FDA’s Advisory Committee for Pharmaceutical Science The Subcommittee on Process Analytical Technologies (PAT): Overview and Objectives Ajaz S. Hussain,

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

ICH Q4B Regulatory Acceptance of Analytical Procedures and/or Acceptance Criteria (RAAPAC) Overview and Update Robert H. King, Sr. Office of Pharmaceutical.
Why does PAT need Rapid Microbiology Methods? S.Lonardi, P.J.Newby, D.Ribeiro, B.Johnson PAT Subcommittee meeting October 23, 2002.
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,
ISO 9001 : 2000.
VALIDATION What is the new guidance?. What is a Compliance Policy Guide? Explain FDA policy on regulatory issues CGMP regulations and application commitments.
Integrating CMC Review & Inspection Industry Recommendations Joe Anisko April 24, 2003.
What’s New in the FDA’s Pharmaceutical Inspectorate and Risk Based Systems Inspection Rick Perlman Chair Food, Drug, and Cosmetic Division ASQ.
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.
Manufacturing Subcommittee of the Advisory Committee for Pharmaceutical Science July 20-21, 2004 Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical.
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 Revisions to 21 CFR Supplements and Other Changes to an Approved Application PhRMA Perspective FDA Public Meeting – 7 Feb 2007.
Radiological Devices Advisory Committee Meeting November 18, 2009 John A. DeLucia iCAD, Inc.
FDA’s Perspective Continued - Where We Are ?. GMP Task Groups.
Ensuring Physical Stability of Pharmaceuticals: Can/should we improve our ability to identify and prevent physical changes? Ajaz S. Hussain, Ph.D. Deputy.
Growth and Success through Partnering & Outsourcing.
Regulatory Update Ellen Leinfuss SVP, Life Sciences.
Achieving and Demonstrating “Quality-by-Design” with Respect to Drug Release/dissolution Performance for Conventional or Immediate Release Solid Oral Dosage.
Risk-Based CMC Review Paradigm
Organizational Gaps in Reaching the “Desired State” Helen Winkle.
One Quality Place Edison, NJ Visit: The Interrelationship.
Nonclinical Studies Subcommittee Advisory Committee for Pharmaceutical Science CMC Issues for Screening INDs Eric B. Sheinin, Ph.D. Acting Deputy Director.
Industry Perspective on Challenges for Product Developers - Drugs Christine Allison, M.S., RAC Associate Regulatory Consultant, Global Regulatory Affairs.
NCI Review of the Clinical Trials Process 6 th Annual National Forum on Biomedical Imaging in Oncology James H. Doroshow M.D. April 7, 2005 Bethesda, Maryland.
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.
1 Supplements and Other Changes to an Approved Application By: Richard J. Stec Jr., Ph.D. February 7, 2007.
Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical Science, CDER, FDA ACPS Subcommittee on Manufacturing Science: Identification and Prioritization.
Ajaz S. Hussain, Ph.D. Office of Pharmaceutical Sciences CDER, FDA October 21, 2003 Welcome and Introduction to the Meeting.
FDA Regulation of Drug Quality: New Challenges Janet Woodcock, M.D. Director, Center for Drug Evaluation and Research, Food and Drug Administration April.
FDA Recommendations: Sampling Plans for Blood Establishments Lore Fields MT(ASCP)SBB Consumer Safety Officer OBRR/CBER/FDA October 19, 2012.
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.
Important informations
Office of Pharmaceutical Sciences
FDA’s Advisory Committee for Pharmaceutical Science The Subcommittee on Process Analytical Technologies (PAT): Opening Remarks Ajaz S. Hussain, Ph.D.
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 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,
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.
COMPARABILITY PROTOCOLUPDATE ADVISORY COMMITTEE FOR PHARMACEUTICAL SCIENCE Manufacturing Subcommittee July 20-21, 2004 Stephen Moore, Ph.D. Chemistry Team.
10:00 A.M. – Noon 7 June 2004 ICH Quality Plenary Meeting.
Blend Uniformity: Update Ajaz S. Hussain, Ph.D.. Background Issue: Assuring and documenting “adequacy of mixing” operations –PQRI’s Proposal Stratified.
Progress in FDA’s Drug Product Quality Initiative Janet Woodcock, M.D. November 13, 2003.
General Aspects of Quality assessment of multisource interchangeable medicines Rutendo Kuwana Technical Officer, WHO, Geneva Training workshop: Assessment.
Process Understanding and PAT
FDA Regulation of Drug Quality: New Challenges Janet Woodcock, M.D. Director, Center for Drug Evaluation and Research, Food and Drug Administration November.
Research in the Office of Vaccines Research and Review: Vision and Overview Jesse Goodman, M.D., M.P.H. Director, Center for Biologics Evaluation and Research.
Pharmaceutical Manufacturing Subcommittee of the ACPS Ajaz S. Hussain, Ph.D. ACPS Meeting October 22, 2002.
1 PAT Subcommittee Closing Report 12 March 2003 Tom Layloff Acting Chair.
Process Analytical Technologies (PAT) Sub-Committee Report ACPS Meeting October 21, 2002 Tom Layloff, Ph.D.
Process Analytical Technologies (PATs), Applications and Benefits Working Group.
ICH Quality Topics Update
CDER / Office of Compliance ACPS October 5, 2006 Joseph C. Famulare Acting Deputy Director Office of Compliance CDER / FDA.
PAT Progress Report: 13 April 2004 ACPS Meeting Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical Science CDER, FDA.
Comparability Protocols Nancy Sager Associate Director, QIS-Chemistry FDA/CDER/OPS.
Second Meeting of the FDA/ACPS Process Analytical Technology: Closing Remarks Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical Sciences.
FDA’s Advisory Committee for Pharmaceutical Science The Subcommittee on Process Analytical Technologies (PAT): Closing Remarks Ajaz S. Hussain, Ph.D. Deputy.
FDA PAT Sub-Committee of Advisory Committee for Pharmaceutical Sciences June 12-13, 2002; Gaithersburg, MD Regulatory Challenges: Post-Approval PAT Applications.
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.
Product & Process Working Group February 26, 2002.
FDA’s IDE Decisions and Communications
The Interrelationship between PAT and Six Sigma
Quality System.
The Interrelationship between PAT and Six Sigma
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:

FDA’s Advisory Committee for Pharmaceutical Science The Subcommittee on Process Analytical Technologies (PAT): Overview and Objectives Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical Science, CDER, FDA February 25, 2002, Gaithersburg, MD.

Outline Overview of the FDA’s PAT Initiative –What? –Why? –When? –How? Goals and Objectives of the PAT- Subcommittee and Working Groups –What does FDA need/expect from you?

What are PAT? Systems for continuous analysis and control of manufacturing processes based on real-time measurements, or rapid measurements during processing, of quality and performance attributes of raw and in-process materials and processes to assure acceptable end product quality at the completion of the process. –Process analytical chemistry tools + information management tools + feedback process control strategies + product & process design and optimization strategies

PAT for Pharmaceuticals: Why? Optimal applications of PAT can improve the capability and the efficiency of pharmaceutical processes while maintaining or improving product quality –improve process understanding and help to ensure quality was “built in” or “by design” –reduce the risk of scrap and recalls –reduce production cycle times and enhance capacity utilization –in the long run, reduce product development time

PAT for Pharmaceuticals: Why? Current level of product quality is generally adequate for the intended use The process by which we achieve this level of quality is inefficient –The current manufacturing paradigm is skewed towards testing to document product quality and rejecting (or recalling) products of unacceptable quality

PAT for Pharmaceuticals: Why? Ensuring high efficiency of the US pharmaceutical manufacturing sector –Provide high quality drugs to the US public in a timely manner by taking advantage of the many new drug development opportunities offered by advances in biology and chemistry –Ensure optimal utilization of public and private resources to meet the growing health care needs of the US public Minimize risks due to sub-optimal pharmaceutical process quality

PAT for Pharmaceuticals: Why? Low manufacturing efficiency, waste (time and resources) and a high “cost of compliance” Need for very high level of regulatory scrutiny (review and inspections) –High proportion of FDA resources needed to ensure adequate product quality –Recurring problems that do not seem to get resolved –Continued debates between FDA-industry, few permanent resolutions

Risks Due to Sub-optimal Pharmaceutical Process Quality Development (Quality specifications) Manufacturing (Process capability)Regulatory approval and compliance

Minimum Regulatory “Sigma” Level for Drugs? Under cGMP when failures/recalls exceeds 10% - no longer “validated.” The minimum regulatory "Sigma” ~ 1.65? FDA Science Board 11/16/01: PricewaterhouseCooper Presentation (Modified by AH) Pharmaceuticals Semiconductor

Pharmaceutical OOS & Batch “Failures” Rates Scrap and rework - we plan for 5-10% (accepted as necessary) PWHC 11/16/01 “It is authors’ experience that... validation exercise precedes a trouble-free time period in the manufacturing area only to be followed by many hours (possibly days or weeks) of troubleshooting and experimental work after a batch or two of product fails to meet specifications. This becomes a never-ending task.” Harwood and Molnar. Using DOE techniques to avoid process problems. Pharm. Dev. Tech

Risks Due to Sub-optimal Pharmaceutical Process Quality Risk of releasing a “poor” quality product –Recalls are not effective quality control tools Drug shortages –Delay in approval of important drug products –High potential for disruption in the availability of important drugs –Production of low volume “essential” drugs may be adversely effected

Risks Due to Sub-optimal Pharmaceutical Process Quality Regulatory commitments on an inefficient manufacturing process –Continued optimization activities in the post approval phase (or live with the “validated” but inefficient process) –Recurring manufacturing difficulties leading to very low efficiency and capacity utilization –Higher manufacturing and regulatory compliance costs “locked-in”

Risks Due to Sub-optimal Pharmaceutical Process Quality Increased risk of non-approval or delayed regulatory approval –Increased potential for quality problems confounding the clinical safety and efficacy databases –Past quality (compliance) problems can delay new drug approvals –Industry and FDA resources being spent on recurring problems

When? When was this initiative started? 3rd quarter 1999 (building on the AOAC International Special Symposium: “ Pharmaceutical Process Control and Quality Assessment by Non-Traditional Means,” October 1993, St. Louis, Missouri) FIP’s Millennium Congress, New Technology Forum of the Royal Pharmaceutical Society, PhRMA Technical Conclave,... –19 July 2001, ACPS Meeting –16 November 2001, FDA Science Board Meeting –28 November 2001, ACPS Meeting Recommendation to form a PAT Subcommittee

When? When can companies submit PAT based applications or submissions to FDA? –Any time a company is ready to do so they should contact the OPS/CDER/FDA to discuss their proposed PAT applications or submissions There are many hurdles that seem to hold back PAT applications –It is widely perceived that FDA will not accept PAT based applications, this is not true

Need for FDA to Facilitate Introduction of PAT Industry is hesitant to introduce PAT in US –Regulatory uncertainty/risk leads to “Don’t Tell” or “Don’t Use” practice New Technology = New Questions –Method suitability, chemometrics and validation Old products + New technology = New Regulatory Concerns –Problems not visible under the current system –Mindset: Why change? PAT application will add to current regulatory requirements

How does FDA plan to facilitate introduction of PAT? Eliminate regulatory uncertainty –#1. FDA will accept PAT applications that are based on “good” science Develop standards for PAT –Method suitability and validation –Multivariate statistical/computer pattern recognition –Critical process control points and specifications –Changes, OOS…. –#2. Current system “adequate for intended use” –#3. Introduction of PAT not a requirement

How does FDA plan to facilitate introduction of PAT? Eliminate regulatory uncertainty –#4. Define conditions under which PAT may replace current “end product release testing” –#5. Process for addressing existing “invisible” problems in marketed products –#6. Review and inspection practices –#7. International harmonization

How does FDA plan to facilitate introduction of PAT?: Two Tracks General Guidance on PAT –Information source: ACPS Subcommittee on PAT and working groups Meeting #1 2/25-26/02 Meeting #2 (6/02?) –Draft Guidance Implementation –CDER-ORA Team Invite companies to propose submissions –Expect to receive proposals for submissions (~3 by 4q 02) –Review-Inspection plans and teams for these submissions Plan for concurrent development -review- inspection

General (principles) Guidance on PAT Proposed Goals and Objectives –General principles and terminology Bring the community on the “same page” –Address issues related to “regulatory uncertainties” – Clarify the regulatory process Review and inspection –Other tangible benefits Serve as a tool for building within-company consensus Promote research and development activities in the pharmaceutical PAT area

Guidance Development Process PAT Steering Committee –CDER (OPS/OC) and ORA Douglas Ellsworth, Mike Olson/Diane Obrien, Joe Famulare, Frank Holcomb, Moheb Nasr, Yuan Yuan Chiu, Ajaz Hussain (Chair) Guidance writer: Raj Uppoor Project management: Chris Cole Communication tools - Web based and electronic tools

Options for Introducing PAT A. Currently marketed “robust” products. PAT to improve efficiency (minimal improvement in quality assurance) B. Currently marketed products that need improvement. Step wise PAT approach - first improve quality and then improve the efficiency C. New products. PAT utilized throughout development and scale-up. Lab based tests to ensure shelf-life and/or for establishing “public standards.”

PAT Subcommittee A major source of information for the FDA’s General Guidance on PAT –At the end of this meeting: Topics to be covered in the guidance (outline) Layout general principles for setting specifications, validation, chemometrics Consensus on benefits, definitions, terminology –Meeting #2? More details on “optimal” applications, identification and control of critical formulation and process variables, specifications, validation, chemometrics, addressing OOS, ……. Illustrative examples (for inclusion in the guidance)

PAT Subcommittee Organization –Industry presentations to focus the discussion –Questions (see background packet) to stimulate and focus discussion –Four working groups Benefits, technology, definitions/terminology Process + Analytical Validation Chemometrics Product/Process Development –Only two meetings planned

Chemometrics (Kowalski and Wold) Multivariate data collection and analysis –DOE, PCA, PLS, non-linear PLS, neural networks,…... –Calibration, process modeling, pattern recognition and classification, signal correction and compression –Statistical process control,…. Need information of the type of tools and general principles (and examples) for “verification” and “validation” of such analyses

Challenges Different perspectives, expertise, and affiliations - can we come on the “same page” by the end of this meeting? Are two meetings sufficient to gather the information necessary to develop the general guidance? Is the “general guidance” approach the most effective approach?