1 DRUG QUALITY SYSTEM FOR THE 21 ST CENTURY PQRI/FDA April 22-24 CHANGES WITHOUT PRIOR APPROVAL AN FDA PERSPECTIVE Dennis M. Bensley, Jr., Ph.D. Center.

Slides:



Advertisements
Similar presentations
U.S. Food and Drug Administration Notice: Archived Document The content in this document is provided on the FDAs website for reference purposes only. It.
Advertisements

Assures that feed… –has the identity and strength, which it purports –meets the quality, purity, and safety requirements, which it is represented to possess.
Fundamentals of Regulatory Affairs eighth edition
Strengthening the Medical Device Clinical Trial Enterprise
Development of Guidance Documents Jennifer Scharpf, M. P. H
VALIDATION What is the new guidance?. What is a Compliance Policy Guide? Explain FDA policy on regulatory issues CGMP regulations and application commitments.
Batch Reworking and Reprocessing
CFSAN’s Peer Review for Risk Assessments Robert L. Buchanan, Sherri Dennis, and Marianne Miliotis.
Integrating CMC Review & Inspection Industry Recommendations Joe Anisko April 24, 2003.
The ICH E5 Question and Answer Document Status and Content Robert T. O’Neill, Ph.D. Director, Office of Biostatistics, CDER, FDA Presented at the 4th Kitasato-Harvard.
Office of New Drug Chemistry (ONDC)
CBER Cooperative Manufacturing Arrangements (Contractors) Jennifer Jones Consumer Safety Officer CBER, OBRR, DBA September 15, 2009.
Manufacturing Subcommittee of the Advisory Committee for Pharmaceutical Science July 20-21, 2004 Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical.
COMPARABILITY PROTOCOLS ACPS March 12-13, 2003 Stephen K. Moore, Ph.D. Chemistry Team Leader CDER/Office of New Drug Chemistry Co-Chair, Comparability.
CBER Managed Review Process Sheryl A. Kochman Deputy Director, DBA, OBRR, CBER September 15, 2009.
Investigational New Drug Application 21 CFR Part 312 A Review for OCRA US RAC Study Group September 2005 Ginger Clasby, MS Promedica International
1 MEASURING THE EFFECTIVENESS OF THE NATION’S FOODSERVICE AND RETAIL FOOD PROTECTION SYSTEM.
Animal Feed GRAS Notifications Geoffrey K. Wong, M.S. Division of Animal Feeds Center for Veterinary Medicine Pet Food Institute Pet Food Institute October.
Mrs. Brandi Robinson Office of New Animal Drug Evaluation Center for Veterinary Medicine Regulating Animal Drugs.
Classification of HLA Devices FDA Introduction & Background Sheryl A. Kochman CBER/OBRR/DBA.
ONDQA Perspective on Post Approval Changes Eric P. Duffy, PhD Director, Division of Post-Market Evaluation, ONDQA, CDER, FDA Public Meeting: Supplements.
1 ACPS November 15, Update Nancy B. Sager, Associate Director Office of Pharmaceutical Science Center for Drug Evaluation & Research Food and.
1 Revisions to 21 CFR Supplements and Other Changes to an Approved Application PhRMA Perspective FDA Public Meeting – 7 Feb 2007.
FDA’s Perspective Continued - Where We Are ?. GMP Task Groups.
Requirements for Standardized Study Data: Update on Guidance Ron Fitzmartin, PhD, MBA Data Standards Program Office of Strategic Programs Center for Drug.
Executive summary prepared by some members of the ICH Q9 EWG for example only; not an official policy/guidance July 2006, slide 1 ICH Q9 QUALITY RISK MANAGEMENT.
FDA Guidance for Industry: Use of Serological Tests to Reduce the Risk of Transmission of Trypanosoma cruzi Infection in Whole Blood and Blood Components.
Achieving and Demonstrating “Quality-by-Design” with Respect to Drug Release/dissolution Performance for Conventional or Immediate Release Solid Oral Dosage.
Center for Veterinary Medicine (CVM) RECALLS.  21 CFR 7.40 provides guidance on the policy, procedures, and industry responsibilities for recalls. 
Nonclinical Studies Subcommittee Advisory Committee for Pharmaceutical Science CMC Issues for Screening INDs Eric B. Sheinin, Ph.D. Acting Deputy Director.
CBER 1 Establishment Submissions Rosia E. Nesbitt, BS, SBB(ASCP), CQA(ASQ) Consumer Safety Officer CBER, OBRR, DBA September 15, 2009.
1 Supplements and Other Changes to an Approved Application By: Richard J. Stec Jr., Ph.D. February 7, 2007.
