Wikipedia

Search results

Wednesday 6 June 2018

Material Control Strategies in Product Development

https://www.pda.org/pda-europe/news-archive/full-story/2015/01/05/raw-material-control-strategy-key-to-overall-control


Section 501 (21 U.S.C. 351) is amended by adding at the end the following flush text: ‘‘For purposes of paragraph (a)(2)(B), the term ‘current good manufacturing practice’ includes the implementation of oversight and controls over the manufacture of drugs to ensure quality, including managing the risk of and establishing the safety of raw materials, materials used in the manufacturing of drugs, and finished drug products.’’.

INTRODUCTION:
Selection of raw materials and excipients to be used in manufacturing process and identification of critical material attributes to define material control strategies at development stage of product life cycle, this includes the following:
·         Discovery research (including all stages of clone development) and
·         Toxicology studies
All users involved in vector development, clone development and process development shall follow the procedure defined below.
Note: As activities of clone development are considered as critical for product lifecycle, a diligent material control with traceability of materials used in each stage shall be demonstrated (Refer ICH Q5B & Q5D and EP 5.2.12).
CONTROL STRATEGY:
MATERIAL CLASSIFICATION:
Biological product development and manufacturing uses diverse range of raw materials. These materials can be categorized as below:
A.   Base on composition materials can be categorized as:
·         Inorganic Salts and liquids,
·         Organic Salts and liquids,
·         Complex chemically defined mixtures of organic/ inorganic origin),
·         Complex materials of biological origin, including animal origin (defined),
·         Complex materials of biological origin, including animal origin (undefined),
·         Complex semi defined mixtures (containing above materials),
Note: In addition to above category there are few fixed materials used in process which though are not categorized as raw materials but are product contact and have tendency of leaching (process) impurities in the product e.g., filters and TFF membranes, chromatography media etc. Criticality of such materials shall be assessed and capture in process development report as part of process impurities.
B.   Based on usage in manufacturing process materials can be categorized as:
·         Product contact materials and
·         Product non-contact materials (such as cleaning agents)
However, there are chances of product contamination through product non-contact materials if cleaning procedures are not validated and may directly impact the product CQAs.
C.   Based material control strategy the materials used in manufacturing process can be classified as under:
·         Biological Starting Materials
·         Raw Materials
·         Drug Substance (Active Pharmaceutical Ingredient)
·         API Starting Material
·         Excipients
Note: Specific requirements for Biological Starting material are outside scope of this document.
MATERIAL CONTROL STRATEGY:
Material control at development stage is limited to first two elements; other elements shall be covered in the subsequent stages of product life cycle.
Elements of material control strategies ensure the safety (including risk of adventitious agent contamination), quality and efficacy of the biological molecule to be manufactured. These are:
1.    Selection of material and understanding the role of the raw materials in the process and criticality assessment of material
2.    Identification of material quality attributes
3.    Developing material testing strategies to ensure raw material quality and evaluating specific tests to ensure consistent material quality
4.    Setting up material/supplier risk assessments for supplier qualification program
5.    Having a supplier qualification system in place which assesses all risks of a raw material: origin risk (safety), supply risk (availability, back-up options, audits, contractual agreements) and quality risk (testing, evaluation of lot-to-lot consistency).
Note: All the elements shall be revisited during initiation of cGMP manufacturing operations such as MCB and WCB preparation, clinical batch manufacturing, process performance qualification (validation) batch manufacturing and commercial manufacturing.
SELECTION OF MATERIAL AND CRITICALITY ASSESSMENT:
Material for each process stage shall have scientific basis for selection and following criteria shall be considered:
1.    Role of material in process stage
2.    Impact of the material on quality attribute of the subsequent stage product
3.    Introduction of impurities in manufacturing process (including adventitious agents).
To ensure material control, user shall perform material risk assessment as per the table below:
Sr.
No
Assessment Question
Answer
(Yes/No)
Risk
(High/Medium/Low)
Comments/
Justification
1
Is the RM complex?

