Presentation Scope. Rapid Microbiological Methods (RMM) Regulatory Acceptance: Policies and Expectations. Rapid microbiological methods:

Similar documents
Rapid Microbiological Methods Understanding the Technologies and Regulatory Expectations for Validation and Implementation

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

Erin Patton, MS Senior Product Specialist Charles River Labs, Microbial Solutions

Questions and Answers on allogenic stem cell-based products for veterinary use: specific questions on sterility

European Pharmacopoeia Chapter Alternative methods for control of microbiological quality

Rapid Methods & Technologies

A risk based approach to managing environmental excursions

Specifications, Methods and precedence of source with particular reference to microbiological requirements in TGOs.

Validation Study of Rapid Assays of Bioburden, Endotoxins and Other Contamination

USP Chapter 823 USP 32 (old) vs. USP 35 (new)

Overview of a sterility assurance program for PET drugs

Regulatory Implications for Global Manufacturing Development of Regenerative Medicines

10/31/2014. Automated and Rapid Microbiological Methods: Selection and Validation. Microbiology: Past. Microbiology: Today

Copyright. Jeremiah J. Kelly (2015). All rights reserved. Further dissemination without express written consent strictly prohibited.

The Parenteral Drug Association

Microbiology Research Associates

FDA s Guidance for Industry

Tests to Support Sterility Claim. Imtiaz Ahmed

The Microbiological Requirements of a Stability Study. Ngoc Anh-Thu Phan 19 th June 2012

Sterile Compounding elearning Course Curriculum 28 lessons with 33 hours of CE. Fundamentals of Sterile Compounding (8 lessons/8 hours CE)

Current Topics For Sterile Generic Drug Products

Guidance. Media Fills for Validation of Aseptic Preparations for Positron Emission Tomography (PET) Drugs DRAFT GUIDANCE

Sterile Compounding elearning Course Curriculum 28 lessons with 33 hours of CE. Fundamentals of Sterile Compounding (8 lessons/8 hours CE)

Guideline on the requirements for quality documentation concerning biological investigational medicinal products in clinical trials

US FDA: CMC Issues for INDs

USP <1116> and Contamination

Beth Hutchins, PhD PhRMA ICH Gene Therapy Discussion Group

Appropriate Control Strategies Eliminate the Need for Redundant Testing of Pharmaceutical Products

Application of Quality Risk Management Tools for Cell Therapy Manufacturing

Flow cytometry tools for characterization of GMP cell products

COMPLIANCE WITH EU ANNEX 15: VALIDATION AND QUALIFICATION

Food and Drug Administration (FDA) 101

Microbiological Cleaning Method Validation

Annex A2. Guidance on Process Validation Scheme for Aseptically Processed Products

Overview of Biologics Testing and Evaluation: Regulatory Requirements and Expectations. Audrey Chang, PhD, Senior Director Development Services

Validation of Sterilizing Grade Filters

GMO Technology Conference

Regulatory considerations for manufacturing and testing of investigational chimeric antigen receptor (CAR) T-cell products

Microbiological Consideration for Non-Sterile Pharmaceutical

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

MICROBIOLOGY AND NEW TECHNOLOGY HOW TECHNOLOGY CAN IMPACT ON MICROBIOLOGICAL CONTROL

á61ñ MICROBIOLOGICAL EXAMINATION OF NONSTERILE PRODUCTS: MICROBIAL ENUMERATION TESTS

MicroSEQ Rapid Microbial Identification System

Guideline on requirements for the production and control of immunological veterinary medicinal products

Contamination Risk Assessment in Aseptic, Non-Sterile and Terminally Sterilized Products March 7 th & 8 th Raleigh, NC

Initiation of Validation

International Journal of Pharma and Bio Sciences DEVELOPMENT OF ACCELERATED STABILITY PROTOCOL FOR SILDENAFIL TABLETS A EUROPEAN PERSPECTIVE REVIEW

2018 North America Lab Services Fee Schedule

Environmental. National Legionella Conference Sample Taking Workshop. Right Solutions Right Partner