MB RESEARCH LABORATORIES Increased Regulatory Vigilance with respect to GLP Test Article Characterization George L. DeGeorge, Ph.D., DABT MB Research Laboratories.
Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical Science, CDER, FDA ACPS Subcommittee on Manufacturing Science: Identification and Prioritization.
Investigational New Drug Application (IND)
Changes without Prior Approval Breakout Session Summary Rick Smith Aventis Pasteur, Inc.
U.S. Food and Drug Administration Notice: Archived Document The content in this document is provided on the FDA’s website for reference purposes only.
1 PAT and Biological Products Tom Layloff FDA-SGE Management Sciences for Health The views expressed here are those of the author and not necessarily.
FDA’s Perspective on the “Pharmaceutical cGMPs for the 21st Century” Initiative David J. Horowitz, Esq. Director, CDER/FDA, Office of Compliance Advisory.
1 Regulatory Aspects of Pharmaceutical Excipients PQRI Workshop Nick Buhay Acting Director Division of Manufacturing and Product Quality Office of Compliance.
FDA Public Meeting on Electronic Records and Signatures June 11, 2004 Presentation of the Industry Coalition on 21CFR Part 11 Alan Goldhammer, PhD Chair.
Preventive Controls for Human Food S upplemental Proposal 1
Foreign Supplier Verification Programs Supplemental Proposal 1.
Proposal for End-of-Phase 2A (EOP2A) Meetings Advisory Committee for Pharmaceutical Sciences Clinical Pharmacology Subcommittee November 17-18, 2003 Lawrence.
Overview of FDA's Regulatory Framework for PET Drugs
PhRMA Perspective on FDA Final Report FDA Advisory Committee on Pharmaceutical Sciences October 20, 2004 G.P. Migliaccio, Pfizer Inc.
COMPARABILITY PROTOCOLUPDATE ADVISORY COMMITTEE FOR PHARMACEUTICAL SCIENCE Manufacturing Subcommittee July 20-21, 2004 Stephen Moore, Ph.D. Chemistry Team.
REGULATION OF COMBINATION PRODUCTS Mark A. Heller Wilmer Cutler Pickering Hale and Dorr LLP MassMEDIC Combination Product Program, March 28, 2006.
IAEA International Atomic Energy Agency Methodology and Responsibilities for Periodic Safety Review for Research Reactors William Kennedy Research Reactor.
Comparability Protocols Lore Fields MT(ASCP)SBB Consumer Safety Officer DBA/OBRR/CBER September 16, 2009.
Investigational Devices and Humanitarian Use Devices June 2007.
Progress in FDA’s Drug Product Quality Initiative Janet Woodcock, M.D. November 13, 2003.
1 Office of Pharmaceutical Science on Jon Clark FDA/CDER/OPS Associate Director for Policy Development.
Evaluate Phase Pertemuan Matakuliah: A0774/Information Technology Capital Budgeting Tahun: 2009.
Pharmaceutical Manufacturing Subcommittee of the ACPS Ajaz S. Hussain, Ph.D. ACPS Meeting October 22, 2002.
CDER / Office of Compliance ACPS October 5, 2006 Joseph C. Famulare Acting Deputy Director Office of Compliance CDER / FDA.
Comparability Protocols Nancy Sager Associate Director, QIS-Chemistry FDA/CDER/OPS.
Examining Drug Quality Regulation Douglas C. Throckmorton, MD Deputy Director Center for Drug Evaluation and Research Public Meeting on 21 CFR February,
DEPARTMENT OF HEALTH CENTER FOR BIOLOGICS AND HUMAN SERVICESEVALUATION and RESEARCH AND HUMAN SERVICES EVALUATION and RESEARCH Update on OCTGT Guidance.
EXCEPTION FROM INFORMED CONSENT IN CPR DEVICE TRIALS: PROTECTION OF PATIENTS’ RIGHTS Circulatory System Devices Panel Meeting September 21, 2004 Elisa.
DEPARTMENT OF HEALTH CENTER FOR BIOLOGICS AND HUMAN SERVICESEVALUATION and RESEARCH AND HUMAN SERVICES EVALUATION and RESEARCH Update on the Somatic Cell.
Guidance Development Merton V. Smith, Ph.D., J.D. Director International Programs and Product Standards Center for Veterinary Medicine, FDA.
POST APPROVAL CHANGE MANAGEMENT PROTOCOLS IN THE EUROPEAN UNION
Introduction Review and proper registration of Human Gene Transfer protocols is very complex. A protocol goes through rigorous review by multiple Committees.
נמטוציטים משושנת ים Eli. S Lec. No.2.
CHANGE CONTROL.
Premarket Notification 510(k) process
FDA’s IDE Decisions and Communications
Pre-Approval-Inspection
Quality System.
Presentation transcript:

1 DRUG QUALITY SYSTEM FOR THE 21 ST CENTURY PQRI/FDA April CHANGES WITHOUT PRIOR APPROVAL AN FDA PERSPECTIVE Dennis M. Bensley, Jr., Ph.D. Center for Veterinary Medicine, FDA

2 OUTLINE INTRODUCTION BACKGROUND CURRENT FDA ASSESSMENT CURRENT RISK ANALYSIS COMPARABILITY PROTOCOL STRATEGIC GOALS CONCLUSION

3 INTRODUCTION Changes Without Prior Review working group established by FDA’s Drug GMP Steering Committee Working Group members: -Ajaz Hussain, Co-chair (CDER) -Nancy Sager, Co-chair (CDER) -Kathy Jordan, Project Manager (CDER) -Nick Buhay (CDER) -Chris Joneckis (CBER) -John Finkbohner (CBER) -Dennis Bensley (CVM) -Nancy Rolli (ORA) -Patricia Alcock (ORA) -Marybet Lopez (ORA) -Janet Showalter (OC, OIP) -Lynn Whipkey (OC, OCC)

4 INTRODUCTION Charge of Working Group: -examine current state of the supplemental change approval process, specifically those manufacturing changes requiring prior FDA approval.

5 INTRODUCTION Charge of Working Group: -examine current state of the supplemental change approval process, specifically those manufacturing changes requiring prior FDA approval. -identify and recommend implementation of other means to reduce reporting requirements, for example the use of risk management tools, comparability protocols, product development information, and process control improvements (e.g., PAT).

6 INTRODUCTION Purpose of Workshop -Present a summary of FDA’s current thinking and activities regarding the supplemental change approval process.

7 INTRODUCTION Purpose of Workshop -Present a summary of FDA’s current thinking and activities regarding the supplemental change approval process. -Stimulate discussion and constructive feedback from stakeholders regarding the current and desired future state of the change approval process.

8 BACKGROUND Legal Requirements - The applicant must notify FDA of each manufacturing change in accordance with section 506A of the Federal Food, Drug, and Cosmetic Act and, when finalized 21 CFR (CDER) and 514.8(CVM).

9 BACKGROUND Legal Requirements - The applicant must notify FDA of each manufacturing change in accordance with section 506A of the Federal Food, Drug, and Cosmetic Act and, when finalized 21 CFR (CDER) and 514.8(CVM). - An applicant must inform the FDA about each change in the product, production process, quality controls, equipment, facilities, responsible personnel, or labeling established in the approved license application(s) (CBER ).

10 BACKGROUND Legal Requirements - The applicant must assess the effects of any change on the identity, strength, quality, purity, and potency of the drug as they may relate to the safety and effectiveness of the drug before distributing product made with the change (Section 506A of Act, Section 116 FDAMA). - Four legal reporting categories under FDAMA: prior-approval, CBE (immediate), CBE-30 and annual report.

11 BACKGROUND Filing Categories: -Prior-approval (major change): Substantial potential to adversely affect the identity, strength, quality, purity, or potency of a product as they may relate to the safety or effectiveness of the product. Product made with change may not be distributed until approval.

