Challenges and Opportunities in Enhancement of the CMC Section of NDAs: Quality – by - Design Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical.

Slides:



Advertisements
Similar presentations
FDA/Industry Statistics Workshop Washington D.C. September 27-29, 2006
Advertisements

Statistical Evaluation of Dissolution for Specification Setting and Stability Studies Fasheng Li Associate Director, Pharmaceutical Statistics Worldwide.
1 Dissolution Measurement System: Current State and Opportunities for Improvement Dr. Lucinda Buhse Director, Division of Pharmaceutical Analysis.
1 Implementation of Quality by Design (QbD): Status, Challenges and Next Steps Moheb M. Nasr, Ph.D. Office of New Drug Quality Assessment (ONDQA), OPS,
Integrating CMC Review & Inspection Industry Recommendations Joe Anisko April 24, 2003.
Chemistry, Manufacturing, and Controls (CMC) and Good Manufacturing Practices (GMPs): The Big Picture of a Long-term Commitment Elizabeth Pollina Cormier,
Division of Pharmaceutical Analysis Research in support of the Critical Path Dimensions Ensuring Safety Demonstrating Medical Utility Industrialization.
Determine impurity level in relevant batches1
Implementation of Quality-by-Design: ONDQA Initiatives Advisory Committee for Pharmaceutical Science October 5, 2006 Chi-wan Chen, Ph.D. Deputy Director.
Office of New Drug Chemistry, OPS, CDER, Food and Drug Administration Establishing Dissolution Specification Current CMC Practice Vibhakar Shah, Ph.D.
Overview of Guidance Documents and Decision process: Biopharmaceutics Section Mehul Mehta, Ph.D. Director Division of Pharmceutical Evaluation I OCPB,
Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical Science, CDER, FDA An Example of Process Understanding Directed Risk Based CMC Regulatory.
Manufacturing Subcommittee of the Advisory Committee for Pharmaceutical Science July 20-21, 2004 Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical.
Pilot Risk-Ranking Model to Prioritize Manufacturing Sites for GMP Inspections Advisory Committee for Pharmaceutical Science Manufacturing Subcommittee.
Assessing Quality-by-Design A CMC Review Perspective
Final Report on Process Analytical Technology (PAT) and Manufacturing Science Ajaz S. Hussain, Ph.D. Deputy Director, Office of Pharmaceutical Science,
PAT Validation Working Group Process and Analytical Validation Working Group Arthur H. Kibbe, Ph.D. Chair June 13, 2002.
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.
Learnings from Pre-approval Joint Inspection of a GSK QbD Product with US-FDA & EMA and the application of Continuous Verification 17 May 2011, Beijing,
Ensuring Physical Stability of Pharmaceuticals: Can/should we improve our ability to identify and prevent physical changes? Ajaz S. Hussain, Ph.D. Deputy.
Achieving and Demonstrating “Quality-by-Design” with Respect to Drug Release/dissolution Performance for Conventional or Immediate Release Solid Oral Dosage.
ACPS Advisory Committee Meeting October , 2002 ACPS Advisory Committee Meeting October , 2002 Scientific Considerations of Polymorphism in.
Quality by Design Application of Pharmaceutical QbD for Enhancement of the Solubility and Dissolution of a Class II BCS Drug using Polymeric Surfactants.
Establishing Drug release/Dissolution Specifications – QBD Approach Moheb M. Nasr, Ph.D. Office of New Drug Quality Assessment (ONDQA), OPS, CDER Advisory.
D. Christopher Watts, Ph.D. Office of Pharmaceutical Science, CDER, FDA Science Seminar Series for the Office of Commissioner April 9, 2004 Process Analytical.
Moving Towards the “Desired State”: Scientific Gap Analysis Ajaz S. Hussain, Ph.D. Deputy Director, Office of Pharmaceutical Science, CDER, FDA 20 October.
1 Supplements and Other Changes to an Approved Application By: Richard J. Stec Jr., Ph.D. February 7, 2007.
Ajaz S. Hussain, Ph.D. Office of Pharmaceutical Sciences CDER, FDA 17 September 2003 Quality by Design: Next Steps to Realize Opportunities?
Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical Science, CDER, FDA ACPS Subcommittee on Manufacturing Science: Identification and Prioritization.
FDA Regulation of Drug Quality: New Challenges Janet Woodcock, M.D. Director, Center for Drug Evaluation and Research, Food and Drug Administration April.
Blend Uniformity - PQRI Research
© 2011 Underwriters Laboratories Inc. All rights reserved. This document may not be reproduced or distributed without authorization. ASSET Safety Management.
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.
Introduction to Topic #1: QbD approach for quality control and assurance of Dissolution Rate Ajaz S. Hussain, Ph.D. Deputy Director, Office of Pharmaceutical.
Quality by Design (QbD) Myth : An expensive development tool ! Fact : A tool that makes product development and commercial scale manufacturing simple !
1 Regulatory Aspects of Pharmaceutical Excipients PQRI Workshop Nick Buhay Acting Director Division of Manufacturing and Product Quality Office of Compliance.
1 An Update on ICH Guideline Q8 – Pharmaceutical Development FDA Advisory Committee for Pharmaceutical Science: 5 Oct 2006 Dr John C Berridge Senior Regulatory.
Waiver of In Vivo Bioequivalence Studies for Immediate Release Solid Oral Dosage Forms Based on a Biopharmaceutics Classification System Ajaz S. Hussain,
Overview of FDA's Regulatory Framework for PET Drugs
Challenges and Opportunities in Enhancement of the CMC Section of NDAs: Quality – by - Design Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical.
1 Basis of the Proposed Tactical Plan for a QbD approach for Quality Control and Assurance of Dissolution Rate Ajaz S. Hussain, Ph.D. Deputy Director,
What Impact should ICH Q8 have on ICH Q6A Decision Trees?
PhRMA Perspective on FDA Final Report FDA Advisory Committee on Pharmaceutical Sciences October 20, 2004 G.P. Migliaccio, Pfizer Inc.
Risk-Based CMC Review - OGD Perspective Gary J. Buehler, R.Ph. Director Office of Generic Drugs July 21, 2004 Advisory Committee for Pharmaceutical Science.
Molecule-to-Market-Place Quality
The USP Performance Test Dissolution Systems Suitability Studies Walter W. Hauck, Ph.D. USP Consultant Presentation to Advisory Committee for Pharmaceutical.
COMPARABILITY PROTOCOLUPDATE ADVISORY COMMITTEE FOR PHARMACEUTICAL SCIENCE Manufacturing Subcommittee July 20-21, 2004 Stephen Moore, Ph.D. Chemistry Team.
Purdue University – Industrial and Physical Pharmacy - Morris Discussion Questions of Polymorphism in ANDAs Ken Morris Industrial and Physical Pharmacy.
Bioequivalence of Locally Acting Gastrointestinal Drugs: An Overview
10:00 A.M. – Noon 7 June 2004 ICH Quality Plenary Meeting.
Drug Release Specification: In Vivo Relevance Ajaz S. Hussain, Ph.D. Deputy Director, OPS/CDER/FDA.
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.
General Aspects of Quality assessment of multisource interchangeable medicines Rutendo Kuwana Technical Officer, WHO, Geneva Training workshop: Assessment.
Using Product Development Information to Address the Bioequivalence Challenges of Highly-variable Drugs Lawrence X. Yu, Ph. D. Director for Science Office.
Introduction What is a Biowaiver?
FDA Regulation of Drug Quality: New Challenges Janet Woodcock, M.D. Director, Center for Drug Evaluation and Research, Food and Drug Administration November.
Second Meeting of the FDA/ACPS Process Analytical Technology: Closing Remarks Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical Sciences.
Evaluation of quality and interchangeability of medicinal products - WHO Training workshop / 5-9 November |1 | Prequalification programme: Priority.
Examining Drug Quality Regulation Douglas C. Throckmorton, MD Deputy Director Center for Drug Evaluation and Research Public Meeting on 21 CFR February,
FDA Advisory Committee for Pharmaceutical Science and Clinical Pharmacology July 22-23, 2008 Introduction and Update Helen N. Winkle Director, Office of.
Topic #2: Quality by Design and Pharmaceutical Equivalence Ajaz S. Hussain, Ph.D. Office of Pharmaceutical Science Center for Drug Evaluation and Research.
开发报批美国 FDA 的仿制药 与相关问题探讨 上海复星普适医药科技有限公司何平. 内容提要 开发仿制药的重要性和机遇 开发仿制药的重要性和机遇 开发仿制药的挑战 开发仿制药的挑战 申报仿制药的分类 申报仿制药的分类 仿制药研发团队 仿制药研发团队 仿制药的研发过程 仿制药的研发过程 QbD 在制剂开发中怎么体现.
Product & Process Working Group February 26, 2002.
An Update on ICH Guideline – Pharmaceutical Development
Quality System.
Office of Pharmaceutical Science, CDER, FDA
Implementation of Quality by Design (QbD): Status, Challenges and Next Steps Moheb M. Nasr, Ph.D. Office of New Drug Quality Assessment (ONDQA), OPS, CDER.
Presentation transcript:

Challenges and Opportunities in Enhancement of the CMC Section of NDAs: Quality – by - Design Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical Science CDER, FDA

Opportunity ► For companies that acquire extensive understanding about their product and manufacturing process and share this with the regulators  Enhanced science and risk-based regulatory quality assessment will be possible ► Setting specifications ► Reduction in the volume of data to be submitted – replaced by more knowledge based submissions ► Flexible post approval continuous improvement

Janet Woodcock, M.D. May 19, 2004

Without adequate product and process development and/or knowledge sharing ► Without “design” consideration, high level of uncertainty with respect to critical attributes, “representative” test sample, and adequacy of risk coverage (e.g., compendail tests) to assure batch quality [Regulatory Concern/Risk] ► Reduce concern/risk by covering all apparent attributes with acceptance criteria based on capability of test methods and/or manufacturing process plus very inflexible SOP’s [Current Regulatory Risk Mitigation Strategy]

To illustrate the current state: A “Case Study” – Dissolution Attribute The “case study” attempts to connect many dots ICH Q6A ICH Q8 Dissolution SUPAC BCS BA/BE Desired State Current State Testing to Document Quality Quality By Design

ICH Q6A DECISION TREES #7: SETTING ACCEPTANCE CRITERIA FOR DRUG PRODUCT DISSOLUTION What specific test conditions and acceptance criteria are appropriate? [IR] dissolution significantly affect BA? Develop test conditions and acceptance distinguish batches with unacceptable BA YES NO YES NO YES NO Do changes in formulation or manufacturing variables affect dissolution? Are these changes controlled by another procedure and acceptance criterion? Adopt appropriate test conditions and acceptance criteria without regard to discriminating power, to pass clinically acceptable batches. Adopt test conditions and acceptance criteria which can distinguish these changes. Generally, single point acceptance criteria are acceptable. How? What? Why? How do we currently establish dissolution specifications

Without adequate product development and/or knowledge sharing we debate frequently …… And then, have CGMP problems?

This can be catastrophic for the business and availability of Important drugs A Warning Letter

OOS or Exceptions Further Increase Cycle Times (Source: G. K. Raju, M.I.T. FDA Science Board Meeting, November 16, 2001) Dissolution

Testing to Document Quality: Requires Less Variable Test Methods ► The current USP 10-mg Prednisone Calibrator Tablets exhibit slower dissolution over time ► If the acceptable test equipment calibration limit is 28-54; what can we say about use of f2 criteria (~mean profile difference of 10%) as a way to document unchanged quality (e.g., SUPAC)? LotDate Mean (n=6) SD (%) USP Limit (%) M4/ M10/ N12/ N11/ N6/ DPA/FDA Data using Apparatus 2; data from only one apparatus shown. Note the USP adjusts the limits of each new lot of calibration tablets to reflect the anticipated decrease in dissolution.

Dissolution Experience at the FDA Division of Pharmaceutical Analysis ► Dissolution testing with USP Apparatus 1 and 2 requires diligent attention to details: mechanical and chemical ► Dosage forms can respond differently to small variations in apparatus set up or degassing ► Large differences in dissolution results are possible unless all parameters are carefully controlled ► The experience at DPA indicates that differences in reproducibility can often be traced to improper mechanical calibration and/or degassing

False Positives and False Negatives!!! Test/Ref. Mean I. J. MacGilvery. Bioequivalence: A Canadian Regulatory Perspective. In, Pharmaceutical Bioequivalence. Eds. Welling, Tse, and Dighe. Marcel Dekker, Inc., New York, (1992)).

Do we need a dissolution specification of every solid oral drug product? No ICH Q6A: Decision Tree #7 (1) Modified release? High solubility? Rapid dissolution? Relationship between Disintegration - Dissolution? No Yes Generally single-point dissolution acceptance criteria with a lower limit Generally disintegration acceptance criteria with an upper limit Establish drug release acceptance criteria: ER: Multiple time point MR: Two stage, parallel or sequential Yes No Yes How? Test – Test Comparison?

Disintegration - Dissolution Relationship 10# screen Fraction dissolved Note: Disintegration and dissolution process in a dissolution apparatus may differ from that in a disintegration apparatus (different hydrodynamics and other conditions)

“Testing to Document Quality” ► The phrase has many dimensions  In-process and end-product release and stability testing  Reliability of specifications (attribute, test method, and acceptance criteria)  Managing post approval changes/continuous improvement (e.g., reduce variability, improve efficiency,..)  Product and process knowledge acquisition and generalization

How can pharmaceutical development knowledge help? ► Demonstrate quality was designed in? ► Specifications based on mechanistic understanding? ► Continuous "real time" assurance of quality? ► Flexible continuous improvement?

ICH Q6A DECISION TREES #7: SETTING ACCEPTANCE CRITERIA FOR DRUG PRODUCT DISSOLUTION What specific test conditions and acceptance criteria are appropriate? [IR] dissolution significantly affect BA? Develop test conditions and acceptance distinguish batches with unacceptable BA YES NO YES NO YES NO Do changes in formulation or manufacturing variables affect dissolution? Are these changes controlled by another procedure and acceptance criterion? Adopt appropriate test conditions and acceptance criteria without regard to discriminating power, to pass clinically acceptable batches. Adopt test conditions and acceptance criteria which can distinguish these changes. Generally, single point acceptance criteria are acceptable. Overall Risk-based CMC: Why? Overall CMC Systems approach (e.g., link to morphic form, particle size, stability failure mechanisms) CMC: Why? Then How? Clin. Pharm. What? Design of Manufacturing and Controls How (reliable)? Product Design (Postulate - Confirmed Based on mechanism and/or empirically) So what?

Based on Quality of Pharmaceutical Development Knowledge can we not evaluate ► ► Overall CMC Systems approach (e.g., link to morphic form, particle size, stability failure mechanisms) and address concerns and risks  Is a dissolution specification needed?  Instead of wet dissolution test, can we use disintegration test?  Real time release and stability based on process controls, and NIR test for capsules?

Not all information “mandatory” ► We are okay with this ► But we wish to avoid confusion and the potential vocabulary that may evolve from this – “two different systems” ► Instead we see this as one system with different levels of QbD  we will use the “process understanding – predictive ability” vocabulary as a means to create a continuous framework and avoid “two different systems” March 2004 ICH Q8 Meeting: FDA’s Goals

Challenges we face ► Common approach to, and more clear articulation of  Not all information “mandatory”  Improved process understanding and control technologies may be afforded reductions in regulatory requirements  An inverse relation is expected between the effectiveness of the Quality by Design and the risk to a patient being exposed to product that is not fit for use  Ensuring continuous improvement and a process for continuous learning and updating of the knowledge base March 2004 ICH Q8 Meeting: FDA’s Goals