Quality and Compliance Challenges for Biopharmaceutical Products Ramon Rivera Gonzalez, Ph.D. Director Quality Assurance Amgen Manufacturing, Limited.

Slides:



Advertisements
Similar presentations
Basic Principles of GMP
Advertisements

Good Manufacturing Practice Regulations
Biopharmaceutical Quality
Radiopharmaceutical Production
1 ASQ Keynote February 13, 2006 A Look Back at 2006 The year of the hurricane Organizational Change National and District Changes in programs Adverse Drug.
Annual Product Review (APR) Product Quality Review (PQR)
CSI Biopharmaceutical Manufacturing: How Manufacturing Failures are Investigated Katie Stewart – Technical Training Manager.
Regulatory Challenges in the Cell Preparation Facility Adrian Gee Center for Cell & Gene Therapy Regulatory Challenges in the Cell Preparation Facility.
VALIDATION What is the new guidance?. What is a Compliance Policy Guide? Explain FDA policy on regulatory issues CGMP regulations and application commitments.
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
Quality control of raw materials In-process control
MANUFACTURING OF API AND INTERMEDIATES
FIELD COMPLIANCE UPDATE CDR Thomas R. Berry, RPh FDA, Investigator RAL-RP / ATL-DO.
INTERNAL AUDITS OF LABORATORIES Sanjay S Shetgar 1.
Pilot Risk-Ranking Model to Prioritize Manufacturing Sites for GMP Inspections Advisory Committee for Pharmaceutical Science Manufacturing Subcommittee.
External Defibrillators: Recalls, Inspections, and the Quality System Regulation Melissa Torres Office of Compliance December 15, 2010.
ISO 9000 Certification ISO 9001 and ISO
QC/QA Mary Malarkey Director, Division of Case Management Office of Compliance and Biologics Quality Center for Biologics Evaluation and Research March.
Good Manufacturing Practices for Blood Establishments
Personnel Basic Principles of GMP Workshop on
4. Quality Management System (QMS)
Top Tactics for Maximizing GMP Compliance in Blue Mountain RAM Jake Jacanin, Regional Sales Manager September 18, 2013.
4. Quality Management System (QMS)
Huzairy Hassan School of Bioprocess Engineering UniMAP.
Radiopharmaceutical Production
© 2009 Michigan State University licensed under CC-BY-SA, original at Corrective Action.
World Health Organization
Regulatory Update Ellen Leinfuss SVP, Life Sciences.
Project co-financed by European Union Project co- financed by Asean European Committee for Standardization Implementing Agency 1 Module 13 GMP Workshop.
Inspection Issues in the Analytical Laboratory: An FDA Perspective Yvonne McKnight Chemist US Food and Drug AdministrationPhone: x
Module 5: Assuring the Quality of HIV Rapid Testing
Good Manufacturing Practice. Good Manufacturing Practice Regulations Establishes minimum GMP for methods to be used, and the facilities or controls to.
Important informations
Aloe Vera of America: A Quality IASC-Certified Producer of Aloe Vera Presented by Walt Jones.
Proposed Rule for Preventive Controls for Animal Food.
Proposed Rule: 21 CFR 507 Proposed Rule for Preventive Controls for Animal Food 1.
Overview of FDA's Regulatory Framework for PET Drugs
Molecule-to-Market-Place Quality
30/02/2008 Dept. of Pharmaceutics 1 Salient Features of Quality Assurance Dr. Basavaraj K. Nanjwade M.Pharm., Ph.D Associate Professor Department of Pharmaceutics.
COMPARABILITY PROTOCOLUPDATE ADVISORY COMMITTEE FOR PHARMACEUTICAL SCIENCE Manufacturing Subcommittee July 20-21, 2004 Stephen Moore, Ph.D. Chemistry Team.
Expectations for Facilities & cGMPs Biological Response Modifiers Advisory Committee Meeting October 9, 2003 Nicholas Obiri, Ph.D. CBER.
Draft Guidance for Industry: CGMPs for Phase 1 INDs Joseph C. Famulare, Director Division of Manufacturing & Product Quality Office of Compliance, CDER,
Final Rule for Preventive Controls for Animal Food 1 THE FUTURE IS NOW.
General Aspects of Quality assessment of multisource interchangeable medicines Rutendo Kuwana Technical Officer, WHO, Geneva Training workshop: Assessment.
CBER Common Problems on Source Plasma Inspections Judy Ellen Ciaraldi BS, MT(ASCP)SBB, CQA(ASQ) CBER, OBRR, DBA September 16, 2009.
The Second Annual Medical Device Regulatory, Reimbursement and Compliance Congress Presented by J. Glenn George Thursday, March 29, 2007 Day II – Track.
Module 2Slide 1 of 26 WHO - EDM Quality Management Basic Principles of GMP Part One.
CDER / Office of Compliance ACPS October 5, 2006 Joseph C. Famulare Acting Deputy Director Office of Compliance CDER / FDA.
Pharmaceutical Quality Control & current Good Manufacturing Practice
Topic #2: Quality by Design and Pharmaceutical Equivalence Ajaz S. Hussain, Ph.D. Office of Pharmaceutical Science Center for Drug Evaluation and Research.
Good Manufacturing Practice (GMP) Compliance: GMPs EXPLAINED.
GMP AND cGMP CONSIDERATIONS
WORKSHOP ON ACCREDITATION OF BODIES CERTIFYING MEDICAL DEVICES INT MARKET TOPIC 9 CH 8 ISO MEASUREMENT, ANALYSIS AND IMPROVEMENT INTERNAL AUDITS.
Quality Control significance in pharmaceutical industry
In the name of God. Common Technical Document On Biotech.
The Regulation on Cell Therapy Products in Japan
GMP -Regulatory Perspective Sukanti Borkar Abbott India Ltd.
GOOD MANUFACTURING PRACTICE FOR BIOPROCESS ENGINEERING (ERT 425)
Quality Control and Assurance
Pharmaceutical Quality Control & current Good Manufacturing Practice
MHRA GMP Inspection Deficiency Data Trend 2016
Quality Assurance and Quality Control in Generics
UK Legal Requirement for Notification of Serious Breaches of Good Clinical Practice or The Trial Protocol John Poland, PhD Senior Director, Regulatory.
Quality System.
Introduction to GMP.
World Health Organization
Radiopharmaceutical Production
Manufacture of Sterile Medicinal Products: Practical Examples
Manufacture of Sterile Medicinal Products: ‘Annex 1’ - DRAFT
Presentation transcript:

Quality and Compliance Challenges for Biopharmaceutical Products Ramon Rivera Gonzalez, Ph.D. Director Quality Assurance Amgen Manufacturing, Limited

2 Outline  Background Information  Regulatory and Process Challenges  Quality by Design  Risk Management  Process Analytical Technology  Key Quality Systems Deviations/Nonconformities Corrective and Preventive Actions  Biological Product Deviations  FDA and EMEA Observations

3 Biotechnology Biotechnology is a set of scientific techniques used to derive valuable products from living organisms Applications  biopharmaceutical drugs  agriculture  waste management

4 Background information  FDA- “sterile drugs should be manufactured by aseptic processing only when terminal sterilization is not feasible”.  Parenterals -Drug administration other than by the mouth or rectum- ex. Injection, infusion or implantation. –Biological products (vials or syringes) solubility, stability, maintain activity

5 Biotechnology BIOLOGY CHEMISTRYENGINEERING

6 Biological Products Manufacturing

7

8 Bacterial Systems (+) –Grow fast –Easy to maintain –High yield (-) –Endotoxins –Low expression or extracellular secretion –No post-translational modifications machinery E. coli

9 Mammalian Cells (+) –Adequate conformation –Post-translational modifications –Preferred for complex proteins (-) –Grow slower than bacteria –Maintenance is expensive –Usually lower yield –Limited manufacturing applications CHO Mammalian

10 Quality and the Product Life Cycle  Adherence to regulations  Control and maintenance of documentation  Quality of suppliers, components and raw materials  Reliability and consistency  Monitor/audit of the manufacturing process  Deviations, unexpected situations - product impact  Correction and prevention - CAPA  Lot release/rejection decision  Customer complaints  Continuous improvement

11 Quality Systems Emphasis  Quality Management  Quality Assurance  Risk Management –Evaluation analysis and quality risk management tools  Preventive Action  Promote product and process improvement (i.e., continuous improvement)  “Continuous Improvement” of the Quality System

12 Regulatory Environment Challenges  Transfer of CBER products to CDER  GMPs for the 21 st. Century  Aseptic Processing Guideline  Risk management  Increased scrutiny of product insert claims  Focus on patient safety  Process Analytical Technology (PAT)

13 Regulatory Environment Challenges  Quality by Design  Bioterrorism  Animal-derived materials  Country-specific regulatory requirements –Mexico, Brazil, Saudi Arabia, Japan  Biogenerics in EU  State of the art technology –Isolators

14 Major Process Challenges Sterile vs. Aseptic Requires the application of microbiological contamination control to prevent infectious organisms to be present in the sterile product Demonstrate “CONTROL” of the process, while technical complexity increases Characterization to identify variability components Application of science and new technologies Maintenance of the cell lines Contamination risks Personnel as “incubators” Source of microbial load

15  “Quality can not be tested into products; it has to be built in by design”  Product quality and performance requires efficient design of manufacturing processes  Product specifications based on deep understanding of how formulation and process factors impact product performance  It provides a framework for continuous "real time" assurance of quality and continuous Improvement Quality by Design

16 The importance of Design  Multidimensional combination and interaction of input variables and process parameters that have been demonstrated to provide an assurance of quality  Operating within design parameters will produce a product meeting designed quality attributes  Working within the design parameters is not considered a reportable change  Movement outside of design space is considered a change – subject to regulatory approval

17 Risk Management  What are the potential hazards to process and product ? Identify potential hazards both prospectively and in a reactive mode  Applies to components, container closure, raw materials, dosing devices, manufacturing process, drug substance, intermediates  How these factors influence variability of process, product performance, product safety and efficacy?

18 Risk Management  Risk management is a regulatory expectation for a modern quality system  Risk assessment, risk control, risk communication and risk review throughout product lifecycle  Decisions should be based upon process and product understanding Balance between the use of risk management and compliance with GMPs  Always include intended use, patient safety and availability

19 Process Analytical Technology  “…any system for continuous analysis and/or control of manufacturing processes based on real-time measurements, or rapid measurements during processing.” Source; FDA Human Drug cGMP Notes, Q1 2002

20 Process Analytical Technology  “At line” - the sample is removed, isolated from, and analyzed in close proximity to the process stream  “On-line” - the sample is diverted from the manufacturing process, and may be returned to the process stream  “In-line” – the sample is not removed from the process stream and can be invasive or non-invasive Source: FDA PAT Guidance

21 PAT - Benefits  Enhancement of process understanding  Sources of variability identified and explained  Meet requirements for validating and controlling process  Quality attributes can be accurately and reliably predicated  Continuous improvement  Integration of development, manufacturing, QA and knowledge management  Acceptability of in-process materials and final product based on process data

22 Management Controls  Written quality policy and objectives  Management reviews – regulations and quality objectives – attendance documented – results, action plans/corrective actions documented  Internal audits –auditors no direct responsibility for matters being audited

23 Deviations/Nonconformities  Classification based on impact/risk  Investigations need to be thorough, stand-alone –Root cause analysis –Product impact Stability data, intrinsic vs. extrinsic, historical data –Toxicological/health hazard evaluations based on route of administration to patient –Corrective and Preventive actions

24 Corrective and Preventive Actions (CAPA)  Corrective = correct existing nonconformity  Prevention = potential recurrence of nonconformity Regulatory expectations: –Identify sources of problems/nonconformities Unfavorable trends –Prioritize based on risk –Defined action plans –Timely implementation –Measure and document effectiveness –Reviewed by Management

25 If a deviation/nonconformity involves distributed product…  Evaluate per 21CFR – Biological Product Deviation (BPD)  “must report any event and any information relevant to the event associated with the manufacturing, to include testing, processing, packing, labeling or storage, or with the holding or distribution, of a licensed biological product if the event meets the following criteria: (1)Either; (i)Represents a deviation from cGMP, applicable regulations, applicable standards, or established specifications that may affect the safety, purity or potency of that product; or (ii)Represents an unexpected or unforeseeable event that may affect the safety, purity, or potency of that product; and (2)Occurs in your facility or a facility under contract with you; and (3)Involves a distributed biological product

26 Some points regarding BPDs  Distributed = biological product has left the control of the licensed manufacturer Contract manufacturing  The decision to report should be based on whether the event had the potential to affect safety, purity, or potency of the product  The license holder is required to file at a date not to exceed 45 calendar days from the date that you, your agent, or another person, acquire information reasonably suggesting that a reportable event has occurred  Must report even if the investigation determined that there was no impact

27 BPD- Reportable Examples  Raw materials that failed specifications used in manufacturing  Aseptic processing not performed according to procedures  Stability testing not performed at required interval  Missing information in label (product type, lot number, storage temperature, concentration)  Distributed product prior to completion of required testing  Product release prior to validation of manufacturing process  Biological Drug Substance stored at the incorrect temperature

28 BPD – Non-reportable Examples  Raw material did not meet specification and was rejected prior to its use  Testing performed incorrectly, invalidated and repeated and found acceptable prior to distribution  Product labeled with a shortened expiration date (not due to failure to meet specification)  Product shipped to the incorrect facility  Customer order filled incorrectly (wrong product, wrong amount), but labeled correctly

29 EMEA Observations  Critical = give rise to product that could be harmful to patient  Major = result in product not meeting marketing authorization, major deviation from EU GMP  Other = usually lack information to be classified

30 EMEA  Critical Design and maintenance of premises Potential for microbiological or chemical/physical contamination  Major/Other Potential for microbial contamination Documentation- Quality system elements/procedures Unauthorized activities requiring regulatory filing Design and maintenance of equipment and premises

31 FDA Observations ( )  Failure Investigations (Nonconformities) –Failure to conduct investigations –Failure to extend investigations to related batches and products –Averaging OOS results with in-specification results to support release –Delays in starting the investigation –Incomplete investigation into root cause –Failure to take immediate corrective action –Failure to address product impact

32 FDA Observations ( )  Record keeping –Batch Production and Control records do not include complete information –“post-it” notes with hand-written original data –Lack of procedures –No revision history for procedures  Validations –Discrepancies against Master Validation Plan –Inadequate cleaning validation –Inadequate process validation –Inadequate validation of analytical methods

33 In Summary  The successful application of quality systems for the manufacturing of biological products requires the comprehensive synergy between: regulations, science/new technologies, process knowledge, Management accountability and efficient risk management

34 Questions?