the FDA agreed to a program enhancement that allows for instances when an early review,“DMF prior assessment,” could be requested by a DMF holder. The early assessment for a Type II DMF would start six months prior to an ANDA or PAS submission referencing the DMF. The early assessment does not apply to Type II API DMFs used to support NDAs, submissions related to ANDAs not described in the guidance, or any other type of DMF.


Another important program enhancement under GDUFA that only applies to ANDAs is the provision that for holders of Type II DS DMFs referenced in ANDAs can request teleconferences in response to first cycle DMF deficiency letters.


Conclusion


Registering active substances allows the holder to disclose valuable confidential or proprietary information and “know-how” to the regulatory authorities related to facilities, processes, or materials used to manufacture, package, and store product without disclosing confidential information from the applicants/sponsors. One additional component, the holder must also evaluate the financial impact/ value

when registering substances to the corporation. Therefore, these applications are solely at the discretion of the holder.






同写意媒体矩阵,欢迎关注↓↓↓





同写意
一个以新药创制为核心,汇集国内一流研发精英的大型交流与价值分享平台。“同筑技术人生路,写意中国新药魂”是同写意的初衷与愿景。秉承汇集“一线、一流”,讲求“实用、实在、时效”的原则。
 最新文章