Product & Process Working Group February 26, 2002.

Slides:



Advertisements
Similar presentations
Basic Principles of GMP
Advertisements

Tips and Resources IASC Cluster/Sector Leadership Training
Technology Module: Technology Readiness Levels (TRLs) Space Systems Engineering, version 1.0 SOURCE INFORMATION: The material contained in this lecture.
Statistical Evaluation of Dissolution for Specification Setting and Stability Studies Fasheng Li Associate Director, Pharmaceutical Statistics Worldwide.
1 COMM 301: Empirical Research in Communication Kwan M Lee Lect4_1.
Statistical Approaches to Addressing the Requirements of the New FDA Process Validation Guidance for Small Molecules 1 Jason Marlin, MS/T Statistics, Eli.
TITLE OF PROJECT PROPOSAL NUMBER Principal Investigator PI’s Organization ESTCP Selection Meeting DATE.
Strategy 2022: A Holistic View Tony Hayes International President ISACA © 2012, ISACA. All rights reserved.
VALIDATION What is the new guidance?. What is a Compliance Policy Guide? Explain FDA policy on regulatory issues CGMP regulations and application commitments.
ASSESSORS ORIENTATION 2008 DEPARTMENTAL SERVICE EXCELLENCE AWARDS.
Supplier SQM Participation. 2 | MDT Confidential What is SQM? Stands for Supplier Quality Managment –Formally referred to as SPACE and SPICE Is a system.
Batch Reworking and Reprocessing
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,
Determine impurity level in relevant batches1
Fundamentals of Information Systems, Second Edition
Process Analytical Technologies Subcommittee Product and Process Development: An Industry Perspective David Rudd PhD Process Technology GlaxoSmithKline.
Pharmaceutical Product Quality Assurance Through CMC Drug Development Process Presented by Darlene Rosario (Aradigm) 21 October 2003 Meeting of the Advisory.
DEFINING JOB PERFORMANCE AND ITS RELATIONSHIP TO ASSESSMENTS.
Developing the Marketing Plan
Training Workshop on Pharmaceutical Development with a Focus on Paediatric Medicines / October |1 | Regulatory Requirement on Dossier of Medicinal.
What are MRLs ? Alfred W. Clark Dawnbreaker, Inc.
PAT Validation Working Group Process and Analytical Validation Working Group Arthur H. Kibbe, Ph.D. Chair June 13, 2002.
FDA’s Perspective Continued - Where We Are ?. GMP Task Groups.
Product Lifecycle Management Cost of Quality Pasi Kaipainen, Mika Huhta.
Huzairy Hassan School of Bioprocess Engineering UniMAP.
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,
Guidelines for Carbon Fiber Tape Prepreg Specifictions W.T.McCarvill S.Ward August 6, 2002.
Annex I: Methods & Tools prepared by some members of the ICH Q9 EWG for example only; not an official policy/guidance July 2006, slide 1 ICH Q9 QUALITY.
Quality By Design - A Generic Industry Perspective
Financial Assessment and
Achieving and Demonstrating “Quality-by-Design” with Respect to Drug Release/dissolution Performance for Conventional or Immediate Release Solid Oral Dosage.
Basic Principles of GMP
N By: Md Rezaul Huda Reza n
1 Supplements and Other Changes to an Approved Application By: Richard J. Stec Jr., Ph.D. February 7, 2007.
Changes without Prior Approval Breakout Session Summary Rick Smith Aventis Pasteur, Inc.
Capability Maturity Models Software Engineering Institute (supported by DoD) The problems of software development are mainly caused by poor process 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.
Quality by Design (QbD) Myth : An expensive development tool ! Fact : A tool that makes product development and commercial scale manufacturing simple !
Important informations
Risk Management in premarketing phase Anshu Vashishtha MD PhD (in individual capacity employer : Watson Pharmaceuticals)
Overview of Portfolio Process Maturity
CHAPTER 28 Translation of Evidence into Nursing Practice: Evidence, Clinical practice guidelines and Automated Implementation Tools.
PhRMA Perspective on FDA Final Report FDA Advisory Committee on Pharmaceutical Sciences October 20, 2004 G.P. Migliaccio, Pfizer Inc.
Molecule-to-Market-Place Quality
COMPARABILITY PROTOCOLUPDATE ADVISORY COMMITTEE FOR PHARMACEUTICAL SCIENCE Manufacturing Subcommittee July 20-21, 2004 Stephen Moore, Ph.D. Chemistry Team.
© G. A. Motter, 2006, 2008 & 2009 Illustrated by Examples Quality Function Deployment and Selection Matrices Customer Driven Product Development.
10:00 A.M. – Noon 7 June 2004 ICH Quality Plenary Meeting.
General Aspects of Quality assessment of multisource interchangeable medicines Rutendo Kuwana Technical Officer, WHO, Geneva Training workshop: Assessment.
Risk Sharing Schemes Dr Rafiq Hasan Director of Market Access
Process Analytical Technologies (PATs), Applications and Benefits Working Group.
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.
Second Meeting of the FDA/ACPS Process Analytical Technology: Closing Remarks Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical Sciences.
Pharmaceutical Quality Control & current Good Manufacturing Practice
Continual Service Improvement Methods & Techniques.
Engineer Exceptionally Capable Project Teams February 2012.
Department of Defense Voluntary Protection Programs Center of Excellence Development, Validation, Implementation and Enhancement for a Voluntary Protection.
European Diagnostic Manufacturers Association EQA contribution and industry expectations Dr. Claude GIROUD – Chairman Standardization, Quality & Risk Management.
Info-Tech Research Group1 Info-Tech Research Group, Inc. Is a global leader in providing IT research and advice. Info-Tech’s products and services combine.
Principal Investigator ESTCP Selection Meeting
Product Lifecycle Management
Quality Metrics for better Quality Compliance
Principal Investigator ESTCP Selection Meeting
Quality System.
Software Quality Engineering
Assessing your total rewards offer
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.
Principal Investigator ESTCP Selection Meeting
Principal Investigator ESTCP Selection Meeting
Presentation transcript:

Product & Process Working Group February 26, 2002

Questions to the Working Group What considerations, during product development, are needed to ensure optimal application of PATs to realize "quality by design" through better understanding of processes and determination of performance-based process controls/end points? Benefits of PAT are under realized Companies have not always seen the benefits they expected

Question 1 continued Still some selling that needs to be done 6 sigma is too high, 3-4 sigma may be more realistic

Questions to the Working Group What areas, from raw material identification/characterization to finished product manufacturing, are amenable to monitoring and control using process analytical technologies? Applicable to most areas of the manufacturing process. Different levels of maturity. Applicable to most areas of the manufacturing process, but different levels of maturity. Nature of the ingredient is a factor. May not work in all cases.

Question 2 continued PAT Technology allows the incorporation of feed back controls such that a whole batch need not be lost Goal during development is to understand and develop robust processes During development look at a lot of parameters to identify what will be important monitor in actual practice. Evaluation of other technologies from other industries may be helpful

Question 2 continued Unit operations (where we have the history) and possible technologies that may be used, should not exclude other promising technologies, we don’t want to limit alternative approaches

Questions to the Working Group How do you anticipate PAT application will change the process for identifying critical process variables, their control, and establishment of product specifications? Development function. Structured approach. Getting into the process early. Process of optimization allows you to identify critical parameters and develop metrics. How you control it is up to you. Online sensors give you additional information to identify your critical endpoints.

