Top Banner
GMP Considerations for Biotechnology Products: How Are These Products Different from Chemical Drugs? Nadine Ritter, Ph.D. Ruth Wolff, Ph.D. [email protected] [email protected] Biologics Consulting Group, Inc. www.bcg-usa.com REGULATORY OVERVIEW Biological/Biotechnological vs. Chemical Pharmaceutical Products 9Raw Materials 9Production Processes 9Handling Conditions 9Formulations 9Methods of Analysis 9Physiochemical Characteristics 9Stability Profile 9Storage Conditions 9Expiration Dating 9Specifications Significant Differences In:
57
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: REGOVRVIEW

GMP Considerations for Biotechnology Products: How Are These Products Different

from Chemical Drugs?

Nadine Ritter, Ph.D. Ruth Wolff, [email protected] [email protected]

Biologics Consulting Group, Inc.www.bcg-usa.com

REGULATORY OVERVIEW

Biological/Biotechnological vs. Chemical Pharmaceutical Products

Raw MaterialsProduction ProcessesHandling ConditionsFormulationsMethods of AnalysisPhysiochemical

CharacteristicsStability ProfileStorage ConditionsExpiration DatingSpecifications

SignificantDifferences In:

Page 2: REGOVRVIEW

Biological/Biotechnological vs. Chemical Pharmaceutical Products

ProteinProteinProductsProducts

ChemicalChemicalProductsProducts

Where is it Written? U.S. Regulatory Documents (www.fda.gov)

Code of Federal Regulation (CFR)U.S. FDA quality practices approved by Congress and enforceable by law (“thou shalt…”).

Guidance for Industry (GFI)Information from FDA on recommended activities to satisfy specific CFR requirements (“how to….”)

Compliance Policy Guidance (CPG)Guidance to FDA reviewers/inspectors on what to look for in product submissions and manufacturing operations (“did they…”)

Page 3: REGOVRVIEW

Where is it Written? EU Regulatory Documents (www.emea.eu.int)

Eudralex Volume 4 - Medicinal Products for Human and Veterinary Use : Good Manufacturing Practice; Commission Directive 2003/94/EC (“thou shalt…”)

Annexes 1 – 19 – General Instructions for Specific Product Types or Operational Elements (“how to…”)

Annex 1 = Sterile Medicinal ProductsAnnex 2 = Biological Medicinal ProductsAnnex 11 = Computerized SystemsAnnex 13 = Investigational Medicinal ProductsAnnex 15 = Qualification and ValidationAnnex 18 = GMP for APIsAnnex 19 = Reference and Retention Samples

Directives - Detailed Guidance for Specific CMC Studies (“how to…”)

Where is it Written? International Regulatory Documents (www.ich.org)

Stability (Q1A, Q1B, Q1C, Q1D, Q1E, Q1F)Test Method Validation (Q2A, Q2B)Product and Process Impurities (Q3A, Q3B, Q3C)Pharmacopoeal Harmonizations (Q4 series)Test Procedures and Acceptance Criteria (Q6A, Q6B)GMP for Active Pharmaceutical Ingredients (Q7)Pharmaceutical Development (Q8, Q8 Annex)Quality Risk Management (Q9)Pharmaceutical Quality Systems (10)

International Conference on Harmonization:U.S., E.U., Japan (Canada, Australia)

ICH Guidance Document SeriesQ = Quality, S = Safety, E = Efficacy, M - Multidisciplinary

Quality Series:

Page 4: REGOVRVIEW

Where is it Written? ICH Q5 Series = Biotech/Biologic Products

Q5A(R1) = Viral Safety Evaluation of Biotechnology Products Derived From Cell Lines of Human or Animal Origin

Q5B = Quality of Biotechnological Products: Analysis of Expression Construct in Cells Used for Production of r-DNA Derived Protein Products

Q5C = Quality of Biotechnological Products: Stability Testing of Biotechnological/Biological Products

Q5D = Derivation and Characterization of Cell Substrates Used for Production of Biotechnological/Biological Products

Q5E = Comparability of Biotechnological/Biological Products Subject to Changes in Their Manufacturing Process

Current Regulatory Guidance Documents Relevantto Designated Biotech Product Types

Monoclonal Antibodies

Recombinant Proteins

Naturally-Derived Products (Allergenic extracts)

Vaccine Products (Protein, DNA, Conjugate, etc…)

Cellular and Tissue-Derived Products

Gene Therapy Products

Plasma-Derived Biopharmaceuticals

Plant-Derived Biopharmaceuticals

Synthetic Peptides (Redacted in 2006, but principles still useful)

Page 5: REGOVRVIEW

ICH M4Q(R1): Content and Format of the Common Technical Document (CTD)

Module 3 outlines details of the specific information Needed on the CMC Elements for Drug Substance (API) and Drug Product

Module 1 = Administrative InformationModule 2 = CTD SummariesModule 3 = Quality (Chemistry, Manufacturing and Controls)Module 4 = Nonclinical StudiesModule 5 = Clinical Studies

3.2.S. Drug Substance1. General Information

1.1 Nomenclature1.2 Structure1.3 General Properties

2. Manufacture2.1 Manufacturer2.2 Manufacturing Process and Controls2.3 Control of Materials2.4 Control of Critical Steps and Intermediates2.5 Process Validation2.6 Process Development

3. Characterization3.1 Structure and Characteristics3.2 Product and Process Impurities

4. Control4.1 Specifications4.2 Analytical Procedures4.3 Validation of Analytical Procedures4.4 Batch Analyses4.5 Justification of Specification

5. Reference Standards6. Container Closure System7. Stability

Page 6: REGOVRVIEW

3.2.P. Drug Product1. Description and Composition2. Pharmaceutical Development

2.1 Components2.2 Drug Product2.3 Manufacturing Process Development2.4 Container Closure System2.5 Microbiological Attributes2.6 Compatibility

