You are on page 1of 57

GMP Considerations for Biotechnology Products:

How Are These Products Different from Chemical Drugs?

REGULATORY OVERVIEW

Nadine Ritter, Ph.D.


nritter@bcg-usa.com

Ruth Wolff, Ph.D.


rwolff@bcg-usa.com

Biologics Consulting Group, Inc.


www.bcg-usa.com

Biological/Biotechnological vs. Chemical Pharmaceutical Products


Significant Differences In:
9Raw Materials 9Production Processes 9Handling Conditions 9Formulations 9Methods of Analysis 9Physiochemical Characteristics 9Stability Profile 9Storage Conditions 9Expiration Dating 9Specifications

Biological/Biotechnological vs. Chemical Pharmaceutical Products Chemical Products

Protein Products

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)

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) Types or Operational Elements (how to) Annex 1 = Sterile Medicinal Products Annex 2 = Biological Medicinal Products Annex 11 = Computerized Systems Annex 13 = Investigational Medicinal Products Annex 15 = Qualification and Validation Annex 18 = GMP for APIs Annex 19 = Reference and Retention Samples

Annexes 1 19 General Instructions for Specific Product

Directives - Detailed Guidance for Specific CMC Studies

(how to)

Where is it Written?

International Regulatory Documents (www.ich.org)


International Conference on Harmonization: U.S., E.U., Japan (Canada, Australia)
ICH Guidance Document Series Q = Quality, S = Safety, E = Efficacy, M - Multidisciplinary
Quality Series:

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)

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 Q5C = Quality of Biotechnological Products: Stability Testing of

Construct in Cells Used for Production of r-DNA Derived Protein Products

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 Relevant to Designated Biotech Product Types


9 Monoclonal Antibodies 9 Recombinant Proteins 9 Naturally-Derived Products (Allergenic extracts) 9 Vaccine Products (Protein, DNA, Conjugate, etc) 9 Cellular and Tissue-Derived Products 9 Gene Therapy Products 9 Plasma-Derived Biopharmaceuticals 9 Plant-Derived Biopharmaceuticals 9 Synthetic Peptides (Redacted in 2006, but principles still useful)

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

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

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

3.2.S. Drug Substance

1. General Information
1.1 Nomenclature 1.2 Structure 1.3 General Properties

2. Manufacture
2.1 2.2 2.3 2.4 2.5 2.6

3. Characterization 4. Control

Manufacturer Manufacturing Process and Controls Control of Materials Control of Critical Steps and Intermediates Process Validation Process Development

3.1 Structure and Characteristics 3.2 Product and Process Impurities 4.1 4.2 4.3 4.4 4.5 Specifications Analytical Procedures Validation of Analytical Procedures Batch Analyses Justification of Specification

5. Reference Standards

6. Container Closure System 7. Stability

3.2.P. Drug Product


2.1 2.2 2.3 2.4 2.5 2.6

1. Description and Composition 2. Pharmaceutical Development

3. Manufacture
3.1 3.2 3.3 3.4 3.5

Components Drug Product Manufacturing Process Development Container Closure System Microbiological Attributes Compatibility Manufacturer Batch Formula Description of Process and Process Controls Control of Critical Steps and Intermediates Process Validation

4. Control of Excipients 5. Control of Drug Product

4.1 Specifications 4.2 Analytical Procedures 4.3 Validation of Analytical Procedures 4.4 Batch Analyses 4.5 Justification of Specification

6. Reference Standards 7. Container Closure System 8. Stability

3.2.A Appendices A.1 Facilities and Equipment A.2 Adventitious Agents Safety Evaluation A.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)

Biological Products

Virus Therapeutic serum Toxin Antitoxin Vaccine Blood, blood component or derivative Allergenic product Trivalent organic arsenic compound Analogous product

Biological products are


Complex mixtures Manufactured in living systems that are, by their nature, inexact Influenced by method of manufacture Along with the manufacturing system, susceptible to their environment Capable of causing a unique set of pathologies due to their origin

Biologic product development is


Challenging Often convoluted Iterative Continuous Susceptible to emerging issues

Examples

Vaccine biologic Tissue-derived protein drug Plasma fractionation product - biologic Therapeutic protein
Biologic Drug if predicate is tissue-derived

Peptide drug

Monoclonal antibodies

Monoclonal antibody conjugates


Biologic, if radiolabeled Biologic, if a toxin conjugate Drug, if a chemical conjugate

Biologic, analogous to therapeutic serum

Monoclonal antibody diagnostic Biologic, if used in vivo Device, if used ex vivo

Legal Basis
Federal Food, Drug and Cosmetic Act
Drugs include biological products

21CFR 312 21CFR 314 21CFR 211 Submission

Investigational drugs Drugs cGMP format NDA

Legal Basis
Public Health Service Act, Section 351

21CFR 312 Investigational drugs 21CFR 600 Biological products 21CFR 211 cGMP Submission format BLA

Biological products only License facility to manufacture product Permits suspension / revocation of licenses Review of the manufacturing facility is integral to review of the product Description of the facility, equipment, utilities and controls must be provided

Legal Basis
Device

Federal Food, Drug and Cosmetic Act 21CFR 812 Investigational Devices 21CFR 814 Devices 21CFR 820 Device Quality Systems Submission format PMA

GMP Considerations for Biotechnology Products:

How Are These Products Different from Chemical Drugs?

Regulatory Considerations for Follow-on Protein Products

Drug
Innovator company files a 505(b)(1) application as a NDA under 21CFR 314 Generic company files a 505(j) application as an ANDA under both 21CFR 314 and 21CFR 320 A 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

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 product However, 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 included Comparability data in some cases A finding of safety and effectiveness is not the same as a finding of substitutability
Woodcock, J. et al. The FDAs assessment of follow-on protein products: a historical perspective. Nature Reviews Drug Discovery. 6, 437 442 (2007).

Factors
Consistency of manufacturing process Conformance to existing regulations Consistency with reference standards or comparators, including comparative PK and PD data Ability to rely on existing clinical data for approved product

Examples

Human Serum Albumin Glucagon Epoetin alfa Human Growth Hormone (Omnitrope)

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 Act Biologics Price Competition and Innovation Act of 2007 (BPCIA)

Provisions of BPCIA
Compromise position Amends Section 351 of the PHS Act Provides a regulatory pathway for safe and interchangeable follow-on biological products Tacit acknowledgment that it may not be possible to dissociate the product from the process

Approval Process
Applicant must demonstrate no clinically meaningful differences in safety, purity and potency between products
Analytical data Animal testing One 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 comparator Influenced 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).

Interchangeable

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

FDA Guidance

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

GMP Considerations for Biotechnology Products:

How Are These Products Different from Chemical Drugs?

Biotech Manufacturing Systems

Mammalian Systems
Closest to 'original' product Older systems have lower productivity Newer systems with higher productivity often require medium additives Strategies to reduce additive levels in production fermenters can lead to substrate stability issues Need to demonstrate removal of additives through downstream purification

Mammalian Systems
Adventitious agents
Raw materials Cell substrate Design downstream purification to partition

agents from product

Complex product profiles


Require extensive characterization Product related substances or product related

impurities?

Microbial Systems
No glycosylation Good for 'simpler' molecules

Engineering specific addition sites Shorter manufacturing cycle


after refolding

Some interferons and growth factors Antibody fragments

Inclusion body processes can have lowered yields

Fewer adventitious agent issues Potential for greater primary sequence heterogeneity Less complex overall product profiles

Processes are dynamic


Changes can be process-related
Modification of process Increase in scale Change in location

Changes can be method-related

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

Improvement in analytical method Replacement of one or more analytical methods

When evaluating changes


Biological products are complex mixtures Control starts with the cell banks and all raw materials The product is at risk from raw materials, operators, and environment No change can be assumed to be neutral Release criteria alone are insufficient to fully evaluate the impact of changes

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 comparability For each step in manufacturing that is changed, all claims made for that step (eg removal of a specific impurity) must be confirmed

Process Development for Biological Products

Goal of Manufacturing Process


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

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 determined Conditions that promote product integrity should be optimized Conditions that promote product compromise should be minimized

Aspects of Manufacturing

Raw materials Purification outcomes Process studies

Raw Materials
Chemical
Reagents Excipients

Biological
Starting materials Reagents Excipients

Resins Container / Closure Systems

Cell Substrate
Freedom from adventitious agents
Virus Bacteria / Fungi Mycoplasma

Level of endogenous agents

Retrovirus Transforming virus if used Amount of genome remaining Amount of infectious virus produced

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

Cell Substrate
Capable of consistent and stable production under defined conditions Stability on storage Adequacy of banking system
Protein Genome

WCB not required, but encouraged Can 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

Synthetic

Biological / biotechnological

Free of solvents and impurities Free of infectious agents Level 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 product Clear adventitious contaminants Remove process-related impurities Remove product-related impurities Ensure 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

Enrich for Desired Product


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

Clear Endogenous and Adventitious Agents


Viral clearance studies Choose a panel of challenge viruses
Challenge data must be reproducible Changes to the process may necessitate repeat studies on one of more purification steps Reevaluation may be needed when agent risks change
Risks in the raw materials Susceptibility of the cell substrate

Platform technologies
Modular approaches to viral clearance have been accepted for early phase clinical development Justification for applicability must be supplied Lack of consensus whether sufficient to support market applications

Process-related Impurities
Residual from cell culture Residual from cell substrate Residual from purification
DNA Host cell proteins Selective agent Medium additive

Affinity ligand Specialty reagent Contaminants from reagents added late in the

purification process

Product-related impurities
Product aggregates Product degradants Modified product
Deamidated Oxidated Misfolded Unconjugated Truncated Glycosylation variants

GMP Considerations for Biotechnology Products:

How Are These Products Different from Chemical Drugs?

Biotechnology Process Validation

Process Understanding

Define equipment Impact of scale Factorial studies Robustness studies Mixing studies Hold time studies
Process intermediates Process buffers Resin lifetime

Equipment
Amount of product required Ease of scale change with equipment included in the initial plan Some technologies do not scale well, either up for production or down for modeling studies Fermentation is an example
Impeller configuration Gas exchange Nutrient availability

Scale
Want to minimize need to perform process validation at full scale Most process critical studies are performed at small scale Relevance 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 parameters Explore parameter interactions Execute challenge studies for clearance Explore improvements Explore anomalies Refine process parameters

Factorial Studies
Two-factor combinations of parameters
Interactions Criticality

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

Robustness
Push the limits of the process parameters identified through the factorial studies
Edge of failure Proven acceptable range Operating range

Hold Time - Intermediates


Harvest from continuous fermentation Primary isolation Loads / Eluates
Pooling Further processing

Product integrity Bioburden

Hold Time - Buffers


Chemical stability Bioburden levels Interaction with container

Hold Time Resin Lifetime


Product yields Performance claims
Virus Host cell proteins DNA Endotoxin Product-related impurities Leachates from resin?

Bioburden

Process Validation Package


Small scale data establish parameters and

interactions Clinical manufacturing data show that the process runs within specified parameters in a reproducible manner Conformance demonstrated through successive, successful runs protocol contains pre-specified criteria for dropping a run No parameters are listed as for information only all information gathering should have been completed in the factorial and robustness studies Supportive validation studies Submitted in the market application

Process Validation Outcomes


Understanding of process capabilities Support for comparability assessments Initial steps in establishment of the design space for product manufacture In 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

GMP Considerations for Biotechnology Products:

How Are These Products Different from Chemical Drugs?

Characterization and Comparability

Heterogeneity of Biotechnological and Biological Products (ICH Q6B)

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

Differences in Heterogeneity? (ICH Q6B)

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

Differences in Drug Safety Issues

Oral Dosage Route (Chemical)


Not Destroyed with Ingestion

Parenteral Dosage Route (Biotech)


Impurities (e.g. host cell proteins) Contaminants (e.g. microbials) microbials)

Differences in Drug Safety Issues

Oral Dosage Route (Chemical)


Decrease in Active Moiety = Efficacy

Parenteral Dosage Route (Biotech)


Change in Protein Impurities = Safety!!

IMMUNOGENICITY

ICH Q6B (August 1999) Specifications: Test Procedures and Acceptance Criteria for Biotechnological/Biological Products
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. Product-related impurities: Aggregates, truncated forms, other modified forms (deamidated, isomerized, oxidized, etc). Degradation products are considered product-related impurities.

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.

Physiochemical and Biological Methods Frequently used with Well-Characterized Protein Products
METHOD pH (if liquid) Karl Fisher (if lyophilized) Appearance UV Absorbance SDS-PAGE (R/NR) SEC-HPLC RP-HPLC, IEX-HPLC, HIC-HPLC Peptide Mapping Mass Spectrometry Isoelectric Focusing Capillary Electrophoresis Immunoassay/ELISA Ligand Binding Assay In Vitro Bioassay N terminal Sequencing Amino Acid Analysis Product Residuals Process Residuals Monosaccharides Oligosaccharide Sialic Acid Circular Dichroism, FTIR AUC ATTRIBUTE General quality Moisture, integrity General quality Concentration Identity, purity, integrity Identity, purity, integrity Identity, purity, integrity Identity, integrity Identity, integrity Identity, integrity Identity, integrity Identity, integrity Identity, potency, integrity Identity, potency, integrity Identity Identity, concentration Purity Purity Identity Identity Identity Conformation Impurities (Aggregates) TYPICAL USE C, C, C, C, C, C, C, C, C, C, C, C, C, C, C, C, C, C, C, C, C, C C R, R, R, R, R, R, R, R, R, R, R, R, R, R, R R R R R* R* R* S S S S S S S S S S S S S S

[* 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 characterization Isotyping ------Verify IgG isotype Peptide 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 content Oligosaccharide profile -------- Determine oligosaccharides present N-terminal and C-terminal -----Determine proportion of C-terminal lysine forms and content N-terminal truncation and/or blockage Western blotting --------- Heavy and/or light chain related species Analytical ultracentrifugation ------ Detect and characterize aggregates
Matrix-assisted laser-desorption ionization time-of-flight (MALDITOF) mass spectrometry -------- Aggregates, breakdown products, verify mass ESI-MS (intact molecule) ----- Aggregates, breakdown products, verify mass,

nonglycosylated forms, and C-terminal variants

Analysis and Structure Characterization of Monoclonal Antibodies BioProcess International (Feb 2004) Schenerman, Sunday, Kozlowski,Webber,

Gazzano-Santoro, and Mire-Sluis

Physiochemical and Biological Methods Being used with Gene Therapy Viral Vectors

METHOD pH Appearance UV Absorbance Colormetric Protein Assay In Vitro Transgene Expression Process Residuals SDS-PAGE (R/NR) Immunoblot/immunoassay IEX-HPLC (particles) RP-HPLC (protein map) Light Scattering Electron Microscopy

ATTRIBUTE General quality General quality Particle count Protein concentration Genetic Stability, Potency Purity Identity Identity Identity, integrity Identity, integrity Integrity

USE C, C, C, C, C, C, C C C C C C R, R, R, R, R, R S S S S S

Bulk and final container characterization assays


Field flow fractionation multiangle light scattering (FFFMALS) Atomic force microscopy (AFM)

Determine particle number and aggregation state Determine particle number and aggregation state Determine particle number

Transmission electron microscopy (TEM)

Size exclusion chromatography multi-angle light scattering (SEC-MALS) TCID, FFA, plaque, or other assays Polymerase chain reaction (PCR) Density gradient centrifugation

Determine particle number and aggregation state

Determine proportion of defective particles based on the difference between total particles and infectious particles Determine proportion of nucleic acid containing particles 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

Schenerman, BioProcess International (April and May, 2006)

Lot Release and Characterization Testing of Live-VirusBased Vaccines and Gene Therapy Products, Gombold, Peden, Gavin, Wei, Baradaran, Mire-Sluis, and

Bulk and final container characterization assays (contd)


Capillary electrophoresis (CE) Reversed-phase HPLC (RPHPLC)

Determine proportion of defective and aggregated particles based on particle mass and charge Determine proportion of defective and aggregated particles based on hydrophobic interaction properties Determine proportion of defective and aggregated particles based on charge state of the particles Determine proportion of defective and aggregated particles based on hydrodynamic sieving properties of particle populations

Ion-exchange chromatography (IEC) Size exclusion chromatography (SEC) SDS-PAGE (or equivalent) Western blot

Determine composition of proteins contained in preparation based on polypeptide chain sizes Determine composition of immunoreactive proteins contained in preparation Quantify process-related impurities

Process residuals (BSA, benzonase, polysorbate, etc.)

Schenerman, BioProcess International (April and May, 2006)

Lot Release and Characterization Testing of Live-VirusBased Vaccines and Gene Therapy Products, Gombold, Peden, Gavin, Wei, Baradaran, Mire-Sluis, and

Peptide Product SpecificationsWhat Parameters are Currently Expected?


Appearance Identity Amino Acid Analysis Purity Potency (Bioassay) Peptide Content Impurity Levels (Individual and Total) Moisture Residual Solvents Counter-Ion Content Microbiological safety

Slide Courtesy D. Lin, BCG

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

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 bridge nonclinical and clinical data generated with pre-change to postchange 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)

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 characterisation studies. 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.

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.

Comparability Studies Utilize Analytical Tests To Answer the Question: Is Batch A Comparable to Batch B? Testing Scheme Outline: Drug Substance Samples
Physiochemical Characteristics ProductProduct-Related Impurities Functional Characteristics ProcessProcess-Related Impurities
Any Differences Might Require In Vivo Studies To Assess Impact Of Changes On PK/PD And Safety

Degraded Drug Substance Samples


Physiochemical Characteristics Functional Characteristics

Product Degradants

Comparability Paradigm Innovator Product

Preclin safety

Clinical safety

Clinical Efficacy

COMPARABILITY STUDIES

Preclin Product Lots

Phase 1 / 2 Product Lots

Phase 3 / Commercial Scale Product Lots

Comparability Paradigm Follow On Biologic (Biosimilar)

Preclin safety

Clinical safety

Clinical Efficacy

COMPARABILITY STUDIES

Preclin Product Lots

Phase 1 / 2 Product Lots

Phase 3 / Commercial Scale Product Lots

Ref Drug

Ref Drug

Ref Drug

Comparison of Major Species in Formulated Drug


Innovator

FOB

Differences In Minor Species?

Some species below LOD of massmass-based protein methods FOB: Impossible to do Direct Comparison at level of API

Current Analytical Expectations for Biotechnology Product and Process Impurities


www.casss.org (look under CMC Strategy Forum publications)
Defining Your Product Profile and Maintaining Control Over It, Part 1: Process-Related Impurities (Simmerman and Donnelly)

BioProcess International June 2005

and Shacter) BioProcess International Sept 2005

Defining Your Product Profile and Maintaining Control Over It, Part 2: Monitoring Host Cell Proteins (Champion, Madden, Dougherty, Defining Your Product Profile and Maintaining Control Over It, Part 3: Product-Related Impurities (Boerner and Clouse) BioProcess Defining Your Product Profile and Maintaining Control Over It, Part 4: Process-Related Impurities - Aggregates (Brorson and Phillips)

International Oct 2005

BioProcess International Nov 2005

GMP Considerations for Biotechnology Products:

How Are These Products Different from Chemical Drugs?

QC Testing: Method Development and Validation for Biotechnology Products

Analytical Test Method TOOL KITS


STABILITY METHODS (S) BATCH RELEASE METHODS (R) CHARACTERIZATION COMPARABILITY METHODS (C)

Analytical Considerations for Biotech Products


No one method is more or less important than another regardless of how simple or complex the technology Specifications are method-dependent different methods will have different levels of sensitivity and specificity Methods developed and validated for bulk substance stability may require re-validation for stability of intermediates, conjugates or formulated product Appropriate internal system suitability measures are critical safeguards for assessing whether variability in test results are related to changes in the method or changes in the product (process)

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 specifications for 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 listed in Compendial monographs. Most new products will not be listed in the Compendia and will therefore require nonnon-compendial analytical procedures for many attributes such as identity, purity/impurities, potency and stability. NonNon-compendial methods require validation per ICH Q2(R1).

Stability-Indicating Method
A procedure that is used to assess the presence/absence of degradants in a product. It is capable of accurately measuring changes in the product that can occur under conditions of physical physical or chemical stress. Methods shown to be stabilitystability-indicating are used in stability protocols to assess the quality of product batches over time under under specified conditions of storage, and to establish the rere-test or expiration dating of products. Methods used in stability protocols must be validated to be stabilitystability-indicating. indicating.

METHOD VALIDATION WHEN?


FDA GFI INDs for Ph 2 and 3 Studies of Drugs, Inc. Specified Therapeutic Biotechnology-Derived Products; CMC Content and Format, Feb 1999 (draft) Phase 1 - A summary of the methods used is provided. 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. ICH Q7A: Good Manufacturing Practice Guidance for Active Pharmaceutical Ingredients, Aug 2001 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.

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) fully validated (e.g. for long-term robustness)

QC test methods used during product development that are not yet

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

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

Qualification versus Validation Studies


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

9 Qualification studies are used to determine method performance


capabilities for parameters such as specificity, linearity, accuracy, precision, etcas required for the intended application.

9 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.

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

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

Qualification versus Validation Studies


Method Validation:
9 Method performance specifications should be established
before starting a validation; validation should not be a discovery study. run for the validation study to pass.

9 These specifications must be met by every validation trial 9 A method can fail validation; if it does, assignable cause for
the failure should be investigated and resolved before the method can be considered fully validated.

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

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

Official Definitions of Validation


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

ICH Q2 (R1)
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

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

Would you want to set pass or fail specifications on these two peaks forever? What factors could impact the reliable performance of this method for this product?

Test Method Validation Study


Experimentally demonstrates that a test method can meet its predetermined specifications for performance of parameters such as: Specificity * Accuracy Precision Linearity/Range LOD/LOQ Robustness *

* NMR The two parameters most often inadequately assessed for the methods ACTUAL intended use.

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] modifications modificationshave been used for product stability testing. These modifications have not been validated or approved by the FDA. FDA.

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 reagents 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 being measured (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

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.

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?


FDA GFI Comparability Protocols, Protein Drug Products and Biological Products (Sept 2003)
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 existing analytical procedure (i.e. retaining the same principle), or to change from one analytical procedure to another.

Making Changes in Analytical Methods?


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

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).

Making Changes in Analytical Methods?


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

When you use the new revised analytical procedure for release or process 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.

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


Compare the current operational capabilities between labs 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

Can the Recipient Lab Meet Method Performance Specifications?


A Tech Transfer Protocol should be clearly designed with predetermined 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?

R&D Methods vs. QC Methods


R&D has the ability to tweak the method into acceptable performance. This may be appropriate to get reliable data while the method is still under development If QC also has to tweak the method, it can be misinterpreted as tweaking the PRODUCT into acceptable performance

Caution to R&D:

Do not transfer methods that require subjective adjustments Do not validate methods that are not optimized for robustness

Caution to QC:

Do not accept methods that are not adequately validated Do not tweak validated methods follow change control

Key CMC Documentation for Analytical Methods:


Method Qualification Protocol, Report, Method SOP (Qualified version) Method Validation Protocol, Report, Method SOP (Validated version) Method Tech Transfer Protocol, Report, Method SOPs (Pre and post transfer versions) Method ReRe-Validation Protocol, Report, Method SOPs (Pre and post rere-validation versions) Comparability/Bridging Protocol, Report, Method SOPs (Old and new method versions)

You might also like