Question 3 continued Multivariate approach/strategy Identifying new variables that may be more pertinent to the process Correlate PAT and specifications where relevant Look at raw material, quality is a key factor

Questions to the Working Group What are some of the issues that arise during the scale up of pharmaceutical manufacturing, using PATs? Do PATs help in the scale-up situations? Yes. What is the endpoint that you’re working towards? Need to know what the process looks like when it’s working well. Insight when the process is working well and when it’s not. Getting an endpoint to work towards. You’ll know where problems may exist and what to monitor. You can often calibrate PAT to scale up and develop scale up coefficients

Question 4 continued Do they cause problems? Yes. Limitations in existing “gold standard” method Engineering issue - Ruggedness of engineering. Critical implementation issue. Applying design to equipment Business issue: Addition of PAT must be value added For new products sensor applications for up-front equipment are easier to put in place and employ PAT measurement may not match your submission parameters but your finished product is still good. The mechanism in which these changes are submitted needs to be clear.

Question 4 continued Moving from a parameter control to an endpoint control. Have to set boundaries (upper limits, work within this window) Low dose drugs, low potency, may be many more exceptions Do they make scale-up transitions easier, and if so, why? Yes. Better understanding of your process always helps

Questions to the Working Group In some situations, PATs may be used only for certain specific unit operations within the overall scheme of a dosage form manufacturing. What are some of the advantages and disadvantages for doing so? No technical downside. It’s a business decision. Has to be “value added” Accurately reflecting what’s going on, it can’t be a disadvantage. Overall weakness, e.g. blend homogeneity does not mean you will not have other problems down stream

Question 5 continued “Go for new technologies, you pay for new technologies” Business decision, development time lines. Speed to market, Cost in time. Allocation of resources (human) Advantage – single implementation, supports doing it incrementally for other similar processes, e.g. dryer

Questions to the Working Group How can PATs be used to minimize, or prevent “out of specification” incidents? If you’re are allowed to go to an endpoint, it will help decrease OOS incidents It will decrease incidents proactively because the developed process is more rugged. Can drive more scientific investigation of problems. Gives you more data to trouble shoot problems.

Questions to the Working Group How can PAT tools be used for predicting the performance of a drug product (e.g. dissolution) based on causal links and data based correlation? Certainly possible. Need to develop correlation's Exercise in benefit/risk assessment More work needs to be done, e.g. use of acoustic data. There are measurement technologies that could be used, but they are not yet mature.

Question 7 continued Case by case basis.

Questions to the Working Group Can PAT tools be used for predicting the stability of a drug product? If yes, what are the factors that should be considered prior to using them? No. Can’t replace stability study. PAT may be used as a predictor. Yes for physical instabilities. Can be possible, but in the general sense it’s pretty limited May reduce risk, but you’ll still need to test. May be able to predict, but will still need data to confirm stability

Question 8 continued One benefit, batch release will be higher quality, product failing during shelf life will be less likely More consistent product

Questions to the Working Group What factors should the Industry and the Agency consider while implementing the use of new PATs for already approved drug products? Benefits – product capability. Building the quality in. You can’t do that retrospectively. Continuous monitoring of an on-going process may yield higher quality product Considerably more prospects with new products

Question 9 continued It could have applications to validation and SUPAC guidelines in the future

Question 9 continued View from industry “If it’s not broken, don’t fix it” When vendor changes, it’s an opportunity to use PAT. Honeymoon period to collect parallel data Team inspections are being planned – review chemist and inspector will work together. Decisions based on sound science. Manufacturers may look at implementing PAT when other changes are made, e.g. site change

Draft Guidance Table of Contents comments  combine III & VI  V. What types of data do we need?  IV. Facilitation of PATs. Agency is open to parallel testing. Should be expanded.  Add reason for doing this in I.Introduction  Use of PATs in product development  Enabling technology (including chemometrics)  Relationship to finished product specification  Worked examples for different product types, e.g. three different dosage forms

Draft Guidance Table of Contents comments Section VII change to review of PAT information in an application  Guidance should address roles and responsibilities of different groups, e.g. quality unit, engineering, process technology as well as the scale mixes