3. Manufacture3.1 Manufacturer3.2 Batch Formula3.3 Description of Process and Process Controls3.4 Control of Critical Steps and Intermediates3.5 Process Validation

4. Control of Excipients5. Control of Drug Product

4.1 Specifications4.2 Analytical Procedures4.3 Validation of Analytical Procedures4.4 Batch Analyses4.5 Justification of Specification

6. Reference Standards7. Container Closure System8. Stability

3.2.A AppendicesA.1 Facilities and EquipmentA.2 Adventitious Agents Safety EvaluationA.3 Excipients

3.2.R Regional Information- Executed Batch Records (USA only)- Method Validation Packages (USA only)- Comparability Protocols (USA only)- Process Validation Scheme for Drug Product (EU only)- Medical Device (EU only)

Page 7: REGOVRVIEW

Biological Products

VirusTherapeutic serumToxinAntitoxinVaccineBlood, blood component or derivativeAllergenic productTrivalent organic arsenic compoundAnalogous product

Biological products are …

Complex mixturesManufactured in living systems that are, by their nature, inexactInfluenced by method of manufactureAlong with the manufacturing system, susceptible to their environmentCapable of causing a unique set of pathologies due to their origin

Page 8: REGOVRVIEW

Biologic product development is …

ChallengingOften convolutedIterativeContinuousSusceptible to emerging issues

Examples

Vaccine – biologicTissue-derived protein – drugPlasma fractionation product - biologicTherapeutic protein

BiologicDrug if predicate is tissue-derived

Peptide – drug

Page 9: REGOVRVIEW

Monoclonal antibodiesBiologic, analogous to therapeutic serum

Monoclonal antibody conjugatesBiologic, if radiolabeledBiologic, if a toxin conjugateDrug, if a chemical conjugate

Monoclonal antibody diagnosticBiologic, if used in vivoDevice, if used ex vivo

Legal Basis

Federal Food, Drug and Cosmetic ActDrugs include biological products

21CFR 312 Investigational drugs 21CFR 314 Drugs21CFR 211 cGMPSubmission format – NDA

Page 10: REGOVRVIEW

Legal Basis

Public Health Service Act, Section 351Biological products onlyLicense facility to manufacture productPermits suspension / revocation of licensesReview of the manufacturing facility is integral to review of the productDescription of the facility, equipment, utilities and controls must be provided

21CFR 312 Investigational drugs 21CFR 600 Biological products 21CFR 211 cGMPSubmission format – BLA

Legal Basis

DeviceFederal Food, Drug and Cosmetic Act21CFR 812 Investigational Devices21CFR 814 Devices21CFR 820 Device Quality SystemsSubmission format – PMA

Page 11: REGOVRVIEW

Regulatory Considerations for Follow-on Protein Products

GMP Considerations for Biotechnology Products: How Are These Products Different

from Chemical Drugs?

Drug

Innovator company files a 505(b)(1) application as a NDA under 21CFR 314Generic company files a 505(j) application as an ANDA under both 21CFR 314 and 21CFR 320A ‘paper NDA’ pathway is available under 505(b)(2), relying on data from clinical studies not performed by the applicant and for which no right of reference was given

Page 12: REGOVRVIEW

Biologics

FD&C Act mechanisms are not available for biological products as these mechanisms require ‘sameness’PHS Act does not provide a mechanism for approving a follow-on biologic productHowever, historically, FDA has approved several “follow-on” biological products on a case by case basis

Basis for approvals

‘Similarity’ rather than ‘sameness’Clinical data were includedComparability data in some casesA finding of safety and effectiveness is not the same as a finding of substitutability

Woodcock, J. et al. The FDA’s assessment of follow-on protein products: a historical perspective. Nature Reviews Drug Discovery. 6, 437 – 442 (2007).

Page 13: REGOVRVIEW

Factors

Consistency of manufacturing processConformance to existing regulationsConsistency with reference standards or comparators, including comparative PK and PD dataAbility to rely on existing clinical data for approved product

Examples

Human Serum AlbuminGlucagonEpoetin alfaHuman Growth Hormone (Omnitrope)

Page 14: REGOVRVIEW

Impasse

Pathway charted by Omnitrope is very extensive … but …Omnitrope is a drug and not subject to the provisions of Section 351 of the PHS ActBiologics Price Competition and Innovation Act of 2007 (BPCIA)

Provisions of BPCIA

Compromise positionAmends Section 351 of the PHS ActProvides a regulatory pathway for safe and interchangeable follow-on biological productsTacit acknowledgment that it may not be possible to dissociate the product from the process

Page 15: REGOVRVIEW

Approval Process

Applicant must demonstrate no clinically meaningful differences in safety, purity and potency between products

Analytical dataAnimal testingOne or more clinical studies, unless FDA deems this not necessary

Requirement for new clinical data

For a demonstration of safety and effectiveness, the amount and type will be influenced by the extent to which the follow-on product can be shown to be sufficiently similar to the comparator to rely on the safety and effectiveness of the comparatorInfluenced by clinical use of product, and the amount and type of clinical experience from the comparator and related products

“Follow-on protein products.” Statement of Janet Woodcock, MD, Deputy Commissioner, Chief Medical Officer, FDA before the Committee on oversight and government reform US House of Representatives, 26 March 2007. FDA web site [on line]. http://www.fda.gov/ola/2007/proteins32607.html (2007).

Page 16: REGOVRVIEW

For a designation of interchangeable, applicant must provide evidence that, in any given patient, the follow-on product yields the same clinical result as the comparator and that it presents no risk to safety or efficacy if the patient alternates or is switched between products

“Interchangeable”

FDA may, but is not required to, issue guidance documents with respect to standards and criteria that will be applied to follow-on productsApplications may be submitted prior to issuance of any guidance documents

FDA Guidance

Page 17: REGOVRVIEW

Biotech Manufacturing Systems

GMP Considerations for Biotechnology Products: How Are These Products Different

from Chemical Drugs?

Mammalian Systems

Closest to 'original' productOlder systems have lower productivityNewer systems with higher productivity often require medium additivesStrategies to reduce additive levels in production fermenters can lead to substrate stability issuesNeed to demonstrate removal of additives through downstream purification

Page 18: REGOVRVIEW

Mammalian Systems

Adventitious agentsRaw materialsCell substrateDesign downstream purification to partition agents from product

Complex product profilesRequire extensive characterizationProduct related substances or product related impurities?

Microbial Systems

No glycosylationGood for 'simpler' molecules

Some interferons and growth factorsAntibody fragments

Engineering specific addition sites Shorter manufacturing cycle

Inclusion body processes can have lowered yields after refolding

Fewer adventitious agent issuesPotential for greater primary sequence heterogeneityLess complex overall product profiles

Page 19: REGOVRVIEW

Processes are dynamic

Changes can be process-relatedModification of processIncrease in scaleChange in location

Changes can be method-relatedImprovement in analytical methodReplacement of one or more analytical methods

To rely on pre-clinical and clinical data, must demonstrate that comparable product has been used throughout the development program

When evaluating changes …

Biological products are complex mixturesControl starts with the cell banks and all raw materialsThe product is at risk from raw materials, operators, and environmentNo change can be assumed to be neutralRelease criteria alone are insufficient to fully evaluate the impact of changes

Page 20: REGOVRVIEW

Guiding Principles

The earlier in the manufacturing process the change is made, the greater the potential for impact of the change, and the greater the body of data required to demonstrate comparabilityFor each step in manufacturing that is changed, all claims made for that step (eg removal of a specific impurity) must be confirmed

Goal of Manufacturing Process

To produce sufficient amounts of quality product in a controlled and reproducible manner

Process Development forBiological Products

Page 21: REGOVRVIEW

To accomplish the goal …

The manufacturing process should be thoroughly investigated and understood

Quality should be designed into the product via the process, rather than tested into the product via the analytical methods

Translation …

The contribution of each step of the manufacturing process to the quality of the product should be determinedConditions that promote product integrity should be optimizedConditions that promote product compromise should be minimized

Page 22: REGOVRVIEW

Aspects of Manufacturing

Raw materialsPurification outcomesProcess studies

Raw Materials

ChemicalReagentsExcipients

BiologicalStarting materialsReagentsExcipients

ResinsContainer / Closure Systems

Page 23: REGOVRVIEW

Cell SubstrateFreedom from adventitious agents

VirusBacteria / FungiMycoplasma

Level of endogenous agentsRetrovirusTransforming virus if used

Amount of genome remainingAmount of infectious virus produced

ICH Q5A: Guidance on Viral Safety Evaluation of Biotechnological Products Derived from Cell Lines of Human or Animal OriginICH Q5D: Guidance on Quality of Biotechnological / Biological Products: Derivation and Characterization of Cell Substrates Used for the Production of Biotechnological / Biological Products

Cell SubstrateCapable of consistent and stable production under defined conditions

ProteinGenome

Stability on storageAdequacy of banking system

WCB not required, but encouragedCan manufacture early clinical material from MCB; generate WCB and switch later

ICH Q5B: Quality of Biotechnological Products: Analysis of the Expression Construct in Cells Used for the Production of r-DNA derived Protein Products

ICH Q5D: Guidance of Quality of Biotechnological / Biological Products: Derivation and Characterization of Cell Substrates Used for the Production of Biotechnological / Biological Products

Page 24: REGOVRVIEW

SyntheticFree of solvents and impurities

Biological / biotechnologicalFree of infectious agentsLevel of impurities understood

The further down the manufacturing process the step in which the raw material is added, the ‘cleaner’ the material must be, as there is often little to no capability to ‘clean up’ the product in the remaining process steps.

Purification Outcomes

Enrich for desired productClear adventitious contaminantsRemove process-related impuritiesRemove product-related impuritiesEnsure that product meets specification

ICH Q5A: Guidance on Viral Safety Evaluation of Biotechnological Products Derived from Cell Lines of Human or Animal Origin

ICH Q6B: Specifications: Test Procedures and Acceptance Criteria for Biotechnological / Biological Products

Page 25: REGOVRVIEW

Enrich for Desired Product

Desired product may be a mixture of product related substancesDefinition of the desired product is an analytical oneDefinition of the desired product may change with increased understanding of the product and its behavior in vivo

Clear Endogenous and Adventitious Agents

Viral clearance studiesChoose a panel of challenge viruses

Risks in the raw materialsSusceptibility of the cell substrate

Challenge data must be reproducibleChanges to the process may necessitate repeat studies on one of more purification stepsReevaluation may be needed when agent risks change

Page 26: REGOVRVIEW

Platform technologies

Modular approaches to viral clearance have been accepted for early phase clinical developmentJustification for applicability must be suppliedLack of consensus whether sufficient to support market applications

Process-related Impurities

Residual from cell cultureSelective agentMedium additive

Residual from cell substrateDNAHost cell proteins

Residual from purificationAffinity ligandSpecialty reagentContaminants from reagents added late in the purification process

Page 27: REGOVRVIEW

Product-related impurities

Product aggregatesProduct degradantsModified product

DeamidatedOxidatedMisfoldedUnconjugatedTruncatedGlycosylation variants

Biotechnology Process Validation

GMP Considerations for Biotechnology Products: How Are These Products Different

from Chemical Drugs?

Page 28: REGOVRVIEW

Process Understanding

Define equipmentImpact of scaleFactorial studiesRobustness studiesMixing studiesHold time studies

Process intermediatesProcess buffersResin lifetime

Equipment

Amount of product requiredEase of scale change with equipment included in the initial planSome technologies do not scale well, either up for production or down for modeling studiesFermentation is an example

Impeller configurationGas exchangeNutrient availability

Page 29: REGOVRVIEW

Scale

Want to minimize need to perform process validation at full scaleMost process critical studies are performed at small scaleRelevance to production scale is absolutely dependent on the accuracy of reproduction of the process when run at full scale

Visit small scale often …

Define process parametersExplore parameter interactionsExecute challenge studies for clearanceExplore improvementsExplore anomaliesRefine process parameters

Page 30: REGOVRVIEW

Factorial Studies

Two-factor combinations of parametersInteractionsCriticality

In some cases, may need to assess the interactions of three parametersThe more complex the product, the more convoluted the studies

Robustness

Push the limits of the process parameters identified through the factorial studies

Edge of failureProven acceptable rangeOperating range

Page 31: REGOVRVIEW

Hold Time - Intermediates

Harvest from continuous fermentationPrimary isolationLoads / Eluates

PoolingFurther processing

Product integrityBioburden

Hold Time - Buffers

Chemical stabilityBioburden levelsInteraction with container

Page 32: REGOVRVIEW

Hold Time – Resin Lifetime

Product yieldsPerformance claims

VirusHost cell proteinsDNAEndotoxinProduct-related impuritiesLeachates from resin?

Bioburden

Process Validation PackageSmall scale data establish parameters and interactionsClinical manufacturing data show that the process runs within specified parameters in a reproducible mannerConformance demonstrated through successive, successful runs – protocol contains pre-specified criteria for dropping a runNo parameters are listed as ‘for information only’ – all information gathering should have been completed in the factorial and robustness studiesSupportive validation studiesSubmitted in the market application

Page 33: REGOVRVIEW

Process Validation Outcomes

Understanding of process capabilitiesSupport for comparability assessmentsInitial steps in establishment of the design space for product manufactureIn combination with thorough knowledge of product attributes and the links to process parameters, may be able to submit an Expanded Change Protocol.

Know your process …

… know your product

To be able to validate the process, the analytical methods for routine tests need to be validated (qualified for all other tests)To know which tests to perform, the product needs to be fully characterized

Page 34: REGOVRVIEW

Characterization and Comparability

GMP Considerations for Biotechnology Products: How Are These Products Different

from Chemical Drugs?

Heterogeneity of Biotechnological and Biological Products (ICH Q6B)

The manufacturer should The manufacturer should define the patterndefine the pattern of heterogeneity of heterogeneity of the desired product and demonstrate consistency with of the desired product and demonstrate consistency with that of the lots used in preclinical and clinical studies. that of the lots used in preclinical and clinical studies.

Page 35: REGOVRVIEW

Differences in Heterogeneity? (ICH Q6B)

When process changes and degradation products result in When process changes and degradation products result in heterogeneity patterns that heterogeneity patterns that differdiffer from those observed in the from those observed in the material used during material used during preclinical and clinical developmentpreclinical and clinical development, the , the significance of these alterations should be evaluated.significance of these alterations should be evaluated.

Differences in Drug Safety Issues

Parenteral Dosage Route (Biotech)Parenteral Dosage Route (Biotech)Impurities (e.g. host cell proteins)Impurities (e.g. host cell proteins)Contaminants (e.g. Contaminants (e.g. microbialsmicrobials))

Oral Dosage Route (Chemical)Oral Dosage Route (Chemical)Not Destroyed with IngestionNot Destroyed with Ingestion

Page 36: REGOVRVIEW

Differences in Drug Safety Issues

Oral Dosage Route (Chemical)Oral Dosage Route (Chemical)Decrease in Active Moiety = EfficacyDecrease in Active Moiety = Efficacy

Parenteral Dosage Route (Biotech)Parenteral Dosage Route (Biotech)

IMMUNOGENICITY

Change in Protein Impurities = Safety!!Change in Protein Impurities = Safety!!

ICH Q6B (August 1999)Specifications: Test Procedures and AcceptanceCriteria for Biotechnological/Biological Products

Process-related impurities: Host cell proteins, host cell nucleic acids, cell culture derived components (e.g. serum, antibiotics), downstream process additives (e.g. enzymes, reducing agents, ligands). Process impurities are often treated as product residuals.

Product-related impurities: Aggregates, truncated forms, other modified forms (deamidated, isomerized, oxidized, etc…). Degradation products are considered product-related impurities.

Product-related substances: Molecular variants of the desired product formed during manufacture and/or storage which are active and have no deleterious effects on the safety and efficacy of the drug product. These variants possess properties comparable to the desired product and are not considered impurities.

Page 37: REGOVRVIEW

Physiochemical and Biological Methods Frequently used with Well-Characterized Protein Products

pH (if liquid)Karl Fisher (if lyophilized)AppearanceUV AbsorbanceSDS-PAGE (R/NR)SEC-HPLCRP-HPLC, IEX-HPLC, HIC-HPLCPeptide MappingMass SpectrometryIsoelectric FocusingCapillary ElectrophoresisImmunoassay/ELISALigand Binding AssayIn Vitro BioassayN terminal SequencingAmino Acid AnalysisProduct ResidualsProcess ResidualsMonosaccharidesOligosaccharideSialic AcidCircular Dichroism, FTIRAUC

General qualityMoisture, integrityGeneral qualityConcentrationIdentity, purity, integrityIdentity, purity, integrityIdentity, purity, integrityIdentity, integrity Identity, integrityIdentity, integrityIdentity, integrityIdentity, integrityIdentity, potency, integrityIdentity, potency, integrityIdentityIdentity, concentrationPurityPurityIdentityIdentityIdentityConformationImpurities (Aggregates)

C, R, SC, R, SC, R, SC, R, SC, R, SC, R, SC, R, SC, R, SC, R, SC, R, SC, R, SC, R, SC, R, SC, R, SC, RC, RC, RC, RC, R*C, R*C, R*CC

METHOD TYPICAL USEATTRIBUTE

[* only in specific instances for selected glycoproteins]

Test methods for drug substance characterization (supplementary tests that should be performed in addition to lot-release testing)

ADCC, CDC, neutralization, etc. ------ Potency characterizationIsotyping ------Verify IgG isotypePeptide mapping with electrospray ionization mass spectrometry (ESI-MS) -------

Truncation, deamidation, oxidation, phosphorylation, substitution, alterations in oligosaccharides, detection incorrect sequence

Focused peptide map ---Detect specific product-related impurity/substance (e.g., oxidation)

Carbohydrate composition ----- Determine monosaccharide and sialic acid contentOligosaccharide profile -------- Determine oligosaccharides presentN-terminal and C-terminal -----Determine proportion of C-terminal lysine forms and

content N-terminal truncation and/or blockageWestern blotting --------- Heavy and/or light chain related speciesAnalytical ultracentrifugation ------ Detect and characterize aggregatesMatrix-assisted laser-desorption ionization time-of-flight (MALDITOF) mass spectrometry -------- Aggregates, breakdown products, verify massESI-MS (intact molecule) ----- Aggregates, breakdown products, verify mass,

nonglycosylated forms, and C-terminal variants

Analysis and Structure Characterization of Monoclonal AntibodiesBioProcess International (Feb 2004) Schenerman, Sunday, Kozlowski,Webber, Gazzano-Santoro, and Mire-Sluis

Page 38: REGOVRVIEW

Physiochemical and Biological Methods Being used with Gene Therapy Viral Vectors

pHAppearanceUV Absorbance Colormetric Protein AssayIn Vitro Transgene ExpressionProcess ResidualsSDS-PAGE (R/NR)Immunoblot/immunoassayIEX-HPLC (particles)RP-HPLC (protein map)Light ScatteringElectron Microscopy

General qualityGeneral qualityParticle count Protein concentrationGenetic Stability, PotencyPurityIdentityIdentityIdentity, integrityIdentity, integrityIntegrity

C, R, SC, R, SC, R, SC, R, SC, R, SC, RCCCCCC

METHOD ATTRIBUTE USE

Bulk and final container characterization assays

Field flow fractionation multiangle light scattering (FFF–MALS) Determine particle number and aggregation state

Atomic force microscopy (AFM) Determine particle number and aggregation state

Transmission electron microscopy (TEM) Determine particle number

Size exclusion chromatography multi-angle light scattering (SEC-MALS) Determine particle number and aggregation state

TCID, FFA, plaque, or other assays Determine proportion of defective particles based on the difference between total particles and infectious particles

Polymerase chain reaction (PCR) Determine proportion of nucleic acid containing particles

Density gradient centrifugation Determine proportion of defective particles based on relative densities of particle populations

Analytical ultracentrifugation (AUC) Determine proportion of defective and aggregated particles based on hydrodynamic properties of particle populations

Lot Release and Characterization Testing of Live-Virus–Based Vaccines and Gene Therapy Products, Gombold, Peden, Gavin, Wei, Baradaran, Mire-Sluis, and Schenerman, BioProcess International (April and May, 2006)

Page 39: REGOVRVIEW

Bulk and final container characterization assays (cont’d)

Capillary electrophoresis (CE) Determine proportion of defective and aggregated particles based on particle mass and charge

Reversed-phase HPLC (RPHPLC) Determine proportion of defective and aggregated particles based on hydrophobic interaction properties

Ion-exchange chromatography (IEC) Determine proportion of defective and aggregated particles based on charge state of the particles

Size exclusion chromatography (SEC)Determine proportion of defective and aggregated particles based on

hydrodynamic sieving properties of particle populationsSDS-PAGE (or equivalent)

Determine composition of proteins contained in preparation based on polypeptide chain sizes

Western blot Determine composition of immunoreactive proteins contained in preparation

Process residuals (BSA, benzonase, polysorbate, etc.) Quantify process-related impurities

Lot Release and Characterization Testing of Live-Virus–Based Vaccines and Gene Therapy Products, Gombold, Peden, Gavin, Wei, Baradaran, Mire-Sluis, and Schenerman, BioProcess International (April and May, 2006)

Peptide Product Specifications-What Parameters are Currently Expected?

AppearanceIdentityAmino Acid AnalysisPurityPotency (Bioassay)Peptide ContentImpurity Levels (Individual and Total)MoistureResidual SolventsCounter-Ion ContentMicrobiological safety

Slide Courtesy D. Lin, BCG

Page 40: REGOVRVIEW

During product development, it is expected that multiple changes in the manufacturing process will occur that could impact drug product quality, safety, and efficacy.

Comparability exercises are generally performed to bridgenonclinical and clinical data generated with pre-change to post-change product in order to facilitate further development and, ultimately, to support the marketing authorisation.

ICH Q5E: Comparability of Biotechnological/Biological Products Subject to Changes in Their Manufacturing Processes (2003)

ICH Q5E: Comparability of Biotechnological/Biological Products Subject to Changes in Their Manufacturing Processes (2003)

Any change with the potential to alter protein structure or purity and impurity profiles should be evaluated for its impact on stability, since proteins are frequently sensitive to changes, such as those to buffer composition, processing and holding conditions, and use of organic solvents.

Furthermore, stability studies might be able to detect subtle differences that are not readily detectable by the characterisationstudies.

Accelerated and stress stability studies are often useful tools to establish degradation profiles and provide a further direct comparison of pre-change and post-change products.

Page 41: REGOVRVIEW

Side-by-Side Comparability Studies –SAVE All Product Batch Retains

Pre-clinical and clinical lots are irreplaceable material – once they are gone, they are gone

Conditions of storage may not maintain perfect stability of the retains, but it is better to have slightly degraded retains than no retains at all

Degradants can be distinguished from heterogeneous impurities via side-by-side stress degradation studies (a key aspect of comparability studies)

Retains are also invaluable for demonstrating the suitability of new analytical methods as compared to older methods – they can prove or disprove the need to modify product specifications when analytical methods become more sensitive or more specific for product impurities.

PhysiochemicalPhysiochemicalCharacteristicsCharacteristics

ProductProduct--RelatedRelatedImpuritiesImpurities

ProcessProcess--RelatedRelatedImpuritiesImpurities

FunctionalFunctionalCharacteristicsCharacteristics

Any DifferencesAny DifferencesMight RequireMight RequireIn Vivo StudiesIn Vivo Studies

To Assess ImpactTo Assess ImpactOf ChangesOf ChangesOn PK/PDOn PK/PD

And SafetyAnd Safety

Comparability Studies Utilize Analytical Tests To Answer the Question: “Is Batch A Comparable to Batch B?”

Testing Scheme Outline:

PhysiochemicalPhysiochemicalCharacteristicsCharacteristics

Product Product DegradantsDegradants

FunctionalFunctionalCharacteristicsCharacteristics

Drug Substance Samples

Degraded Drug Substance Samples

Page 42: REGOVRVIEW

Comparability Paradigm – Innovator Product

PreclinPreclinProductProductLotsLots

Phase 1 / 2Phase 1 / 2ProductProductLotsLots

PreclinPreclinsafetysafety

ClinicalClinicalsafetysafety

ClinicalClinicalEfficacyEfficacy

COMPARABILITY STUDIES

Phase 3 /Phase 3 /Commercial ScaleCommercial Scale

ProductProductLotsLots

Comparability Paradigm – Follow On Biologic (Biosimilar)

PreclinPreclinProductProductLotsLots

Phase 1 / 2Phase 1 / 2ProductProductLotsLots

PreclinPreclinsafetysafety

ClinicalClinicalsafetysafety

ClinicalEfficacy

COMPARABILITY STUDIES

Phase 3 /Phase 3 /Commercial ScaleCommercial Scale

ProductProductLotsLots

RefDrug

RefDrug

RefDrug

Page 43: REGOVRVIEW

Comparison of Major Species in Formulated Drug

InnovatorInnovator

FOBFOB

DifferencesDifferencesIn MinorIn MinorSpecies?Species?

FOB: Impossible to FOB: Impossible to do Direct do Direct

ComparisonComparisonat level of APIat level of API

Some species below Some species below LOD of massLOD of mass--basedbased

protein methodsprotein methods

Page 44: REGOVRVIEW

Current Analytical Expectations for Biotechnology Product and Process Impurities

Defining Your Product Profile and Maintaining Control Over It, Part 3: Product-Related Impurities (Boerner and Clouse) BioProcess International Oct 2005

Defining Your Product Profile and Maintaining Control Over It, Part 4: Process-Related Impurities - Aggregates (Brorson and Phillips) BioProcess International Nov 2005

Defining Your Product Profile and Maintaining Control Over It, Part 2: Monitoring Host Cell Proteins (Champion, Madden, Dougherty, and Shacter) BioProcess International Sept 2005

Defining Your Product Profile and Maintaining Control Over It, Part 1: Process-Related Impurities (Simmerman and Donnelly) BioProcess International June 2005

www.casss.org (look under CMC Strategy Forum publications)

QC Testing: Method Development and Validation for Biotechnology Products

GMP Considerations for Biotechnology Products: How Are These Products Different

from Chemical Drugs?

Page 45: REGOVRVIEW

Analytical Test Method“TOOL KITS”

STABILITY METHODSSTABILITY METHODS(S)(S)

BATCH RELEASE METHODSBATCH RELEASE METHODS(R)(R)

CHARACTERIZATION CHARACTERIZATION COMPARABILITY METHODSCOMPARABILITY METHODS

(C)(C)

No one method is more or less important than another – regardlessof how simple or complex the technology

Specifications are method-dependent – different methods will havedifferent levels of sensitivity and specificity

Methods developed and validated for bulk substance stability mayrequire re-validation for stability of intermediates, conjugates or formulated product

Analytical Considerations for Biotech Products

Appropriate internal system suitability measures are critical safeguards for assessing whether variability in test results arerelated to changes in the method or changes in the product (process)

Page 46: REGOVRVIEW

Compendial Methods

An analytical method published in a Pharmacopoeia.

General Chapters = overview of issues relevant to the production and testing of pharmaceutical products

Methods = general procedures for chemical, physical, biological, microbiological, immunological methods

Monographs = product-specific specificationsfor release, label claims, and storage conditions

Many ‘old’ biological products (vaccines, plasma products, natural products (e.g. porcine insulin) and ‘herbals’ have monographs

Few ‘new’ biotechnology products (WCBP, conjugate vaccines) have monographs (yet…).

Non-Compendial Method

A procedure that is used to test products that are not A procedure that is used to test products that are not listed in listed in CompendialCompendial monographs. monographs.

Most new products will not be listed in the Most new products will not be listed in the CompendiaCompendia and and will therefore require nonwill therefore require non--compendial analytical compendial analytical procedures for many attributes such as identity, procedures for many attributes such as identity, purity/impurities, potency and stability. purity/impurities, potency and stability.

NonNon--compendial methods require validation per ICH compendial methods require validation per ICH Q2(R1).Q2(R1).

Page 47: REGOVRVIEW

Stability-Indicating Method

A procedure that is used to assess the presence/absence of A procedure that is used to assess the presence/absence of degradants in a product. It is capable of accurately measuring degradants in a product. It is capable of accurately measuring changes in the product that can occur under conditions of physicchanges in the product that can occur under conditions of physical al or chemical stress. or chemical stress.

Methods shown to be stabilityMethods shown to be stability--indicating are used in stability indicating are used in stability protocols to assess the quality of product batches over time undprotocols to assess the quality of product batches over time under er specified conditions of storage, and to establish the respecified conditions of storage, and to establish the re--test or test or expiration dating of products.expiration dating of products.

Methods used in stability protocols must be Methods used in stability protocols must be validated to be validated to be stabilitystability--indicatingindicating..

METHOD VALIDATION – WHEN?

Phase 2 - A complete description of the analytical procedure and supporting validation data should be available on request.

Phase 3 - A complete description of the non-USP analytical procedures with appropriate validation information should be provided.

Phase 1 - A summary of the methods used is provided.

FDA GFI INDs for Ph 2 and 3 Studies of Drugs, Inc. Specified Therapeutic Biotechnology-Derived Products; CMC Content and Format, Feb 1999 (draft)

APIs for use in Clinical Trials While analytical methods performed to evaluate a batch of API for clinical trials may not yet be validated, they should be scientifically sound.

ICH Q7A: Good Manufacturing Practice Guidance for ActivePharmaceutical Ingredients, Aug 2001

Page 48: REGOVRVIEW

“Qualified” Test Methods

Methods used ONLY for process characterization or process validation (not for routine cGMP manufacturing)

Methods used ONLY for product characterization and comparability (not for routine cGMP release or stability testing)

QC test methods used during product development that are not yetfully validated (e.g. for long-term robustness)

“Method Qualification vs Validation – What is the Difference? CMC Strategy Forum July 2003” Ritter, Simmerman, Advant, McEntire,

Hennessey, Mire-Sluis, Joneckis; BioProcess International Sept 2004

There are no pre-determined method performance specifications; however, there may be minimal method performance capability requirements based on the intended application.

Qualification studies are used to determine method performancecapabilities for parameters such as specificity, linearity, accuracy, precision, etc…as required for the intended application.

A method cannot fail qualification; it can (and should) be re-optimized until it can achieve acceptable performance, or it should be rejected for the application.

Method Qualification:

Qualification versus Validation Studies

“Method Qualification vs Validation – What is the Difference? CMC Strategy Forum July 2003” Ritter, Simmerman, Advant, McEntire,

Hennessey, Mire-Sluis, Joneckis; BioProcess International Sept 2004

Page 49: REGOVRVIEW

“Method Qualification vs Validation – What is the Difference? CMC Strategy Forum July 2003” Ritter, Simmerman, Advant, McEntire,

Hennessey, Mire-Sluis, Joneckis; BioProcess International Sept 2004

Method performance specifications should be established before starting a validation; validation should not be a discovery study.

These specifications must be met by every validation trial run for the validation study to pass.

A method can fail validation; if it does, assignable cause forthe failure should be investigated and resolved before the

method can be considered fully validated.

Method Validation:

Qualification versus Validation Studies

Official Definitions of Validation

“Establishing through documented evidence a high degree of assurance that an analytical method will consistently yield

results that accurately reflect the quality characteristics of the material tested.”

Title 21 Code of Federal Regulations 211.222

“The objective of validation of an analytical procedure is to demonstrate that it is suitable for its intended purpose.”

ICH Q2 (R1)

Page 50: REGOVRVIEW

What factorscould impact the

reliable performance of this method for

this product?

Could any analyst run this assay on any day, and get the same results?

Would you want toset pass or fail

specifications onthese two peaks forever?

Test Method Validation Study

Experimentally demonstrates that a test method can meet its predetermined specifications for performance of parameters such as:

Specificity *Accuracy PrecisionLinearity/RangeLOD/LOQRobustness *

* NMR – The two parameters most often inadequately assessedfor the methods ACTUAL intended use.

Page 51: REGOVRVIEW

Check Out Existing Method Validations

-Verify that the methods currently in place are validated for their intended use

- Review the validation packages (protocols/reports) to assess if the studies were conducted in alignment with current ICH expectations

- Determine if there have been any changes made to the method following validation

- Confirm if the changes required re-validation of the method

FDA Warning Letter on Changes Made in Stability Methods

““Various [method] modificationsVarious [method] modifications……have have been used for product stability testing. These been used for product stability testing. These

modifications have not been modifications have not been validated or approved by the FDA.validated or approved by the FDA.””

Page 52: REGOVRVIEW

Method Re-Validation – When and How?

Changes to the product that could affect method performance(e.g. formulation excipients, product concentration)

Changes in critical assay reagentsreagents that cannot meet prior specifications (e.g. gel or blot materials, enzymes)

Changes in instrumentation that cannot perform equivalent procedures (e.g. AAA hydrolysis systems)

Changes in the procedure to improve robustness of the method (e.g. new qualification procedures for critical reagents, new sample preparation steps)

Changes in the specifications for the product parameter beingmeasured (e.g. increased specificity or sensitivity for impurities)

After the Validation: Monitoring Method Performance

Adequately validated test methods should be able to consistently meet their intended use.

Validated test methods should be capable of performing within specifications, demonstrating a true state of control.

Methods that cannot perform to their validated specifications may require re-optimization and re-validation.

FDA Guidance for Industry: Investigating Out of Specification Test Results for Pharmaceutical Production, CDER, September 2006

Page 53: REGOVRVIEW

Hidden Sources of Variability in Biotech Methods:

Assay Materials and Reagents

Potentially “Critical” Assay Reagents for Biotech Methods Have One or More of These Characteristics:

• Complex molecules, often biologically derived• Demonstrated to be a key assay component• Sensitive to operational or assay conditions• Selected characteristics may vary from lot to lot• Limited concurrent availability of multiple lots• Single-source product manufacturer

Ritter, N and Wiebe, M (2001) Validating Critical Reagents Used in cGMP Analytical Testing, BioPharm 14:5, pp 12-20.

Potentially Critical Assay Components

HPLC - columns (resins and packing procedures), unique mobile phase components

Capillary electrophoresis - capillaries, electrode buffers, prepared kit components

Gel electrophoresis - gel matrix components, unique buffers, precast gels, stains, dyes

Immunoassays - immunoreagents, detection agents, unique blocking materials

Peptide maps - reduction/alkylation reagents, digestion enzymes, HPLC columns

Colormetric methods - commercial standards, chromogenic agents, prepared assay kits

Amino acid analysis - hydrolysis reagents, derivatization reagents

Protein sequencing - coupling, cleavage and conversion reagents; de-blocking enzymes

Bioactivity assays - substrates, cofactors, ligands, cell cultures, media components

Sample preparation - unique buffer components, filters, membranes, culture plates, vials and stoppers

Ritter, N and Wiebe, M (2001) BioPharm 14:5, pp 12-20.

Page 54: REGOVRVIEW

Tracking/Trending Test Lab Operations

Management should ascertain the significance that OOS results represent in the overall quality assurance program.

Management should be especially alert to developing trends.

FDA Guidance for Industry: Investigating Out of Specification Test Results for Pharmaceutical Production, CDER, September 2006

Making Changes in Analytical Methods?

“A comparability protocol for changing an analytical procedure should provide the plan for validation of the changed analytical procedure and indicate whether the protocol will be used to modify the existinganalytical procedure (i.e. retaining the same principle), or to changefrom one analytical procedure to another.”

FDA GFI Comparability Protocols, Protein Drug Products and Biological Products (Sept 2003)

Page 55: REGOVRVIEW

Making Changes in Analytical Methods?

“We recommend that you design the comparability protocol to demonstrate that the proposed changes in the analytical procedures improve or do not significantly change analytical procedure characteristics that are relevant to the type of analytical procedure, its validation, and intended use (e.g., accuracy, precision, specificity, detection limit, quantitation limit, linearity, range).”

FDA GFI Comparability Protocols, Protein Drug Products and Biological Products (Sept 2003)

Making Changes in Analytical Methods?

“When you use the new revised analytical procedure for release orprocess control, you should not delete a test or relax acceptance criteria that we approved in your application, unless and until FDA informs you that the approved acceptance criteria are no longer required.”

FDA GFI Comparability Protocols, Protein Drug Products and Biological Products (Sept 2003)

Page 56: REGOVRVIEW

Assay Transfer Protocol -R&D to QC, Site to Site, Sponsor to CMO

Exact replication of validated method with identical systems? Transfer protocol could be similar to a repeatability study

Implementation of validated method with similar systems?

Transfer protocol should contain repeat of key parameters

Adaptation of validated method to new systems?

Transfer protocol might have to be re-validation protocol

Compare the current operational capabilities between labs

Can the Recipient Lab Meet Method Performance Specifications?

A Tech Transfer Protocol should be clearly designed with pre-determined criteria for confirming a successful transfer:

• Number of replicates within each run• Total number of runs (analysts, instruments, days)• Number of product lots to be used in each run• Expected correlation of test results between labs

Transfer of qualified vs validated methods:How much do you know about robustness parameters?

Page 57: REGOVRVIEW

R&D Methods vs. QC Methods

R&D has the ability to tweak the method into acceptable R&D has the ability to tweak the method into acceptable performance. This may be appropriate to get reliable data performance. This may be appropriate to get reliable data while the method is still under developmentwhile the method is still under development

If QC also has to tweak the method, it can be misinterpreted If QC also has to tweak the method, it can be misinterpreted as tweaking the PRODUCT into acceptable performanceas tweaking the PRODUCT into acceptable performance

Caution to R&D:Caution to R&D:Do not transfer methods that require subjective adjustmentsDo not transfer methods that require subjective adjustmentsDo not validate methods that are not optimized for robustnessDo not validate methods that are not optimized for robustness

Caution to QC:Caution to QC:Do not accept methods that are not adequately validatedDo not accept methods that are not adequately validatedDo not tweak validated methods Do not tweak validated methods –– follow change controlfollow change control

Key CMC Documentation for Analytical Methods:

•• Method Qualification Protocol, Report, Method SOP Method Qualification Protocol, Report, Method SOP (Qualified version)(Qualified version)

•• Method Validation Protocol, Report, Method SOP Method Validation Protocol, Report, Method SOP (Validated version)(Validated version)

•• Method Tech Transfer Protocol, Report, Method SOPs Method Tech Transfer Protocol, Report, Method SOPs (Pre and post transfer versions)(Pre and post transfer versions)

•• Method ReMethod Re--Validation Protocol, Report, Method SOPs Validation Protocol, Report, Method SOPs (Pre and post re(Pre and post re--validation versions)validation versions)

•• Comparability/Bridging Protocol, Report, Method SOPs Comparability/Bridging Protocol, Report, Method SOPs (Old and new method versions)(Old and new method versions)