12 BACKGROUND Filing Categories: -Prior-approval (major change): Substantial potential to adversely affect the identity, strength, quality, purity, or potency of a product as they may relate to the safety or effectiveness of the product. Product made with change may not be distributed until approval. -CBE or CBE-30 (moderate change): Moderate potential….Product distribution upon receipt of application to FDA (CBE) or 30 days after receipt of application to FDA, if acceptable (CBE-30).

13 BACKGROUND Filing Categories: -Prior-approval (major change): Substantial potential to adversely affect the identity, strength, quality, purity, or potency of a product as they may relate to the safety or effectiveness of the product. Product made with change may not be distributed until approval. -CBE or CBE-30 (moderate change): Moderate potential….Product distribution upon receipt of application to FDA (CBE) or 30 days after receipt of application to FDA, if acceptable (CBE-30). -Annual Report (minor change): Minimal potential….Annual reportable and immediate product distribution.

14 CURRENT FDA ASSESSMENT Did Section 116 of FDAMA (506A of the Act) meet the expectation of: -providing regulatory relief by lessening the reporting requirements of manufacturing changes without compromising the drug’s “quality”, safety or effectiveness?

15 CURRENT FDA ASSESSMENT The reporting of many manufacturing changes has been provided through regulations and guidances: -Section 506A of FFD&C Act (CDER, CBER and CVM) -Regulations (being revised , 514.8, ). -”Changes” Guidances: Changes to an Approved NDA or ANDA (CDER) Changes to an Approved Application: Biological Products (CBER) Changes to an Approved Application for Specified Biotechnology and Specified Synthetic Biological Products (CDER/CBER) Changes to an Approved NADA or ANADA (CVM) -Various PAC and SUPAC Guidances

16 CURRENT FDA ASSESSMENT Impact of FDAMA on Filing CVM A significant number of CMC changes are currently being reported in CBE supplements or in annual reports.

17 CURRENT FDA ASSESSMENT Impact of FDAMA on Filing CDER Data (%Prior Approval/CBE Supplements) FY99FY00FY01 NDAs Prior Approval62%48%39% CBE38%52%61% ANDAs Prior Approval82%26%34% CBE18%74%66%

18 NON-PDUFA PRODUCTSPDUFA PRODUCTS REPORTING CBER CMC CHANGES IMPACT OF FDAMA/ GUIDANCE

19 CURRENT FDA ASSESSMENT FDAMA has significantly reduced the reporting requirements for manufacturing changes, however FDA recognizes that there could be additional improvements in the change reporting process.

20 CURRENT FDA ASSESSMENT Concerns with current regulatory supplemental change process: -Though the relative percentage of prior- approval supplements as compared to other reporting categories have significantly decreased, the number of prior-approval supplements are starting to increase.

21 CURRENT FDA ASSESSMENT Concerns with current regulatory supplemental change process: -Though significantly reduced overall from “pre- FDAMA” times, the number of reported prior approval changes remain high for certain product types/processes (e.g., sterile products).

22 CURRENT FDA ASSESSMENT Concerns with current regulatory supplemental change process: -Recognize that any prior approval change could impact business planning and possibly impede innovation.

23 CURRENT FDA ASSESSMENT Concerns with current regulatory supplemental change process: -No guarantee that the prior approval supplement will be approved during “first round.” CVM’s experience is that approximately 40% of “first round” prior approval supplements are found to be incomplete (data from ).

24 CURRENT FDA ASSESSMENT Concerns with current regulatory supplemental change process: -Regulatory compliance dilemma in finding unacceptable, i.e., inadequately assessed or documented, CMC changes in CBE or annual reports.

25 CURRENT FDA ASSESSMENT Potential solutions identified by FDA to lessen reporting requirements: - Use of comparability protocols;

26 CURRENT FDA ASSESSMENT Potential solutions identified by FDA to lessen reporting requirements: - Use of comparability protocols; - Drafting and publishing more PAC/SUPAC guidance documents;

27 CURRENT FDA ASSESSMENT Potential solutions identified by FDA to lessen reporting requirements: - Use of comparability protocols; - Drafting and publishing more PAC/SUPAC guidance documents; - Identifying potential risk management tools;

28 CURRENT FDA ASSESSMENT Potential solutions identified by FDA to lessen reporting requirements: - Use of comparability protocols; - Drafting and publishing more PAC/SUPAC guidance documents; - Identifying potential risk management tools; - Encouraging the use of product development information and process control improvements (e.g., Process Analytical Technologies).