2
Is the RM well defined?
3
Is the material of animal/human origin?
4
Is the TSE/BSE assessment available?
5
Is the RM added in the late steps of the process?
6
Is there a need to demonstrate that the process will reduce the RM level to a safe residual level?
7
Is a relevant analytical method available to assess its clearance?
8
Is the level of quality of RM susceptible to impact product CQA?
9
Is the production process of the RM generating high variability in the RM quality attributes
Remarks: (For additional assessment and comments)
Raw Material categorized as: Critical ¨   /   Non-Critical ¨
Note 1: Excipients are not accessed based on above criteria. Procedure for excipient selection and assessment is provided below.
Note 2: Assessment shall be performed for all raw materials, including chromatography resins and media. In column for ‘Answer’ enter ‘Yes’ or ‘No’, identify the risk associated in each evaluation step and provide comments and justification for each risk categorized (irrespective of low, medium, high) with current and recommended mitigation plan.
From the above assessment a list of critical and non-critical materials will be generated.
FOR CRITICAL MATERIALS: Higher grade material with more control on material attributes shall be selected, preferable to use compendial grade material manufactured under cGMP environment.
FOR NON-CRITICAL MATERIALS: Grade of material is flexible however consistency in supply of material with identified attributes shall be ensured.
SELECTION OF EXCIPIENTS:
Excipients are added for the purpose of production enhancement, patient acceptability, improving stability, controlling release etc. Though termed as inactive components, excipients can have an impact on the absorption, distribution, metabolism and elimination process of the co-administered drug, which is important information when selecting excipient for any formulation.
As manufacturer of the chemicals supply their material for different applications and end users (such as cosmetics, food additives etc.), it is responsibility of the user to select the excipient of appropriate type and grade to meet additional quality functionality and safety requirements.
Below are the basic check points used during selection of excipients:
1.  
Name of the excipient in the formulation:
2.  
Is the excipient complex chemical: (Define physical and chemical properties, material (excipient) specifications including impurities and residual solvent)
3.  
Is the excipient well defined: Define physical and chemical properties, material (excipient) specifications including impurities and residual solvent)
4.  
Is the proposed material (excipient) of animal/human origin:
5.  
Is the production process of the material (excipient ) anticipated to generate high variability in the quality attributes:
6.  
Is the level of quality of material (excipient) susceptible to impact product CQA. List and define critical material attributes (CMA) of the material (excipient):
7.  
Is the TSE/BSE assessment available for the excipient:
8.  
Mention role of material (excipient) in the formulation (such as diluent, buffering compound, antioxidant, stabilizer, enhances solubility, enhances bioavailability etc.):
9.  
Mention intended route of administration of finished product (topical, ophthalmic, oral, parenteral etc.):
10.  
Is the material previously selected as excipient for intended route of delivery:
11.  
Is the proposed material identified as established excipient:
(If yes, in which IPEC category does it fall)
1.    Existing chemical excipient- first in human:
(These are class of excipients where animal safety data does exist, as data may have been used in  another regulatory applications)
2.    Existing chemical excipient:
(These are excipients that have been used in man, but for another route of administration, higher dose etc., and additional safety may be required)
3.    New modification of existing excipient:
(Established excipient is the new modifications of combinations, which would not require safety evaluation)
12.  
Is the excipient approved by any of the regulatory agencies: (Refer different resources and mention wherever listed, such as EMA's Excipients Drafting Group (ExcpDG), FDA Inactive ingredient database, Safety and Toxicity of Excipients for Paediatrics (STEP) Database, listed in GRAS database, FAP database, wherever available DMF File number shall be provided).
13.  
Is an official monograph available in any of Pharmacopeia (USP-NF, EP, JP, IP etc.):
14.  
Is the material identified as new chemical excipient:
15.  
Does the selected vendor follow IPEC (The International Pharmaceutical Excipients Council) cGMP Guidelines:
16.  
Does the selected vendor has ISO 9001 certification:
17.  
Does the selected vendor has EXCiPACT certification:
18.  
Does the selected vendor material meet the designated Pharmacopeial requirements, if yes, mention the pharmacopeia (attach monograph copy):
19.  
Note: Though supply security and quality agreements are not expected during discovery research and toxicology studies, however given the criticality of the excipient and impact of change on regulatory filing, wherever possible below expectations shall be ensured:
·         Manufacturing process for excipient shall be sought from vendor,
