Process Validation of a Multi-SKU Drug Product Kevin Maloney, PhD Biogen, Technical Development, Cambridge, MA

Similar documents
Systems-Based Inspections for Cleaning Validation

Further Stability Considerations

ANDAs: Stability Testing of Drug Substances and Products- Industry Perspective

Dr. Earl Dye CMC/GMP Considerations for Expedited Development Programs

Develop an Effective Stability Strategy for Product Distribution

Session 2-Product Design FIM to commercial for a lyophilised (NBE) product Michael Siedler, Abbvie

SESSION 11 Stability Data Evaluate, Set Specifications and Prepare Reports

Changes to a Potency Bioassay for Biotechnology Products: a Regulatory Perspective Kathleen A. Clouse, Ph.D., Director Division of Monoclonal Antibodi

Clinical qualification of specifications - a Regulator s view

Recent experience in scientific advice and marketing authorisations

Risk-Based Strategies for Analytical Method Qualification/Validation Studies to Support Accelerated Product Development

Drivers and Challenges of Developing High Concentration Injectables 7 th Annual PFS & Injectable Summit September 2017

Application of enhanced process and product knowledge to facilitate the lifecycle

Process Validation Lifecycle Approach: A Return to Science

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

A holistic regulatory approach to accelerated CMC development

Hot Topics in Drug Product Process Validation: A Reviewer s Perspective

Setting Specifications for Biotech Products

Extractable and Leachable Challenges From a generic injectable drug development perspective

Systems-Based Inspections for Cleaning Validation

PPQ-to-Approval Timelines Acceleration Approaches at BMS

EVALUATION FOR STABILITY DATA

Commonly Seen Drug Product Related Quality Deficiencies

Key-points of Post approval variation in Korea

A Potential Innovative CMC Solution: Responding To Public Health Needs With An Accelerated Clinical Pathway A Vaccine Example

How to Identify Critical Quality Attributes and Critical Process Parameters

MDI Manufacturing Services

Field trial with veterinary vaccine

Challenges Integrating Regulatory Filings and Pre-Approval Inspection with the Expectations of Current Regulatory Guidance

Challenges with Establishing a Control Strategy for Biosimilars

ICH Q3D RISK ASSESSMENT: REGULATORY SUCCESS AND STANDARDIZED METHODOLOGY FOR NEW FILINGS WILLIAM STEVENS- MERCK & CO., INC.

3M Drug Delivery Systems. April 26, 2011

Preparing the CMC section of IMPD for biological/biotechnology derived substances

CMC Strategy Forum Europe 2016

Biopharmaceuticals A Regulatory Perspective

API Testing Requirements to Support the EI Risk Assessment. Elisabeth Corbett Associate Director, GRS-CMC, Bristol-Myers Squibb November 9, 2016

QUALITY ASSESSMENT METHODS FOR NEW PRODUCT LAUNCHES: PROCESS VALIDATION LIFECYCLE

Accelerated Development for Biopharmaceutical Products. Josh Grieco February 5th, 2018

Strategic Outsourcing for Success

Biowaiver Approaches for Solid Oral Dosage Forms in New Drug Applications Poonam R. Delvadia, Ph.D. Division of Biopharmaceutics\ONDP\OPQ\CDER\FDA

Advantages, Opportunities & Challenges of Adopting the Post Approval Change Management Plan (PACMP)

PROCESS VALIDATION ROCHAPON WACHAROTAYANKUN, PH.D.

Regulatory Perspective on Analytical Method Validation During Product Development

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

GMPs for Method Validation in Early Development: An Industry Perspective (Part II)

A Hands-On Guide to Ultrafiltration/Diafiltration Optimization using Pellicon Cassettes

Cleaning Validation Challenges in Personal Care. June 15, 2017

The Role of Quality Risk Management in New Drug Development and Manufacturing

Developing the Control Strategy for Enhanced Testing and Continuous Monitoring

Regulatory Consideration for Biotechnology Products: Clonality of the Production Cell Bank

Systems-Based Inspections for Cleaning Validation

Lifecycle Management of Commercially Approved QC Potency Assay for a Biotech Product. A Case Study

Clare Simpson 21 st November Content courtesy of Steve Jones, Paul Illot, Bert Frohlich, Thomas Ryll Biopharm Services

A Late-Stage Monoclonal Antibody in the FDA QbD Pilot Program: Moving from Concepts to Implementation

Risk Management in Drug Product Manufacturing with Emphasis on Batch Documentation/Execution

Established Conditions: Reportable CMC Changes for Approved Drug and Biologic Products Guidance for Industry

RapidFACT: Accelerated Formulation Development for Poorly Soluble Drugs and Modified Release Products

QbD In Drug Development. Mathew Cherian Ph.D. Director & Senior Fellow Pfizer, USA

Common Issues in Qualification and Validation of Analytical Procedures

Effect of Freezing on Lyophilization Process Performance & Drug Product Cake Appearance of Biologics: A Case Study

2017 AAM CMC Workshop

V&V Best Practices. CASSS, CMC Strategy Forum Steven W. Badelt, PhD Managing Partner Suttons Creek, Inc. SUTTONSCREEK.COM

Practical Considerations in Developing High Concentration Antibody Formulations

Pharma&Biotech. ADC Process Transfer from a CMO Perspective: How to Make a Collaboration Successful

ANALYTICAL VALIDATION CHALLENGES DURING THE RAPID DEVELOPMENT OF KEYTRUDA

Comparability study to support commercial process change via stability study

Technical Report No. 60 Process Validation: A Lifecycle Approach

CMC Consideration for the Development of Regenerative Medical Products

Essentials in Stability Analysis and Expiry Determination

GUIDELINE FOR THE STABILITY TESTING

FDA Guidance and Current Experience with New Drug Submissions

LEGAL REQUIREMENTS FOR STABILITY

Preview of New PDA Technical Report on Process Validation

Lifecycle of NIR Analytical Methods: Regulatory Perspective

Reference Standards: Overview and Strategy for Development to Commercialization

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

Page 1 of 12. Muscle Relaxant Property of Company X. Compound X (Study No. YYY) DATE

Pharmaceutical Development (Drug Substance & Drug Product) for Visceral Leishmaniasis candidate DNDI-6148

Collaborative Process Engineering Framework for Ground Vehicle Systems Manufacturing #193

GPhA Fall Technical Conference Nov 2-5, 2015 Bethesda, MD ICH M7 Guidance Overview and Current FDA Perspectives

Data Integrity Prioritization and Gap Analysis to Prove Data Integrity

EUROPEAN COMMISSION ENTERPRISE AND INDUSTRY DIRECTORATE-GENERAL. EudraLex The Rules Governing Medicinal Products in the European Union

Paving the way to FDA? tips and pitfalls. The Nextar start up support program Dr. Orna Dreazen- CEO

EU Regulatory Perspective and Expectations. Sven-Erik Hillver Senior expert, QWP delegate Medical Products Agency, Sweden

A Hands-On Guide to Ultrafiltration/ Diafiltration Optimization using Pellicon Cassettes

Guidance for Industry Process Validation: General Principles and Practices; A Contract Manufacturing Organization's Approach

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

Guidance for Industry

Elemental Impurities and Animal Drugs An Update from CVM

Modern Approaches to Process Understanding

Being Clinically Relevant While Setting Specifications

Integration of Risk Management into Product Strategy and Portfolio. Lori Richter Senior Consultant: QRM and Quality Systems ValSource, LLC

Introduction to CMC Regulatory Affairs

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

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

Delivery of materials (usually finished product from a distribution center) to a first paying customer external to Site

How we set specifications for impurities (including Genotoxic impurities) 24 May 2017 Elisabeth Kovacs, Apotex CSO Chemistry and Analytical Sci.

Graduate Certificate in Pharmaceutical Regulation Chemistry, Manufacturing and Controls (CMC) Workshop March 2019

Challenges and Opportunities in the Development of integrated Drug Device Combination Products CMC Strategy Forum, Tokyo, 5 December 2017

PDA: A Global. Association. Matrix Approach to Media Fills. (c) 2012 Catalent Pharma Solutions. All rights reserved.

Transcription:

Process Validation of a Multi-SKU Drug Product Kevin Maloney, PhD Biogen, Technical Development, Cambridge, MA CASSS CMC Forum, DP Validation, July 2016, Gaithersburg, MD 1

Multi-SKU, Weight-Based, DPs Versus Fixed Dose DPs: More to Contend with in Lifecycle Approach to Validation SKU = defined here as a single dose contained in one container Fixed Dosing Weight-Based Dosing Example Weight (kg) Delivered Volume (ml) of for Fixed Versus Weight (kg) Fixed Concentration (e.g., 150 mg/ml) Fixed Container Size (e.g., 5 ml) 1X mg/kg 10X mg/kg 1X mg/kg 10X mg/kg 60 70 80 90 100 One Container, One Strength, One Fill Target N = 3 PPQ 60 0.4 6.7 ml required for 70 the full dose level range 80 90 Multiple DP fill volumes 100 required for single strength N = Bracketed PPQ Min. 2 doses required Potential lifecycle issues with weight-based DPs: Improper # of SKUs and fill targets can encourage dose -splitting and impact COGs Number of SKUs can complicate Design/PPQ Activities, DP design work can mushroom Bracketed stability design and expiry management Label/pack can further expand finished goods configurations Significant resource burden for DS/DP Mfg changes (Changes, or New Sites) 2

Weight-Based DPs: Leverage Science to Select Design Elements to Control Complexity Case Study Fully formulated DS at the highest expected DP dose strength helps maintain flexibility Eliminates the need for DP compounding unit operations. Single-concentration, varied fill volume approach can help simplify with process and stability control strategies For well-formulated proteins, concentration will drive stability. Expect a reasonably broad, dose-independent stability profile. Aside from fill weight controls, enables a common, dose-independent, process-wide control strategy to be developed Enables future lifecycle options for alternate routes of delivery Benefits of approach are more apparent for large # of doses Maximize DS utilization, minimize DP leftover volumes 3

Expected Stability Outcomes: Constant Strength vs Constant Volume Vary fill volume, same conc. Vary concentration, same fill vol. Stability Stability Lowest Vol. Highest Vol. Lowest Conc. Highest Conc. Would expect very limited or no impact of fill volume on stability for the same formulation matrix and product concentration Contrast to same matrix but with different product concentrations; expect regions of similar stability but overall trend towards decreasing stability Design of pre-validation activities will inform the likelihood of this approach Managing a varied stability profile more complex situation to manage overall 4

Stage 1 Design Stage 2 - Qualify Stage 3 - Verify Case Study: Stage 1 and 2 Lifecycle Strategy File ICH Stage 1/Design Stage 2/PPQ markets Follow-on Site PPQ Pre-Launch Line Commercial Launch Line Post-Launch, New DS/DP Sites File ICH, Global Dose Selection Knowns: 2 or more ISO vial container sizes, 6-8 DP doses, same formulation, same concentration, vary fill volume per dose, use of a platform fill-finish process Challenges: Process and stability design work being performed on Pre-Launch line in commercial DP facility. PPQ to be performed on Launch Line in same facility. Both lines considered equivalent. Dose selection could vary 10-fold, may need to initiate PPQ prior to final dose selection Expecting 2 or more manufacturing sites to meet global supply demands What enables this approach? What are the risks? 5

Stage 1 Enablers: Stability Bracketing on Line 1 Fill-finish platform established on Pre-Launch Line Stage 1 Design Stage 2 - Qualify Stage 3 - Verify Weight (kg) 60 70 80 90 100 Delivered Volume (ml) of X mg/ml strength 1X mg/kg 10X mg/kg 0.4 6.7 ml required for the full dose level range Multiple DP fill volumes required for individual doses (eg, 150 mg/ml) Container 1: 2 fill targets (smallest doses) Container 2: 6 fill targets (largest doses) 6 of the 8 doses intermediate doses Stability Bracketing (reduced design per ICH Q1D): N=3 coverage for extreme conditions ; process & stability (bracket ends) Factors: Surface/Vol, Headspace/Vol, Stopper area/volume, etc. One lot each for the 6 intermediate doses, multiple DS batches to be used to evaluate risk of DS variation on DP stability 12 ICH studies from pre-launch Line to establish stability bracket ahead of PPQ on Launch Line. For PPQ & follow on-sites, how long do you continue repeating brackets? Stability and bracketing is also a significant issue for other non-ich adopters. 6

Stage 1 Design Stage 2 - Qualify Stage 3 - Verify Stage 1 2 Enablers: Process and Facility Platform process with prior knowledge of antibodies (150-200 mg/ml) Pre-Launch vs Launch Line: shared technologies, equivalent capabilities Both lines in the same facility risks of transfer will be documented Raw materials will be identical. Control strategy will be the same. Goal with successful transfer, and comparability package is leveraging of stability data from Pre-Launch to PPQ/Launch material Mitigates launch risks of limited PPQ stability data availability at time of BLA/MAA filing 7

Stage 1 Design Stage 2 - Qualify Stage 3 - Verify Stage 2 PPQ: 12 runs for 8 SKUs SKU: Lowest to Highest dose SKU 1 (lowest dose) Container Size: A or B PPQ Run s CCI val. Stability Lots Batch Size Hold Time A 3 Y 3 1 max Max (2) SKU 2 A 1 N 1 SKU 3 B 1 N 1 SKU 4 B 1 N 1 SKU 5 B 1 N 1 SKU 6 B 1 N 1 SKU 7 B 1 N 1 SKU 8 (highest dose) B 3 Y 3 1 min Max (1) PPQ approach mirrors strategy started from Pre-Launch line 3 runs at both best/worst-case process & stability conditions: 6 runs including hold time, batch size and CCI validation activities. 1 PPQ run for each of the intermediate strengths Issues/Risks for Intermediate SKUs and non-ich markets. Requirements for complete, non-bracketed data sets. Contrasts greatly with bracketing approaches for ICH-regions, highly impactful to drug development and filing process. 8

Stage 1 Design Stage 2 - Qualify Stage 3 - Verify Follow On DP Mfg Sites; Leveraging Site 1 Prior Knowledge for Risk-Based Site 2 PPQ Site 1: PK = 24 stab lots, 12 PPQ runs Site 2; N =? PPQ runs SKU: Lowest to Highest dose SKU 1 (lowest Container Size: A or B PPQ Runs CCI val. Stability? A 3 Y Y (3) dose) SKU 2 A 1 N Y SKU 3 B 1 N Y SKU 4 B 1 N Y SKU 5 B 1 N Y SKU 6 B 1 N Y SKU 7 B 1 N Y SKU 8 (highest dose) B 3 Y Y (3) Batch Size 2 max/1 min 1 max/2 min Hold Time Max (3) Max (3) SKU: Lowest to Highest dose SKU 1 (lowest dose) SKU 2 SKU 3 SKU 4 SKU 5 SKU 6 SKU 7 SKU 8 (highest dose) Container Size: A or B PPQ Runs CCI val. Stability? A 3 Y 3 A B B B B B B 3 Y 3 Batch Size Hold Time Validate at site 2 focusing on risks (CCI, 6 runs across 2 containers): Same platform process & control strategy, no new facility, raw material or equipment risks 9

Bracketing and Risks Don t assume you can bracket Establish it: Stage 1 can drive PPQ scope of work, or establish in Stage 2 for follow on sites Change in dose after Stage 2 initiation to either widen or narrow the initial bracketing Narrowing No technical impact to state of validation of the process, nor any impact on stability, still within your bracket What is the regulatory risk? The original high & low SKUs at time of filing will not be marketed. The new bracket data set is not n=3. Widening To either higher or lower dose SKUs A more impactful technical and regulatory issue, you have exceeded the bracket 10

Summary Validation concept for a high volume, multi SKU DP requires effective use of bracketing, identification of best/worst case process and stability conditions, and leveraging of prior knowledge should be used for follow on manufacturing sites. Design elements can be chosen ahead of time to minimize complexity for multi-sku DPs Stage 1 activities are opportunities to assemble significant knowledge space information on the process and establish stability profiles well ahead of PPQ stage This equates to significant reduction in risk at both initial and follow-on DP sites. Due to risk being low (Stage 1 and Stage 2 prior knowledge), follow-on site Stage 2 activities should be feasible under reduced protocols, scope, and filing burden. 11

Acknowledgements Biogen colleagues: Technical Development: Curtiss Schneider, Steve Lantz, and John Ruesch Quality Stability: Brian Nunnally, Philip Pue-Gilchrist Regulatory: Kimberly Wolfram 12