29 CURRENT RISK ANALYSIS SIGNIFICANT MODERATE MINIMAL HIGH SOME LOW YES NO* NO Potential for CMC Change To Adversely Affect Drug Levels of Risk Prior Approval Supplement? *CBE Supplements Required

30 CURRENT RISK ANALYSIS FDA’s determination of a CMC change as major or requiring prior approval: -What is the likely impact of the change on the identity, strength, quality, purity and/or potency of the drug product? If FDA believes there is a potential adverse affect, then major change.

31 CURRENT RISK ANALYSIS FDA’s determination of a CMC change as major or requiring prior approval: -Will additional clinical or other “non-CMC” (e.g., toxicological) studies be required? If yes, then likely major change.

32 CURRENT RISK ANALYSIS FDA’s determination of a CMC change as major or requiring prior approval: -Is the reported change either not well described, too complex or is the potential impact on the drug’s safety or effectiveness not certain? If yes, then likely major change.

33 CURRENT RISK ANALYSIS FDA’s determination of a CMC change as major or requiring prior approval: -If applicable, what is the current GMP status? If unacceptable, then likely major change.

34 CURRENT RISK ANALYSIS The question to address when a risk assessment is performed regarding a CMC change: What is the potential (risk) for the change to adversely affect the identity, strength, quality, purity, or potency of a product as they may relate to the safety or effectiveness of the product?

35 CURRENT RISK ANALYSIS The potential risk for a CMC change increases when the knowledge regarding the potential impact of the change decreases.

36 CURRENT RISK ANALYSIS The potential risk for a CMC change increases when the knowledge regarding the potential impact of the change decreases. What is the purpose of prior approval supplements for specified CMC changes?

37 CURRENT RISK ANALYSIS The potential risk for a CMC change increases when the knowledge regarding the potential impact of the change decreases. What is the purpose of prior approval supplements for specified CMC changes? -Identified major changes are those changes that have a substantial potential to adversely affect the drug.

38 CURRENT RISK ANALYSIS The potential risk for a CMC change increases when the knowledge regarding the potential impact of the change decreases. What is the purpose of prior approval supplements for specified CMC changes? -Identified major changes are those changes that have a substantial potential to adversely affect the drug. -Allows FDA time to review and concur (or not concur) with the proposed major change and its assessment prior to product distribution.

39 CURRENT RISK ANALYSIS FDA tends to be conservative in regard to accepting levels of risk, i.e., if we are not certain about the potential risk, then a higher filing category will likely be required.

40 CURRENT RISK ANALYSIS FDA employees use risk analysis daily, for example: -Deciding whether a change is major or moderate (30-day CBE assessment) -Deciding whether the assessment of the change is satisfactory or not (review process) -Deciding whether a cGMP inspection is required (e.g., CBER’s SOPP 8410: Determining When Prelicense/Preapproval Inspections Are Necessary)

41 CURRENT RISK ANALYSIS FDA employees use risk analysis daily, for example: -Deciding whether a change is major or moderate (30-day CBE assessment) -Deciding whether the assessment of the change is satisfactory or not (review process) -Deciding whether a cGMP inspection is required (e.g., CBER’s SOPP 8410: Determining When Prelicense/Preapproval Inspections Are Necessary) However, risk assessments for CMC changes are neither formalized nor uniformly structured throughout FDA.

42 CURRENT RISK ANALYSIS Possible ways to reduce the risk potential include the use of: 1. Comparability protocols Premise: FDA’s acceptance of proposed assessment of anticipated change will likely lessen risk for implementing the change and should lead to less burdensome reporting category

43 CURRENT RISK ANALYSIS Possible ways to reduce the risk potential include the use of: 2. An applicant may establish their own filing criteria based on developmental information in original or supplemental applications (“Make your own SUPAC”). Premise: Increase in scientific understanding/knowledge of change’s impact may lessen risk for implementing change and could lead to less burdensome reporting category

44 CURRENT RISK ANALYSIS Possible ways to reduce the risk potential include the use of: 3. incorporating significant process control improvements (e.g., PAT). Premise: Improvement in process controls, may lessen risk for producing poor product and could lead to less burdensome reporting category

45 CURRENT RISK ANALYSIS Can other risk analysis models be used to identify the level of risk for implementing CMC changes? For example, can we identify, through risk assessment, low-risk drugs, dosage forms, processes, etc. and significantly reduce the number of changes requiring prior approval before implementation?

46 COMPARABILITY PROTOCOL What is a CP? A well-defined, detailed written plan that prospectively specifies the tests and studies that will be performed, analytical procedures that will be used, and acceptance criteria that will be achieved to assess the effect of specific CMC changes for specific products.

47 COMPARABILITY PROTOCOL Draft Guidance for “small molecules” recently published (public comment by 6/25/03). CP described in regulations (current and/or proposed , and ). FDA believes that additional prior approval changes can be reported in CBEs or annual reports through the use of a CP.

48 COMPARABILITY PROTOCOL Uses and benefits of CP: -Can allow for a reduced reporting category of CMC changes covered by the approved CP. -CP can describe single or multiple-related CMC changes including those that may occur sequentially over a period of time.

49 COMPARABILITY PROTOCOL Uses and benefits of CP (cont.): -Earlier implementation of manufacturing changes. -Likely reduction in incomplete/deficiency letters issued by FDA (more “first- round” approvals), because the means of assessing the change has been approved in the CP.

50 COMPARABILITY PROTOCOL Uses and benefits of CP (cont.): -Allows sponsors to design own “changes” filing and documentation criteria based on experiences with the drug product or similar drug product, e.g. developmental studies. “Make your own SUPAC” concept.

51 COMPARABILITY PROTOCOL Uses and benefits of CP (cont.): -Allows sponsors to continually improve manufacturing processes without necessarily requiring prior FDA approval (potential for PAT implementation). -Reduces the potential risk for the change to adversely affect the drug. -A potential win-win situation for public, industry, and FDA.

52 COMPARABILITY PROTOCOL Limited CDER experience and no CVM experience. CBER experience: -More than 100 comparability protocols have been successfully used for CMC changes for all product classes since Submission of developmental information in CPs has convinced CBER to accept reduced reporting categories for some CMC changes.

53 Strategic Goals Publish Draft Comparability Protocol for “Large Molecules”

54 Strategic Goals Publish Draft Comparability Protocol for “Large Molecules” Finalize both CP Guidances.

55 Strategic Goals Publish Draft Comparability Protocol for “Large Molecules” Finalize both CP Guidances. Continue to amend or introduce new PAC/SUPAC guidances.

56 Strategic Goals Publish Draft Comparability Protocol for “Large Molecules” Finalize both CP Guidances. Continue to amend or introduce new PAC/SUPAC guidances. Publish final “changes” regulations , and

57 Strategic Goals Conduct study to evaluate existing data on prior approval changes and identify opportunities for further reducing of reporting categories: -Determination of the number and types of prior approval supplements submitted to each Center over a designated time period.

58 Strategic Goals Conduct study to evaluate existing data on prior approval changes and identify opportunities for further reducing of reporting categories: -Determination of the number and types of prior approval supplements submitted to each Center over a designated time period. -Identify other potential risk models or other means for reducing reporting categories.

59 Strategic Goals Conduct study to evaluate existing data on prior approval changes and identify opportunities for further reducing of reporting categories: - Determination of the number and types of prior approval supplements submitted to each Center over a designated time period. -Identify other potential risk models or other means for reducing reporting categories. -Consider additional ideas as result of discussion and feedback received during this workshop.

60 CONCLUSIONS The following are the discussion points for consideration during the individual breakout sessions:  Scientific risk-based approaches for identifying low risk manufacturing changes that can be implemented without prior FDA approval

61 CONCLUSIONS The following are the discussion points for consideration during the individual breakout sessions:  Draft guidance on comparability protocols for small molecules and development of a comparability protocol guidance for proteins

62 CONCLUSIONS The following are the discussion points for consideration during the individual breakout sessions:  Effective use of development data and other information to justify less burdensome filing requirements for post-approval manufacturing changes

63 CONCLUSIONS This is your opportunity to comment on and provide your ideas on the current supplemental approval process and your view of the ideal future state!