·         Understanding with the manufacturer to ensure change notification in manufacturing process and CQA of material
·         Long term supply commitment
Note: Regulatory filing process for new excipients is outside the scope of this document.
User shall compile all the above information with relevant evidences as annexures in single report for excipient selected.
MATERIAL CONTROL STRATEGY
As the scope of this document is limited to discovery research and toxicology studies, material control strategy for these stages is defined below.
Drug Discovery:
·         Material and vendor selection as per study plan (assessment defined in 5.3.2 can be performed and recorded)
·         COA and expiry of all incoming process material documented for use in relevant documents (LNBs, protocols & records etc.)
·         Material (Safety Data Sheet) (MSDS/ SDS) understood and to be made available at site of usage for ready reference along with COA.
·         Storage and expiry as per vendor recommendation.
·         Material for each unit operation stage is fixed based on the development data (assessment defined in 5.3.2 mandatory and shall be compiled in single report with relevant annexures). Specification of all the materials to be listed with test method references wherever available.
Toxicology Studies:
·         Material and vendor selection performed as per 5.5.1 point number v. Refer table in section 5.6 for requirements for each stage.
·         Details of expiry of all incoming process material documented in Batch Manufacturing Record and COA’s attached to it
·         MSDS understood and to be made available at site of usage for ready reference.
·         Storage and expiry as per vendor recommendation.
·         Compilation of material and vendor documentation in a single report.
DOCUMENTATION DURING DEVELOPMENT AND TOXICOLOGY BATCH MANUFACTURING:
Collection of following documents for all materials (where applicable) as below:
Material Documentation Requirements
Drug Discovery
Toxicology Batch
-   Material Name
Ă¼
Ă¼
-   Material grade
Ă¼
Ă¼
-   Vendor Name
Ă¼
Ă¼
-   Vendors Address
Ă¼
Ă¼
-   Vendor Catalogue Number
Ă¼
Ă¼
-   Vendor Specification
Ă¼
Ă¼
-   Vendor COA
Ă¼
Ă¼ (multiple batches)
-   List of potential impurities
Ă¼
Ă¼
-   List of solvents used in process and residual solvents in product, if any
Ă¼
Ă¼
-   MSDS/ SDS
Ă¼
Ă¼
-   Certificate of Material Origin
Ă¼
Ă¼
-   Certificate for TSE/BSE
Ă¼
Ă¼
-   Risk of Viral and other adventitious agents
Ă¼
Ă¼
-   Kosher / HALA certifications
Optional
Ă¼
-   Allergen certificate
Optional
Ă¼
-   Aflatoxin certificates
Optional
Ă¼
-   Certification/ accreditations of vendor
Optional
Ă¼
-   Manufacturing Process Flow
Optional
Ă¼
-   Characterization Data
Optional
Ă¼
-   Toxicological data
Optional
Ă¼
-   Logistic mapping and supply change map
Optional
Ă¼
Note: Material control strategy for clinical and commercial manufacturing are part of corporate/global systems, however for the sake for clarity highlights of material control strategy for subsequent stages of product life cycle are listed below:
Ă˜  Clinical Phase 1 / 2 Manufacturing:
-   Identification of CMA and preparation of material control specification
-   Development of non compendial test methods
-   Verification of vendor COA for received material against specifications
-   Testing and release of material for parameters such as identification, appearance, safety (bioburden, endotoxin etc) and identified CMA. Availability of approved test methods for these attributes.
-   Vendor qualification for material traceability, supply security with logistic mapping
Ă˜  Clinical Phase 3 Manufacturing:
-   Material (customer) specification finalization with all CMA listed
-   Availability of test methods for verification of all listed specification parameters for materials.
-   Testing and release of material as per customers’ specification. Full testing for all critical materials.
-   Test values vs material attributes (as per specification) to be trended for better material control as per QMS requirements
-   Risk assessment to be revisited for all critical materials.
-   Due diligence and vendor audits to be performed based on risk assessment.
-   Detailed vendor assessment and qualification.
-   Quality agreements, supply security agreements to be executed and made available.
Ă˜  Marketing (including PPQ) and Post Marketing:
-   Material to be qualified and trends for high critical tests for critical materials made available. Specifications can be revisited for better control.
-   Vendors to be audited and compliance ensured. All agreements in place.
-   Alternate vendor development can be initiated.
-   Risk assessment based reduced testing can be explored.
DEFINITIONS
Active Pharmaceutical Ingredient (API) (or Drug Substance): Any substance or mixture of substances intended to be used in the manufacture of a drug (medicinal) product and that, when used in the production of a drug, becomes an active ingredient of the drug product. Such substances are intended to furnish pharmacological activity or other direct effect in the diagnosis, cure, mitigation, treatment, or prevention of disease or to affect the structure and function of the body. (Reference ICH Q7).