ALTERNATIVE METHODS FOR CONTROL OF MICROBIOLOGICAL QUALITY

ICH Q8/Q8(R)

Activities of the USP Microbiology Subcommittee of Revision During the Revision Cycle

Agenzia Italiana del Farmaco

The long anticipated draft of the FDA s

MANUFACTURING MICROBIOLOGY. Chad Ronholdt, B.Sc., MBA Vice President Strategic Development

cgmp/iso CLIA Experience Unsurpassed Quality

WHO GUIDELINE Stability testing of active pharmaceutical ingredients and finished pharmaceutical products

European Directorate for the Quality of Medicines & HealthCare (EDQM)

Development of non-substantially manipulated cell-based ATMPs 1 : flexibility introduced via the application of the risk-based approach

Applying Cleaning Validation Principles in the Manufacture of Medical Devices and Diagnostics August 20, 2014 IVT San Diego

sterility assurance Prove the power and your processes

Replacing Analytical Methods for Release and Stability Testing CBER Perspective

Final text for addition to The International Pharmacopoeia

PRAXIS. A publication by Bioengineering AG

Update to the Manufacturing Principles for Medicinal Products

MICROBIAL LIMIT TESTS FOR RAW MATERIALS AND NON-STERILE PRODUCTS REVIEW-DISCUSSIONS-INTERPRETATIONS

Bioburden Contamination Control: A Holistic Overview

EU and FDA GMP Regulations: Overview and Comparison

Avoid recurrent microbial contamination using trending of historical data. El Azab Walid Technical Service Manager STERIS

Setting Specifications for Biotech Products

PAT & Risk-Based Initiatives: Implementation Issues PDA New England - 8 th Dec Cliff Campbell B.E., C.Eng. CC&A Ltd., Cork, Ireland

STS Directory Accreditation number: STS 0268

HCT/P Regulation vs 361 Products

ASEPTIC BIOPHARMACEUTICAL MANUFACTURING: FAQs

Case Study: Examples Relating to the Quality Control of Cell-based Products

Starting Material Selection for Type II Drug Master Files

Review Validation of aseptic processes for pharmaceuticals

Protocol Reference: Verification Protocol

Environmental Monitoring of Aseptic Processing Areas - 1

Guidance Document. Microbiological Process Validation & Surveillance Program [No. 5, version 2, July 18, 2016]

BIOPHARMACEUTICAL PROCESS EVALUATED FOR VIRAL CLEARANCE

Preparing Your Aseptic Processing Facility for an FDA Inspection. Valerie Welter, Director Quality Bayer HealthCare March 10, 2014

TECHNICAL AND REGULATORY CONSIDERATIONS FOR PHARMACEUTICAL PRODUCT LIFECYCLE MANAGEMENT Q12

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

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

Automated methods in the Microbiology Lab-Issues and Troubleshooting! Presented By: Dr B. Oboudi

Microbiological Quality Control as Described in the Compendia. Scott Sutton, Ph.D.

3M Purification Inc. Technical and Scientific Services Global Support for the Life Science Industry. Global Expertise delivered locally

INTERNATIONAL RESEARCH JOURNAL OF PHARMACY ISSN Review Article

INFORMATION ON JAPANESE REGULATORY AFFAIRS

Handbook of Microbiological Quality Control

The largest network of harmonized bio/ pharmaceutical GMP product testing labs worldwide, Eurofins BioPharma Product Testing enables companies to

Method Suitability Report Membrane Filtration Sterility Test with QTMicro Apparatus

Modern Analytics for Biologics: Platforms, Multi-Attribute Methods and Real-Time Release

FINAL DOCUMENT. International Medical Device Regulators Forum. Medical Device Regulatory Audit Reports

PERFORMANCE QUALIFICATION PROTOCOL HVAC SYSTEM

DISINFECTION QUALIFI CATION TESTING CONSIDERATIONS FOR THE ASEPTIC AND CLEANROOM MANUFACTURING ENVIRONMENT

Growth Promotion Test Guide for Media Used in Sterility Tests

Transcription:

Rapid Microbiological Methods (RMM) Regulatory Acceptance: Policies and Expectations Karen Longstaff Director, Microbiology Section Laboratories Branch Medical Devices and Product Quality Division, TGA EDQM International Microbiology Symposium 10 October 2017 Presentation Scope Rapid microbiological methods: Overview of relevant TGA legislation Relevant guidance documents TGA s expectations TGA s experience 1

Who is Australia s Regulator? The Therapeutic Goods Administration was established in 1990 to safeguard and enhance the health of the Australian community through effective and timely regulation of therapeutic goods Health Safety Regulation It provides a national system of controls relating to the quality, safety, efficacy and timely availability of therapeutic goods used in, or exported from, Australia 2 TGA How We Operate Part of the Australian Government Department of Health Decisions based on the Therapeutic Goods Act 1989 Main offices in Canberra satellite offices in Sydney, Melbourne, Adelaide and Brisbane Operations are primarily cost recovered (98%): Industry pays fees for making applications & annual charges for products they are responsible for 3

Laboratories Branch: Fast Facts Origins in the National Biological Standards Laboratory 1958 Laboratories hold ISO 17025 accreditation Average staffing level: ~ 90 FTE Five Sections (plus small management & coordination unit): Biochemistry Biomaterials and Engineering Chemistry Immunobiology Microbiology All sections perform essentially the same work: Testing Evaluation of quality aspects for products seeking entry on ARTG Advice Standards development 4 Laboratory Testing Assesses QUALITY Quality: Composition, strength, potency, stability, purity, bioburden, design, construction and performance Testing: Assess compliance with required standards or guidelines, e.g.: Default Pharmacopoeias Therapeutic Goods Orders International standards Compositional guidelines 5

TGA s Default Standards USP, Ph. Eur. and BP recognized as default standards since 1 July 2009 under Therapeutic Goods Act 1989 Permit alternative methods of analysis for control purposes Referee Method: Official pharmacopoeial method 6 TGO 77 Microbiological Standards for Medicines Requirements for finished products (effective January 1, 2010): Sterile medicines Multidose medicines Non-sterile medicines Refers to Ph. Eur., BP and USP test for sterility, preservative efficacy and microbial limits test methods: Allows alternative methods of analysis 7

RMM Guidance Documents No TGA guideline document Default Standards: USP <1223> Validation of Alternative Microbiological Methods BP SC IVL/Ph. Eur. 5.1.6 Alternative Methods for Control of Microbiological Quality PDA Technical Report No. 33 (2013) Evaluation, Validation and Implementation of Alternative and Rapid Microbiological Methods ISO17025 General requirements for the competence of testing and calibration laboratories: Validation of non-standard methods (cl. 5.4.4 & 5.4.5.) 8 RMM: Administrative Aspects TGA Legislation: Approval based on individual finished product registration No mechanism to provide separate approval for equipment/rmm TGA cost recovery: Evaluation fees charged in relation to specific product not process GMP inspection and manufacturer licensing fees Investigate possible options to recover costs for evaluation of an RMM: Evaluate RMM, then charge fee for each product variation application? Charge fee for RMM master evaluation and allow self-assessable changes for subsequent product using that RMM? Consider changes to existing legislation to facilitate evaluation of new technologies? 9

RMM: Administrative Aspects TGA acknowledges and is supportive of new technologies: No specific policy statement published on requirements RMM evaluation by the TGA: As GMP inspection follow-up (e.g. in-process testing) During marketing application (e.g. batch release testing, stability) Via post-market variation (e.g. batch release testing, media fills) Commercial-in-confidence considerations: Primary validation studies conducted by equipment supplier Ability to access technology master file: Might require separate liaison with RMM supplier to access information not available to RMM user 10 Examples of RMMs Used for Product Supplied to Australia RMMs not common: Primarily compendial methods: Raw material, process water, in-process and batch release testing Locally and overseas manufactured product Test for Sterility: Milliflex Rapid System (ATP Bioluminescence) ScanRDI (fluorescent cell labelling and laser scanning (solid phase cytometry) Microbiological contamination testing of blood and tissue-based products: BacT/ALERT Microbial Detection System 11

Examples of RMMs Used for Product Supplied to Australia Bioburden: Celsis Systems (ATP bioluminescence): Non-sterile manufacturing: Screening out negatives in low bioburden product Soleris Optical System: Non-sterile manufacturing: Finished product testing complementary medicines Validation in progress BioLumix System (optical system): Aware of general interest in system 12 Examples of RMMs Used for Product Supplied to Australia Media Fills: ScanRDI Organism Identification: MALDI-TOF (matrix assisted laser desorption ionisation time-of-flight) MicroSEQ Rapid Microbial Identification System Whole genome sequencing (isolate reference laboratory) Phylogenetic analysis outbreak clusters 13

TGA s Validation Expectations DQ: RMM manufacturer IQ and OQ: RMM manufacturer PQ (MPQ): Often jointly by RMM manufacturer and user on sample types Then verification in-house by user Validation criteria (as applicable): Accuracy Precision Specificity Limit of detection Limit of quantification Linearity Range Ruggedness Robustness Equivalence/comparative testing 14 TGA s Expectations Versus Experience Some companies think and talk about using RMM Discuss intentions with TGA: We encourage adoption of RMM We discuss regulatory expectations on case-by-case basis Validation work often already performed by Company Company might continue with compendial method: Why? Expect regulatory hurdles? Delays to product marketing? Prefer to rely on referee method for product batch release? Cost? 15

Reluctance to change from compendial method Access to relevant microbiological expertise: Limited or no microbiological expertise on site Samples contract testing laboratory Contract testing laboratory uses compendial method High initial costs for RMM: Complex technological platforms and sophisticated equipment: More complex than traditional compendial techniques Validation: Whole system, software, database customisation, data integrity, microbiological performance Validation effort and possible challenge by Regulator might be a barrier Low sales volume for inexpensive product Starting to see change: Consider business risk and management of product quality Cost/savings over the long term 16 Reluctance to change from compendial method Not willing to be the first to dip toes into the water RMM cell count might be higher than compendial method: Historical trends might be affected Does it mean my product is unsafe? Does product have a history of safe use? Is it manufactured using a well-controlled process? Higher cell count doesn t necessarily mean a new patient safety risk exists User to assess risk of using RMM for potential for more positive results New method might allow for improved quality decisions on product 17

Reluctance to change from compendial method Referee method: Compendial method is referee method Dispute a test result with Regulator: Generally use referee method and abide by result Option for all parties to agree on actual method to be used: Compendial method might not be suitable: E.g. Burkholderia cepacia complex contamination and microbial limits test method Results of agreed method prevail 18 Summary Recognition that timely microbiological data is vital for: Process monitoring and control Product release RMM offer risk reduction, faster time to result and cost savings: Manufacturers, laboratories and Regulators are adapting to change Starting to move cautiously from traditional methods to implement RMM New method: Provide scientifically sound measure of microbial quality Its limitations should not exceed those of compendial method Should be able to detect same adverse trends as compendial method Important to identify user requirements and determine how these can be met: Work closely with RMM suppliers, technical advisors, and regulators Equipment selection, validation, documentation, training, maintenance, ongoing support etc. 19

Opportunities for Rapid Microbiology Methods in Pharmaceutical Manufacturing Dr. Lynne A. Ensor Director Division of Microbiology Assessment Office of Process & Facilities Office of Pharmaceutical Quality Center for Drug Evaluation & Research/FDA Disclaimer This presentation reflects the views of the presenter and should not be construed to represent FDA s views or policies. 2

Center for Food Safety and Applied Nutrition Center for Drug Evaluation and Research Center for Biologics Evaluation and Research Center for Devices and Radiological Health Center for Tobacco Products Center for Veterinary Medicine National Center for Toxicological Research Office of Regulatory Affairs 3 OPQ/OPF Division of Process Assessment I DPA II DPA III Division of Inspectional Assessment Division of Microbiology Assessment 4

Explore the Possibilities focus on your specific needs Validation Biological Indicators In-Process Environmental monitoring Water Bioburden Sterility Finished Product Sterility Microbial Limits Other???? 5 Validation Guidance & Recommendations USP <1223> Validation of Alternative Microbiological Methods PDA Technical Report 33 Evaluation, Validation and Implementation of Alternative & Rapid Microbiological Methods ICH Q2A Validation of Analytical Procedures The objective of validation of an analytical procedure is to demonstrate that it is suitable for its intended purposes EP 5.1.6 USP Stimuli Article The Development of Compendial Rapid Sterility 6

RMM Validation Criteria Table 1. Validation Parameters by Type of Microbiological Test (USP<1223>) Validation Parameter Qualitative Tests Quantitative Tests Accuracy No Yes Precision No Yes Specificity Yes Yes Limit of detection Yes Yes Limit of quantification No Yes Linearity No Yes Operational (dynamic) range No Yes Robustness Yes Yes Repeatability Yes Yes Ruggedness Yes Yes Equivalency Yes Yes 7 RMM Validation Recommendations Provide Equivalent Product Quality Method yield equivalent or better results Choose your statistical analysis methods carefully Use microorganisms that are relevant to your product and manufacturing environment Understand limitations of your RMM and perform studies using worst-case scenarios Product interference with test method? 8

Potential Submission Strategies NDA, ANDA or BLA Original Submission Supplement Guidance for Industry (GFI): Changes to an approved NDA or ANDA (2004) GFI: Changes to an Approved Application for Specified Biotechnology and Specified Synthetic Biological Products (1997) Prior Approval (for Release Test) Changes Being Effected (CBE-0 or CBE-30) Annual Report GFI: CMC Postapproval Manufacturing Changes to be Documented in Annual Reports (2014) GFI: CMC Postapproval Manufacturing Changes for Specified Biological Products to be Documented in Annual Reports (2017) Comparability Protocol GFI: Comparability Protocols for Human Drugs & Biologics: Chemistry, Manufacturing, and Controls Information (Draft 2016) Drug Master File 9 Potential Submission Strategies (2) Emerging Technology Team - CDER/OPQ & ORA representatives - Mechanism for pre-submission interactions - CDER-ETT@FDA.HHS.GOV Encourage Pre-submission Interactions - Increases likelihood for first review cycle approval 10

Current Pharma Usage of RMM Environmental Monitoring Surface Viable Air Water Monitoring In-Process Bioburden Release testing Microbial enumeration in drug product release (non-sterile) Sterility 11 Potential Barriers to change Reservations for Industry Resources Validation Increased sensitivity of test Concern for regulatory scrutiny or approval (review & inspection) 12

Case study #1 Non-sterile, aqueous drug product Proposed Microbial Limit RMM Test 2 Tier decision tree process: Tier 1: RMM using ATP bioluminescence test Negative for ATP No further testing Positive for ATP Tier 2: Classic USP<61> & <62> testing performed, along with in-house test method for BCC 13 Case study #1 (2) Provided: RMM validation data & BCC method and validation data Requested: RMM method/sop Method verification for Tier 2 test (USP <61> & <62>) requested Resolution: Requested RMM method and USP <61> & <62> method verification data provided 14

Case study #2 Non-sterile, multi-dose topical drug product In Process & Release Specifications for DP: Microbial Limits Total aerobic microbial count (TAMC) Total yeast and mold count (TYMC) Pseudomonas aeruginosa Staphylococcus aureus Salmonella E.coli Burkholderia cepacia Limit NMT 200 cfu/ml (Max) NMT 20 cfu/ml (Max) Negative Negative Negative Negative Negative 15 Case study #2(2) Proposed Microbial Limit RMM Test 2 Tier decision tree process: Tier 1: RMM using ATP bioluminescence test Negative for ATP No further testing Positive for ATP Tier 2: Classic USP<61 & <62> testing performed (pour plates), along with in-house test method for BCC 16

Case study #2(3) Adequate validation data provided in application Facility concerns 17 Case study #2(4) The FDA acknowledges the Burkholderia cepacia complex (BCC) testing is included in the release specification and data are also provided for the exhibit batches. However, more information is needed. Please address the following points: 1. Identify potential sources for introduction of BCC during the manufacturing process and describe the steps to minimize the risk of BCC organisms in the final drug product. 2. We recommend that potential sources are examined and sampled as process controls. These may include raw materials and the manufacturing environment. 3. A risk assessment for this species in the product and raw materials is recommended to develop sampling procedures and acceptance criteria. 18

Case study #2(5) Provided expanded EM program to collect in-process bioburden data & cleaning surveillance data Data used to developed criteria for these in-process tests (for various samples and stages of manufacturing) Cleaning Validation Master Plan developed 19 Case study #2(5) Cleaning Validation Master Plan: Risk Assessment Uses both USP and In-house isolates for cleaning & sanitization efficacy studies Established bioburden baseline criteria (prior to cleaning) Confirmation of cleaning process effectiveness for objectionable organisms Established frequency for re-validation of cleaning processes 20

Case study #2(5) 21 Case study #3 Sterile lyophilized powders & Sterile solutions Proposed change Switch from USP <71> sterility testing on product release to a RMM RMM based on membrane filtration and fluorescent cell labeling 22

Provided: - Validation: Case study #3 (2) - Generally followed lists of validation parameters from USP <1223> & PDA TR 33 - Studies included a variety of microorganisms (including slow growers and environmental isolates) - Used relevant inoculation levels - Included examination of all products for interference with fluorescent detection system - Criteria for invalidating the test and retesting described - Can revert to USP <71> Sterility test results on 10 batches of product using both test methods not necessary! 23 Case study #4 Sterile aqueous solution Proposed change Switch from USP <71> sterility testing on release to a RMM RMM based on CO 2 production and detection of colorimetric change 24

Case study #4 (2) Provided: RMM validation data Requested: Sample incubation temperatures, which appeared to be undefined A variety of microorganisms be used in validation studies (including slow growers or stressed cells) A tighter limit of detection study acceptance criterion (as 10 CFU was too high) Resolution: Method validation deficiencies addressed in amendment 25 Contact Information Lynne A. Ensor, Ph. D 240-402-8627 lynne.ensor@fda.hhs.gov 26

27

www.pei.de Rapid Microbiological Methods: Current Status and Regulatory Perspective Dr. Oleg Krut Section Head EDQM-Conference 10-11. 10. 2017 PEI as NCA for biologicals products ensures Microbial and Pyrogenic Safety of Vaccines and Monoclonal Antibodies Blood Products e.g. - albumin, IVIG - platelet- or erythrocyte concentrates Tissue preparations e.g. - cornea - musculoskeletal tissue - heart valves, vessels - bone marrow-derived stem cells Advanced Therapy Medicinal Products (ATMPs) e.g. - genetically engineered cells - somatic cell therapy - tissue engineered products 2 Cell and tissue products

Cell and tissue products are challenging for microbial safety testing Final product cannot be sterilized (loss of function): - Viable human material (cells/tissues) Source material sterility cannot be guaranteed - contamination rates up to 50-90% -> aseptic procurement - establishment of cell banks usually not possible - limitations of donor exclusion criteria -> subclinical infections Cultivation or storage at conditions permissive for growth of pathogens Interference with testing procedures - highly heterogeneous material (solid organs, skin biopsies, bone marrow, adipose tissue) - contain antibiotics to reduce initial bioburden => masking possible contamination Compendial methods for microbiological control are not rapid 3 Current status of microbiological control for cell and tissue medicinal products Parameter Test Compendial Method Duration bacterial / Sterility / 2.6.1 14 days Sterility Testing (2.6.1 / 2.6.27) fungal Microbiological 2.6.27 7 days contamination Incl. Mycoplasma Control 2.6.12 mycoplasma Mycoplasma 2.6.7 (NAT 1 day) pyrogen BET / MAT / Pyrogen 2.6.14 2.6.30 2.6.8 (1-2 days) No applications for rapid microbiological methods were submitted to PEI recently Is the situation satisfactory? 4

Compendial methods are adequate only for some cellular and tissue medicinal products Manufacturing Drug product Freezing Shelf Life > 14 d Application in patient Examples: Acellular tissue Hematopoetic Stem Cells 14 d (7 d) Ph. Eur 2.6.1 (2.6.27) Final Result Implementation of Rapid Microbial Methods are optional and up to Manufacturer 5 Frequently compendial methods are suboptimal Substantial manipulation Manufacturing In process control Drug product Shelf Life <<14d Drug Product testing Application in patient Examples: Most ATMPs 14 d (7 d) 2.6.1 (2.6.27) (negative to date) 14 d (7d) 2.6.1 (2.6.27) (final result) => RMM development for the Drug Product characterization is desirable 6

Sometimes compendial methods are inadequate Manufacturing and In process control Drug product Shelf Life <<<7d Product test Application in patient 7-14 d 2.6.27 (2.6.1) (negative to date) Examples: Platelet Concentrates Some ATMPs Quality by Design + donor selection + preventive treatment + strict limit of shelf life => Urgent need for RMM 7 Are there rapid microbiological methods on the market? Detection Name* Properties To replace? Duration metabolic byproducts Fluorescence Bactec/ Bact/Alert Bactometer Growth Direct Culture Automates Microcolony Detection Bioluminescence Miliflex ATP Bioluminescence Flow cytometry Cell component detection Bactiflow FACS MicroCount MicroSeq ID PyroSense Live / Dead Fluorescent staining DNA / RNA Detection Endotoxin Growth qualitative Growth quantitative Growth quantitative Direct detection No clear Live / Dead discrimination 2.6.1 2-7 days 2.6.1 2.6.12 2.6.1 2.6.12 10 hours - 2 days 1-5 days 2.6.12 Hours?? Hours * by respective manufacturers 8

Are there rapid microbiological methods on the market? Detection Name* Properties To replace? Duration metabolic byproducts Fluorescence Bactec/ Bact/Alert Bactometer Growth Direct Culture Automates Microcolony Detection Bioluminescence Miliflex ATP Bioluminescence Flow cytometry Cell component detection Bactiflow FACS MicroCount MicroSeq ID PyroSense Live / Dead Fluorescent staining DNA / RNA Detection Endotoxin Growth qualitative Growth quantitative Growth quantitative Direct detection No clear Live / Dead discrimination 2.6.1 2-10 days 2.6.1 2.6.12 2.6.1 2.6.12 10 hours - 2 days 1-5 days 2.6.12 Hours 2.6.12? 2.6.1?? Hours * by respective manufacturers 9 Problem: direct / component detection methods are not suitable for sterility testing Detection Name* Properties To replace? Duration metabolic byproducts Fluorescence Bactec/ Bact/Alert Bactometer Growth Direct Culture Automates Microcolony Detection Bioluminescence Miliflex ATP Bioluminescence Flow cytometry Cell component detection Bactiflow FACS MicroCount MicroSeq ID PyroSense Live / Dead Fluorescent staining DNA / RNA Detection Endotoxin Growth qualitative Sterility Growth quantitative Growth quantitative Direct detection No clear Live / Dead discrimination 2.6.1 2-10 days 2.6.1 10 hours 2.6.12-2 days 2.6.1 1-5 days 2.6.12 2.6.12 Hours Bioburden 2.6.12? 2.6.1?? Hours => Choose a right method or change product characteristics! * by respective manufacturers 10

Question for Discussion: Do we require sterility for inherently non-sterile cell and tissue medicinal products? Is it not better to accept a certain limit for bioburden? (and exclude objectionable organisms) This would allow broader application of growthindependent methods for microbiological control 11 Rationale for this question Relationship of bacterial species and bacterial load to occurrence and severity of transfusion reactions. <100 CFU/ml (5000 CFU) cuture automates @ time of detection It may be more safe release product with result < 100 CFU/ml by direct method vs negative to date by sterility test => Vigilance is important 12 Jacobs MR Clin Infect Dis. 2008

Why RMM are not preferred to compendial methods? Detection Name* Properties To replace? Duration metabolic byproducts Fluorescence Bactec/ Bact/Alert Bactometer Growth Direct Culture Automates Microcolony Detection Bioluminescence Miliflex ATP Bioluminescence Flow / Solid phase cytometry Cell component detection Bactiflow FACS MicroCount MicroSeq ID PyroSense Live / Dead Fluorescent staining DNA / RNA Detection Endotoxin Growth qualitative Growth quantitative Growth quantitative Direct detection No clear Live / Dead discrimination 2.6.1 2-10 days 2.6.1 2.6.12 2.6.1 2.6.12 10 hours - 2 days 1-5 days 2.6.12 Hours 2.6.12? 2.6.1?? Hours = > Validation Necessary (Ph Eur 5.1.6) * by respective manufacturers 13 Validation accordingly to Ph. Eur 5.1.6 Activity Supplier Normally carried out by User Primary validation + - User Requirement Specification (instrument, application) - + Description of the technique + - Risk benefit analysis - + Design qualification (DQ) - + Installation qualification (IQ) - + Operational qualification (OQ) - + Performance qualification (PQ): - verification of primary validation data given by the supplier; - + - verification for the intended use (e.g. sterility testing, TAMC/TYMC, ); - + - method suitability test - + 14

Criterion Validation criteria in PQ Qualitative test Quantitative test Identification test Accuracy + + + Precision - + - Specificity + + + Detection limit + - - Quantitation limit - + - Linearity - + - Range - + - Robustness + + + Suitability testing + + - Equivalence testing + + => Must be performed by every user even for the same product?! 15 Burden of repeated performance qualification Example 1: - Large contract laboratory - Validation of ATP-based test as a sterility test (7 vs 14 days) - Selection of microorganisms (aerobic, anaerobic, yeast, fungi, spores, slow-growing, fast-growing, mixture, stressed, environmental isolates) - >700 Samples tested alone for equivalence test - 3-4 Person-year spent Example 2: - Big Pharma Co. Validation of single RMM (incl. authorization by regulators) will take 3-5 years and have estimated costs of 1-2 Mil. 16

Most applicants are unable to perform proper validation of rapid microbiological methods > 90% small & medium enterprises, start-ups, research facilities & universities, tissue banks > 80% with less than 50 employees -> limited human & financial resources -> limited regulatory experience 17 Question for Discussion: should PQ be performed once for particular method? Prerequisites: Example: - Same product (matrix) - Appropriate method has been validated (elsewere) - Validation data are available for user - Method is established at user site Microbial control of cornea transplants - same tissue type - same culture media - same detection methods (culture automates as sterility test) Full ( Master ) validation performed by single tissue bank validation plan, protocol and results are provided to other banks reduced validation plan performed by others (method transfer) => changes in 5.1.6 might be necessary 18

How to Regulate such two-stage Validation? Pioneer User RMM method developer Publication Validation Plan Raw Data Statistical Analysis Add-on Product Review by Board: RMM Working Party @ EDQM Regulatory Authority - Assessors - Method experts - Statisticians -?? 19 How to Regulate such two-stage Validation? Publication New User + Method qualification Regulatory Authority Add-on Product compendial method 20

Thank you for the Attention Oliver Karo Holger Lößner Ingo Spreitzer Marcel Prax Marie Anders-Maurer Björn Becker Anja Schneider Philipp Windecker Isabelle Bekeredjian-Ding 21