CHANGE CONTROL MANAGEMENT FOR A STABILITY PROGRAM

Similar documents
INVESTIGATE OOT AND OOS IN STABILITY STUDIES

FDA S GUIDANCE FOR INDUSTRY ANDAS: STABILITY TESTING OF DRUG SUBSTANCES AND PRODUCTS

Quality Issues for Clinical Trial Materials: The Chemistry, Manufacturing and Controls (CMC) Review

CGMP Requirements for Investigational Products

Author's personal copy

Excipients Facing Increased Scrutiny How to Use Secondary Reference Standards to Help Maintain Regulatory Compliance

Control Strategy. Implementation of ICH Q8, Q9, Q10

Experience with Health Canada s Approach for Post-Approval Changes. Kiran Krishnan Vice President US Regulatory Affairs September 2014

PHARMACEUTICAL TESTING

Drug Quality Assurance: Systems at ChemCon Author: Dr. Peter Gockel

CMC Considerations for 505(b)(2) Applications. Monica Cooper, Ph.D. FDA/CDER/OPS/ONDQA AAPS Annual Meeting Washington, D.C.

Phase Appropriate GMPs for IMPs. Presented by: Karen S. Ginsbury For: IFF, October 31, Nov 02, 2017

Pharmaceutical Reference Standards: Overview and Role in Global Harmonization

Process Design Risk Management A Proactive Approach

Impact factor: 3.958/ICV: 4.10 ISSN:

Process Drift: When Do We Detect it? Richard L. Friedman Director, DMPQ CDER/Office of Compliance PQRI Process Drift Workshop December 1, 2010

Future of Question-based Review and Regulatory Submissions

How do you know the drug you are using is safe and effective?

While the recognition

LEGAL REQUIREMENTS FOR STABILITY

Review article Rapports De Pharmacie 2015;1(2):46-53 ISSN:

Commonly Seen Drug Product Related Quality Deficiencies

EUROPEAN INDUSTRIAL PHARMACISTS GROUP. Guidance on CPD for QUALIFIED PERSONS

Best Practices and Application of GMPs for Small Molecule Drugs in Early Development IQ Workshop, Feb 4-5, 2014, Washington, D.C.

Post Approval Change Management Protocols/Comparability Protocols (PACMPs/CPs) Current and Future State

The Application of Pharmaceutical cgmp to Live Bacterial Products. James Harris Business Development Manager

Ancillary Materials for Cell & Tissue Therapies Definitions, US Regulatory Approach, and USP s Risk-Tiered Approach

Subpart B Organization and Personnel, 21 CFR Responsibilities of the quality control unit.

Post-approval Change Management Protocols - Current Status and Next Steps on the Way towards a Global Tool

Perspectives on Method Validation: Importance of Adequate Method Validation

Pharmaceutical Compounding Sterile and Nonsterile Preparations. Jeanne Sun, PharmD

Question-based Review (QbR)

Introduction to CMC Regulatory Affairs

ICH Quality Implementation Working Group POINTS TO CONSIDER

22 January Division of Dockets Management (HFA 305) Food and Drug Administration 5630 Fishers Lane, rm Rockville, MD 20852

Quality Risk Management (QRM) of active pharmaceutical ingredients during transportation by using FMEA tools and methodology

Workshop Summaries. Discussions of the Global Stability Workshop are summarized for the following topics:

How to Avoid Common Deficiencies in INDs and NDAs. Ramesh Raghavachari, Ph.D. Branch Chief, Branch IX ONDQA/OPS/CDER

PROCESS VALIDATION ROCHAPON WACHAROTAYANKUN, PH.D.

DECISION TREE #1: ESTABLISHING ACCEPTANCE CRITERION FOR A SPECIFIED IMPURITY IN A NEW DRUG SUBSTANCE

Impacto de las Nuevas Tendencias Regulatorias en la Producción de Parenterales. Innovacion en Packaging Primario

Inspections, Compliance, Enforcement, and Criminal Investigations

San Jose, California, USA

Advanced Concepts for Change Control of Analytical Procedures with ICH Q12 Coming

9/2/2014. USP Monograph Modernization. Todays topics. USP basic. Todays topics. - USP basic. - USP publications. - USP monograph modernization

ICH Q10/WHO TRS A Regulatory Perspective

ICH Q12 Technical and Regulatory Considerations for Pharmaceutical Product Lifecycle Management

EMPROVE For Raw and Starting Materials & For Filtration Devices and Single Use Systems. Jan Thomsen Warsaw, November 15 th, 2016

Application of Quality by Design (QbD) in product development. James E. Polli September 16, 2015

Regulatory Assessment

Inspection. Implementation of ICH Q8, Q9, Q10

Question-based Review: A New Quality Assessment System for Generic Drugs

Quality of raw materials and manufacturing of advanced therapies. Fouad Atouf, Ph.D. Head, Global Biologics July 12, 2018

US FDAs Perspective on Product Quality and Bioequivalence

Implement an Effective Change Management System throughout GMP and Validation Environments. Eileen Cortes February 23, 2017

Validation/Verification of Test Methods An FDA Perspective. Laure H. Kairawicz, Ph.D. Senior Scientist Expert Witness

4 Key Considerations When Engaging A New GMP Contract Service Provider

Expiration Dating of Unit-Dose Repackaged Solid Oral Dosage Form Drug Products Guidance for Industry

Pharmaceutical Good Manufacturing Practice. Contents are subject to change. For the latest updates visit

Impurities from degradation of Drug Substances

Method Validation Studies How GLP Interacts With Guidance Documents

API Stability Protocols and. Chris Byrne Tasmanian Alkaloids

Current Status and Perspectives on Pharmaceutical Products in Japan

2. Risk-based compliance was also a key component in the FDA's new approach for dealing with electronic records and signatures: 21 CFR Part 11.

ABB Industries PAT Validation

Setting Specifications for Biotech Products

ASMF/DMF Quality Assessment Report (QAR) IGDRP Quality Working Group

BRIEFING. . Over time, 466 may be used less frequently and may be withdrawn.

Derivation and Justification of Safety Thresholds

Regulation of Active Pharmaceutical Ingredients (API)

A Generic Industry Perspective on Establishing Impurity Limits And a Corresponding Control Strategy

NEWSMAGAZINE FEB 13 REGULATORY SCIENCES. AAPS ELEARNING Advanced Computer Modeling Seen as Way to Dissect Drugs Complex Effects

Supplier Oversight PQRI. September Steven Lynn, MS, CMQ/OE Vice President Global Quality Compliance Mylan

ICH Q9 An Industry Perspective: Ensuring Quality to Patients in a Risk-Based Regulatory Environment

Inspection Trends. American Society for Quality Richmond, VA Section March 8, 2016

Post Approval Changes & Product Lifecycle Management

Synopsis: FDA Process Validation Guidance

Industry Perspectives on OINDP Regulatory Challenges in Global Environment

Challenges in Implementing Knowledge Management within ICH Q10

Textvergleich. Verglichene Dokumente MEDIA3917.pdf. ICH_Q10_Step4.pdf

ICH Q8/Q8(R)

October 10, Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, Rm Rockville, MD 20852

Cell Therapy Product Manufacturing Considerations. July 17, 2017 CMC Strategy Forum Mo Heidaran, Ph.D.

QBD BIOLOGICS: REGULATORY PERSPECTIVE. 8Jun2015 Mark Alasandro

EU and FDA GMP Regulations: Overview and Comparison

Good Manufacturing Practice ( GMP ) Compliance: GMPs EXPLAINED

Industry Perspective on Manufacturing in Early Development

COMPLIANCE BY DESIGN FOR PHARMACEUTICAL QUALITY CONTROL LABORATORIES INSIGHT FROM FDA WARNING LETTERS

OOS: Back to Basics. Compliant, Effective, Efficient PATH

Enabling Improvement through the Product Lifecycle: Change Management within a PQS

Validation, Verification and Transfer of Analytical Methods (Understanding and implementing guidelines from FDA/EMA, USP and ICH)

Library Guide: Active Pharmaceutical

WHO SAYS COMPLIANCE NEEDS TO BE COMPLICATED?

MANUFACTURE OF INVESTIGATIONAL MEDICINAL PRODUCTS

QbD implementation in Generic Industry: Overview and Case-Study

Process Validation Guidelines. Report highlights 23 rd February 2018

Quality by Design (QbD)

FDA Quality Metrics, Data Integrity and Application of Statistics Throughout Process Validation in a Global Economy

How to Identify Critical Quality Attributes and Critical Process Parameters

Regulatory expectations on impurities in drug substances - Pavia, October 2, Luisa Torchio Euticals SpA

Transcription:

Potential Changes and Minimizing the Impact of Registration CHANGE CONTROL MANAGEMENT FOR A STABILITY PROGRAM Kim Huynh Ba Eecutive Director PHARMALYTIK www.pharmalytik.com Biography Kim Huynh Ba has over 25 years of eperience in analytical development, project management, strategic drug development and stability sciences. She currently is the Eecutive Director of Pharmalytik (www.pharmalytik.com), where she provides consulting and training services to pharmaceutical companies, including companies operating under FDA s Consent Decree on harmonization and optimization of analytical best practices since 2003. In 2011 2012, she took a 2 year leave to join the U.S. Pharmacopeia (USP) as their Director of Pharmacopeial Education Department, where she invigorated their education programs worldwide. Her clients range from large pharmaceutical companies to small contract laboratories in U.S. and also internationally. Kim is an Adjunct Professor at Temple University School of Pharmacy, Widener University and Illinois Institute of Technology (IIT) teaching Pharmaceutical Analysis, cgmps, ICH guidelines and Quality Audit graduate courses. She is also a short course instructor on cgmp compliance and quality topics for several global organizations such as American Chemical Society (ACS), American Association of Pharmaceutical Scientists (AAPS), Pittsburgh Conference, and many other international training groups. Kim is a member of the USP Council of Eperts (2015 2020), where she chairs the Chemical Medicines IV Epert Committee. She is also a member of the Impurities of Drug Products Epert Panel and Food Adulteration Epert Panel. She was the Chair of the USP Good Documentation Practices (GDP) Epert Panel. She is an active member of AAPS and an Alternate Councilor of ACS Delaware Section. She serves on steering committees of AAPS Stability, CMC, and Pharmaceutical Impurities Focus Groups. She is a member of the Eecutive Committee of Governing Board of Eastern Analytical Symposium (EAS) and was their 2013 President. She is also a member of PQRI Stability Shelf Life Committee. Kim has authored numerous technical publications, and book chapters. She is the editor of 2 well known Stability books: the Handbook of Stability Testing in Pharmaceutical Development: Regulations, Methodologies and Best Practices (2008) and Pharmaceutical Stability Testing to Support Global Markets (2010). She is also working on a manuscript for the Analytical Chemistry: An Introduction to Pharmaceutical GMP Laboratory, that is epected to be available in 2018. Kim can be reached at kim.huynhba@pharmalytik.com www.pharmalytik.com (copyright) 1

Overview 1. Managing changes as part of Quality Systems Regulatory epectations to support changes Monitoring Stability as a critical quality attribute Managing changes to stability program 2. Elements of a change control process Preparing changes Managing changes Reinforcing changes 3. Different types of changes affecting stability profile Formulation development Selection of packaging system Analytical procedure API and raw materials Warning Letter Failure to establish and maintain adequate procedures for validating device design as required in 21 CFR 820.30(g). For eample, there were four revisions of the [red acted] shelf life environmental design verification protocol. The first revision was never used, the second revision was used at the start of the validation and the third revision was used at the completion of the real time aging of the units and the verification/validation of the tests. The final revision requires that the real life units be tested at and y months. The testing that was done was done at v and w months, instead of and y months. The testing report was reviewed and accepted by a design cross functional group which included the project manager, regulatory affairs, quality assurance and research and development. QSR; Design Controls; Design Validation 2 Jul 2010 US medical device company www.pharmalytik.com (copyright) 2

Change Control System CFR 211.194 Complete record shall be maintained of any modification of an established method employed in testing. Such records shall include the reason for the modification and data to be verified that the modification produced results that are at least as accurate and reliable for the material being tested as established method. ICH Q7A A formal change control system should be established to evaluate all changes that could affect the production and control of the intermediate or API. Written procedures should provide for the identification, documentation, appropriate review, and approval of changes in raw materials, specifications, analytical methods, facilities, support systems, equipment, processing steps, labeling and packaging materials and computer software. WHY DO WE NEED TO CONTROL THE CHANGES? www.pharmalytik.com (copyright) 3

Change Control Change control is another well known CGMP concept that focuses on managing change to prevent unintended consequences. The CGMP regulations provide for change control primarily through the assigned responsibilities of the quality control unit. Certain major manufacturing changes (e.g., changes that alter specifications, a critical product attribute or bioavailability) require regulatory filings and prior regulatory approval (21 CFR 314.70, 514.8, and 601.12). Guidance for Industry Quality Systems Approach to Pharmaceutical cgmp Regulations, Sep. 2006. Change Control Effective change control activities (e.g., quality planning and control of revisions to specifications, process parameters, procedures) are key components of any quality system. In this guidance, change is discussed in terms of creating a regulatory environment that encourages change towards continual improvement. This means a manufacturer is empowered to make changes subject to the regulations based on the variability of materials used in manufacturing and process improvements resulting from knowledge gained during a product s lifecycle. Guidance for Industry Quality Systems Approach to Pharmaceutical cgmp Regulations, Sep. 2006. www.pharmalytik.com (copyright) 4

Change Management Process Preparing for change Define your change Prepare your team to evaluate and review the change Develop your model & Classify your change Managing Change Develop plan, identify the impacted systems/documents Conduct the Risk Assessment Change Approval Take actions & Implement plans Reinforcing Change Collect and analyze data Verify change implementation Identify gaps and manage resistance, if needed. Monitoring and continuous improvement Drug Product Stability Stability Testing is required by cgmp Stability is a Critical Quality Attributes Stability Data used to set product specifications Stability is to establish a storage condition for labeling Stability is to establish an epiry for commercialization www.pharmalytik.com (copyright) 5

STABILITY TESTING Required by cgmp Regulations (21 CFR 211.166) and global regulations (ICH Q1A(R2), WHO, ASEAN, etc.) FDA defined Stability is a Critical Quality Attribute (CQA) To establish a storage condition for labeling To establish an epiry for commercialization Stability characteristics of API and DP rely heavily on end Product Testing Typically, Quality is measured instead of designed Factors impacting Drug Stability Stability of the Active Pharmaceutical Ingredient (API) from storage Interaction between the API and ecipient Formulation Development Selection of dosage form and Manufacturing process of drug product Selection of container closure packaging system Effect of storage (temperature, humidity and light) Selection of marketing image Handling of the finished products www.pharmalytik.com (copyright) 6

Purpose of Stability Testing Stability studies are used as quality assessment tool to ensure the changes have no adverse impact on the quality of the product over its epiry The purpose of stability testing is to provide evidence on how the quality of a drug substance or drug product varies with time under the influence of a variety of environmental factors such as temperature, humidity and light. Stability testing permits the establishment of recommended storage conditions, retest periods, and shelf lives. ICH harmonized Tripartite Guideline for Stability Testing of New Drug Substances and Products [ICH Q1A] WHAT CHANGES SEEN DURING STABILITY? * FOR DEVELOPMENT PRODUCTS * FOR COMMERCIAL PRODUCTS www.pharmalytik.com (copyright) 7

CMC Changes impact Stability Profile Stability Testing Formulation Development Process Development Packaging Development Method Development API * Quality of a drug substance or drug product in a packaging system must remain within established specifications throughout its retest/epiry. * Specifications = identity + strength + quality + purity www.pharmalytik.com Why changes? Heat Humidity Light Pharmaceutical Product Stability studies are used as quality assessment tool to ensure the changes have no adverse impact on the quality of the product over its epiry. www.pharmalytik.com (copyright) 8

Change of API & Formulation API Selection: Physical: Polymorphic forms, Dissolution, Resuspendability, Aggregation Chemical: Potential degradation products, Degradation mechanism Chirality (as applicable) Impurities control Change of Formulation Eternal factors (Light, Temperature, Humidity, ph) stress studies Accelerated Studies: 40C/75%, 50C Other special conditions thermal cycling freeze-thaw www.pharmalytik.com (copyright) 9

Change of Process Ecipient Compatibility Studies Evaluating drug-ecipient interactions (Q8) For liquids: buffer, ph For solids: Formulation selection Ecipient supplier Process selection Packaging Changes Consider information from API selection from Formulation Development from Process development Link to mechanisms of instability to determine the packaging requirement (protected from light or moisture, etc.) Effect of storage conditions Effect of container orientation Potential for leachables and etractables www.pharmalytik.com (copyright) 10

Analytical Changes Change of analytical technology (UV to HPLC) Change of test method (different columns, NPLC vs RPLC) Change of sample preparation (solvent, buffer, etraction procedure) Addition of new specified degradation products (same specs) Revised method with new specified degradation products Change specifications (wider the specs or shift the specs) Mostly are prior approval or CBE-30 Typically doing both or continue ongoing studies www.pharmalytik.com Impact of Instability Safety and efficacy of drug product are established during development via clinical studies If drug product stability changes beyond established acceptance criteria, established safety and efficacy are no longer applicable. Change of Drug Stability would risk patient safety Quality of finished products decrease (Product is unsafe or ineffective) Potential sub-potent or over-dose products Potential toic unknown impurities Modification of content uniformity/bioavailability Loss of container-closure or microbial integrity Loss of cosmetic or functional aspects Uncontrolled process product investigation product recalls cgmp violations consent decree criminal prosecution www.pharmalytik.com (copyright) 11

Severity of Impact Nature and Compleity of the Drug Product i.e. Dosage form Nature and Compleity of the Changes One change or multiple changes Simple change or comple changes Important to be able to bridge the data before and after changes to evaluate stability of the product Major impact on timelines and resources Stability Requirements 21 CFR 211.166: There shall be a written stability testing program to assess the stability of drug products. 21 CFR 58.31(d): Assure that test and control articles or mitures have been appropriately tested for identity, strength, purity, stability, and uniformity, as applicable. ICH Q7A: GMP for API: Requires stability monitoring of APIs: A documented, on-going testing program is needed to monitor the STABILITY of APIs; results should be used to confirm appropriate storage conditions and retest or epiry dates. www.pharmalytik.com (copyright) 12

Stability Studies Through Product Life Cycle 1. Studies supporting product development Product/Formulation design Manufacturing process Container/closure system 2. Studies supporting registration or marketing application Provide evidence to support Epiration dating periods Label recommendations for storage conditions 3. Studies monitoring product quality Verify the successful eecution of the development 4. Studies supporting post approval changes Make critical changes Typical Changes Leading to Risks Not meeting current product specifications or compliance standards Survival Need for continuous improvement Adaptation Process changes Method changes New Science and/or New Technology New scientific or clinical findings Complying with regulatory or compendial changes Changes caused by Desired Product improvement Changing of clinical/market needs Cost reduction Materials availability (New supplier/vendor) New sites/new facilities Market epansion www.pharmalytik.com www.pharmalytik.com (copyright) 13

Impacts to Product Quality Change of Drug Stability would risk patient safety Quality of finished products decrease (Product is unsafe or ineffective) Potential sub potent or over dose products Potential toic unknown impurities Uncontrolled process product inves ga on product recalls Costly Bad reputa on Lost of customers Compliance Standards cgmp violations 483 s Observa on Warning Le ers consent decree criminal prosecution Lost of License Lost of Registration Lost of customers Impact to Stability Program Major changes Start New Studies Moderate changes Continue Ongoing studies and Start New Studies Stability Program Minor changes Continue Ongoing studies www.pharmalytik.com (copyright) 14

Change Control Process Evaluate the changes What is the reason? What needs to get done? Risk Assessment Is the impact of the change significant? Can it harm a person? Can it hurt the company? Is there a regulatory concern? Plan the Effort Was it the result of a problem? Was it an update? Is it a high priority, visible project? www.pharmalytik.com Risk Assessments Use different tools to assess readiness to change. Assess the scope of the change How big is this change? How many people/products are affected? Is it a gradual or radical change? Assess the readiness of the impact by the change What is the value system and background of the impacted groups? How much change is already going on? What type of resistance can be epected? Assess the strengths of your change management team. Assess the change sponsors and take the first steps to enable them to effectively lead the change process. www.pharmalytik.com www.pharmalytik.com (copyright) 15

Severity of Impact Nature and Compleity of the Drug Product i.e. Dosage form Nature and Compleity of the Changes One change or multiple changes Simple change or comple changes Important to be able to bridge the data before and after changes to assess stability of the product Major impact on timeline and resource Quality By Design Concept: Know what we want Design Space Knowledge Space Control Space www.pharmalytik.com (copyright) 16

QbD Concept Multi variable CQA 1 Knowledge Space CQA 2 Optimum Design Design Space CQA 3 QbD Concept Impact of Changes CQA 1 Knowledge Space CQA 2 Optimum Design Design Space CQA 3 www.pharmalytik.com (copyright) 17

QbD Concept Legacy Product CQA 1 Knowledge Space CQA 2 Optimum Design Design Space CQA 3 Principles of Risk Management The evaluation of the risk to quality should be based on scientific knowledge and ultimately link to the protection of the patient The level of efforts, formality and documentation of the QRM should be commensurate with the level of risk ICH Q9 www.pharmalytik.com www.pharmalytik.com (copyright) 18

Risk Assessment For a scale from 1 to 10 How likely is it to happen? Likelihood of Detection? Severity of the impact? X Probability Detection Severity Risk www.pharmalytik.com Areas of Risk to be identified System Risk Quality Risk Process Risk Product Risk Facility & people Organization Process operations Safety & efficacy Interfaces, operators risk, environment, components (equipment, IT, design elements) Quality systems, controls, measurements, documentatio n, regulatory compliance Quality parameters Quality attributes: measured data according to specifications www.pharmalytik.com www.pharmalytik.com (copyright) 19

CONCLUSION Drug Development is a Process - Changes are inevitable; mostly for continuous improvement. Major changes can derail on-going stability studies and cause delay. Anticipate change and plan in advance. Get input early from stake holder (process, formulation, packaging). Use Science/Data to justify and bridge the changes. Communicate with Regulatory Agencies. Have a solid change control process. Balance the impact of changes (safety, time, resources & money) Culture Changes Industry: More collaboration among business units and across disciplines to apply Quality Systems throughout Less regulatory burden and enhance efficiency Regulatory: Integrate review, inspection and compliance (across disciplines) Implement Quality Systems review Higher assurance of product quality Patients: Higher assurance of product availability and quality www.pharmalytik.com (copyright) 20

References K. Huynh-Ba, ed., Pharmaceutical Stability Testing to Support Global Markets, Springer, 2010. S. Baertschi, K. Alsante & R. Reed, ed., Pharmaceutical Stress Testing: Predicting Drug Degradation, 2 nd ed, Informa, 2012. G. Buehler, Regulatory Perspectives on Product Stability, AAPS 2007 Workshop, Bethesda, MD. L. Wu, Understanding Pharmaceutical Quality By Design, presented at AAPS Webinar. Handbook of Stability Testing in Pharmaceutical Development: Regulations, Methodologies and Best Practices, K. Huynh-Ba (ed.), Springer Publisher, Nov. 2008. Pharmaceutical Stability Testing to support Global Markets, K. Huynh-Ba (ed.), Springer Publisher, Jan. 2010 Handbook of Stability Testing in Pharmaceutical Development: Regulations, Methodologies and Best Practices, K. Huynh-Ba (Ed.), 2008, Springer Publishing. www.springer.com - Post Approval Changes Doing the right thing STABILITY PROGRAM Thank you! Any Question? KIM HUYNH BA Kim.huynhba@pharmalytik.com www.pharmalytik.com www.pharmalytik.com (copyright) 21