API Starting Material: A raw material, intermediate, or an API that is used in the production of an API and that is incorporated as a significant structural fragment into the structure of the API. An API starting material can be an article of commerce, a material purchased from one or more suppliers under contract or commercial agreement, or produced in-house. API starting materials are normally of defined chemical properties and structure. (Reference ICH Q7).
Contamination: The undesired introduction of impurities of a chemical or microbiological nature, or of foreign matter, into or onto a raw material, intermediate, or API during production, sampling, packaging, or repackaging, storage or transport. (Reference ICH Q7)
Control Strategy: A planned set of controls, derived from current product and process understanding, that assures process performance and product quality. The controls can include parameters and attributes related to drug substance and drug product materials and components, facility and equipment operating conditions, in-process controls, finished product specifications, and the associated methods and frequency of monitoring and control. (Reference ICH Q10)
Critical Material Attribute: A physical, chemical, biological or microbiological property or characteristic of an input material that should be within an appropriate limit, range, or distribution to ensure the desired quality of output material. (Reference Raw Materials in the Manufacture of Biotechnology Products: Regulatory Considerations Ruth Cordoba-Rodriguez of the FDA’s Center for Drug Evaluation and Research (CDER in Bethesda, MD)
Excipient: Substances other than the API that have been appropriately evaluated for safety and are intentionally included in a drug delivery system. (Reference: USP <1078>)
Impurity: Any component present in the intermediate or API that is not the desired entity. (Reference ICH Q7)
Lifecycle: All phases in the life of a product from the initial development through marketing until the product’s discontinuation (Reference ICH Q8)
Raw material: Any element or component used in the manufacture of a biotechnology product that comes in contact with the API or the API starting material. A raw material can be reactive or non-reactive with the API. (Reference Raw Materials in the Manufacture of Biotechnology Products: Regulatory Considerations Ruth Cordoba-Rodriguez of the FDA’s Center for Drug Evaluation and Research (CDER in Bethesda, MD)
Materials: A general term used to denote starting materials, reagents, and solvents intended for use in the production of intermediates or APIs. (Reference ICH Q7)
Starting material for biological medicinal products: Any substance of biological origin such as micro-organisms, organs and tissues of either plant or animal origin, cells or fluids (including blood or plasma) of human or animal origin, and biotechnological cell constructs (cell substrates, whether they are recombinant or not, including primary cells).” (Reference Dir. 2001/83/EC)
Biological starting materials: starting materials derived from a biological source that mark the beginning of the manufacturing process of a drug, as described in a marketing authorization or licence application, and from which the active ingredient is derived either directly (for example, plasma derivatives, ascitic fluid and bovine lung) or indirectly (for example, cell substrates, host/ vector production cells, eggs and viral strains). (WHO TRS 999, Annex 2)
REFERENCES:
BioProcess International September 2009: Raw Material Control Strategies for Bioprocesses by Gregory Beck, Mark Schenerman, John Dougherty, Ruth Cordoba-Rodriguez, Christopher Joneckis, Anthony Mire-Sluis, and Lorna D. McLeod.
EBE Concept Paper: Management and Control of Raw Materials Used in the Manufacture of Biological Medicinal Products,  29 November 2017, Version 1.
European Journal of Pharmaceutical Sciences, 2015: Pharmaceutical excipients — quality, regulatory and biopharmaceutical considerations. David P. Elder, Martin Kuentz, RenĂ© Holm.
Dir. 2001/83/EC: Directive 2001/83/EC of The European Parliament and of the Council. The Community Code Relating To Medicinal Products For Human Use.
EP 5.2.12: Raw materials of biological origin for the production of cell-based and gene therapy medicinal products.
ICH Q5B: Quality of Biotechnological Products: Analysis of the expression Construct in Cells used for Production of r-DNA Derived Protein Products.
ICH Q5D: Derivation and Characterisation of Cell Substrates Used for Production of Biotechnological/Biological Products.
ICH Q10: Pharmaceutical Quality System.
ICH Q11: Development and Manufacture of Drug Substances (Chemical Entities and Biotechnological/ Biological Entities).
ICH Q7: Good Manufacturing Practice.
ICH Q8 Pharmaceutical Development.
ICH Q9: Quality Risk Management.
IPEC Europe, 2008: Qualification of Excipients for Pharmaceutical Use.
PDA TR 56 (Revised 2016): Application of Phase-Appropriate Quality System and cGMP to the Development of Therapeutic Protein Drug Substances (API of Biological Active Substance).
USP 1078: Good Manufacturing Practices for Bulk Pharmaceutical Excipients.
WHO TRS 999, Annex 2: WHO good manufacturing practices for biological products.



1 comment: