Specification of Drug Substances and Products
Development and Validation of Analytical Methods
- 3rd Edition - September 22, 2024
- Editors: Christopher M. Riley, K. Lien Nguyen
- Language: English
- Paperback ISBN:9 7 8 - 0 - 4 4 3 - 1 3 4 6 6 - 1
- eBook ISBN:9 7 8 - 0 - 4 4 3 - 1 3 4 6 7 - 8
Specification of Drug Substances and Products: Development and Validation of Analytical Methods, Third Edition contains chapters discussing the unique requirements for the unive… Read more
Purchase options
Institutional subscription on ScienceDirect
Request a sales quoteSpecification of Drug Substances and Products: Development and Validation of Analytical Methods, Third Edition contains chapters discussing the unique requirements for the universal critical quality attributes, as well as the specific tests required to characterize and control different types of products, ranging in complexity from small molecules in immediate release oral dosage forms to complex products such as drug-antibody conjugates and mRNA-based products. This substantially expanded revision of the second edition will serve as practical comprehensive reference for scientists, managers, educators, and consultants involved in the development and regulation of pharmaceutical products.
- Presents critical assessment, potential impact, and application of the recent revisions to ICH guidelines on method validation (Q2) (as well as the latest guideline on Analytical Method Development (Q14), and the special regional requirements in non-ICH regions.
- Addresses comprehensive treatment of the development and validation of analytical methodologies used in the analysis, control, and specification of a variety of different types of dosage forms, ranging from traditional oral solid dosage forms to proteins, nRNA-based drugs, vaccines, and gene therapy. This book will also address drug–device combination
products such as digital drug delivery systems, transdermal systems, and inhalation products. - Presents detailed treatment of latest statistical approaches, including new approaches to the treatment of validation data method, specification setting, and shelf-life prediction (based on stability data).
- Cover image
- Title page
- Table of Contents
- Copyright
- Contributors
- About the editors
- Section I. Introduction
- Chapter 1. Introduction
- 1.1. Historical perspective
- 1.2. Specifications: General principles
- 1.3. Analytical methods
- 1.4. Validation of analytical methods and the reporting of analytical results
- 1.5. Conclusions
- Section II. Regulatory considerations and statistical approaches
- Chapter 2. Principles for setting specifications and shelf lives
- 2.1. Introduction
- 2.2. Regulatory requirements and guidelines
- 2.3. Specification setting process
- 2.4. Elements to include in specifications
- 2.5. Scientific considerations in setting acceptance criteria
- 2.6. Statistical considerations for setting acceptable limits
- 2.7. Evaluation of stability data and determination of shelf life
- 2.8. Conclusions
- Chapter 3. Stability studies: General regulatory considerations and regional differences
- 3.1. Introduction
- 3.2. Global stability programs to support drug development
- 3.3. ICH stability program
- 3.4. WHO stability guideline
- 3.5. ASEAN stability guideline
- 3.6. ANVISA stability guideline
- 3.7. Vaccine stability program
- 3.8. Drug-device combination product stability
- 3.9. Conclusions
- Chapter 4. Kinetics and mechanisms of drug degradation
- 4.1. Introduction
- 4.2. General principles
- 4.3. Mechanisms of drug degradation
- 4.4. Conclusions
- Chapter 5. Analytical procedure life-cycle management
- 5.1. Introduction
- 5.2. Analytical method development and method performance control
- 5.3. Modality, technology, and development phase-specific considerations
- 5.4. Conclusions
- Chapter 6. Process analytical technology
- 6.1. Introduction
- 6.2. Drivers behind the development and implementation of process analytical technology
- 6.3. Quantifiable benefits of process analytical technology
- 6.4. Additional considerations of process analytical technology
- 6.5. Conclusions and future directions
- Chapter 7. Pharmacopeial methods and tests
- 7.1. Introduction
- 7.2. Legal recognition
- 7.3. Requirements for inclusion in a pharmacopeia
- 7.4. Revision process
- 7.5. Input from regulatory authorities
- 7.6. Pharmacopeial harmonization
- 7.7. Content of monographs
- 7.8. Compendial reference standards
- 7.9. Conclusions
- Chapter 8. Development of United States Pharmacopeia-National Formulary (USP–NF) Monographs and General Chapters
- 8.1. Introduction
- 8.2. Structure of the USP-NF compendium
- 8.3. The standard setting process for the USP-NF
- 8.4. Revision process for Monographs and General Chapters
- 8.5. The appeals process
- 8.6. Harmonization of Monographs and General Chapters
- 8.7. New dosage forms of existing drugs
- 8.8. Analytical method management
- 8.9. Further method development to implement a procedure
- 8.10. Conclusions
- Chapter 9. The distinctiveness of the British Pharmacopoeia and its relationship to other Pharmacopeia
- 9.1. The British Pharmacopoeia
- 9.2. Content, organizational structure and management
- 9.3. Harmonization
- 9.4. Tests and methods: An academic user's perspective
- 9.5. Reference standards
- 9.6. Recent developments
- 9.7. Conclusions
- Chapter 10. Specifications and analytical procedures in registration applications
- 10.1. Introduction
- 10.2. Guidelines relevant for specifications and analytical methods in registration applications
- 10.3. Preparation of a core submission dossier for registration applications
- 10.4. Interactions with regulatory authorities
- 10.5. Regulatory perspective of GMP aspects and inspections
- 10.6. Post-approval activities
- 10.7. Conclusions
- Chapter 11. Specifications and analytical procedures in clinical trial applications
- 11.1. Introduction
- 11.2. Principles for the CMC part of a CTA/IND
- 11.3. Guidance relevant for the CMC content in CTA/IND submissions
- 11.4. Preparation of a quality dossier for clinical trial applications
- 11.5. CTA/IND submission, review, approval and amendments
- 11.6. Interactions with regulatory authorities
- 11.7. Regulatory perspective of GMP aspects and inspections
- 11.8. Conclusions
- Section III. Critical quality attributes
- Chapter 12. Description and identification
- 12.1. Introduction
- 12.2. Description
- 12.3. Drug substance identification testing
- 12.4. Drug product identification
- 12.5. Conclusions
- Chapter 13. Assay and impurities: Specifications of new chemical entities (“small molecules”)
- 13.1. Introduction
- 13.2. Analytical methodology
- 13.3. Calculations
- 13.4. Analysis of samples
- 13.5. Specification of assay and impurities
- 13.6. Other applications of assay methods
- 13.7. Conclusions
- Appendix A.13. Rules of rounding, significant figures and use of spreadsheets
- Chapter 14. Assay and impurities: Method development as part of analytical life-cycle management
- 14.1. Introduction
- 14.2. Rational design of analytical methods
- 14.3. Techniques and mode selection based on physicochemical properties
- 14.4. Maximizing orthogonality using column screening and mobile phase screening
- 14.5. Systematic and efficient column and mobile phase screening
- 14.6. Method optimization and robustness using simulation software
- 14.7. Enhanced selectivity, peak capacity, orthogonal methods and two-dimensional chromatography
- 14.8. Cases studies: Examples of systematic method development
- 14.9. Conclusions
- Chapter 15. Assay and impurities: Method validation
- 15.1. Introduction
- 15.2. Validation of methods for assay, impurities, and related procedures
- 15.3. Conclusions
- Chapter 16. Mutagenic impurities
- 16.1. Introduction
- 16.2. Assessment and control as a function of the life-cycle
- 16.3. Developing control strategies
- 16.4. Impurities to be considered for mutagenic risk assessment (MRA)
- 16.5. Analytical challenges and strategies
- 16.6. Case study 1 – galunisertib
- 16.7. Case study 2 – pazopanib hydrochloride
- 16.8. Case study 3 – N-nitrosamines in sartans
- 16.9. Conclusions
- Chapter 17. Residual solvents
- 17.1. Introduction
- 17.2. Residual solvent classes
- 17.3. Supplier management
- 17.4. Risk assessment
- 17.5. Specifications and acceptance criteria
- 17.6. Analytical methods
- 17.7. Analytical methods validation
- 17.8. Conclusions
- Chapter 18. Elemental and inorganic impurities
- 18.1. Scope
- 18.2. Introduction
- 18.3. Compliance requirements
- 18.4. Elemental impurity classification
- 18.5. Permitted daily exposure limits
- 18.6. Compliance options
- 18.7. Preparing a risk assessment
- 18.8. Analytical techniques
- 18.9. Method development
- 18.10. Method validation
- 18.11. Other compendial methods for inorganic impurities
- 18.12. Life-cycle management
- 18.13. Conclusions
- Chapter 19. Extractables and leachables
- 19.1. Introduction
- 19.2. Materials risk assessment
- 19.3. ISO 10993-18 – medical device
- 19.4. Extractable analysis
- 19.5. Leachables study
- 19.6. Leachables study design
- 19.7. Impact of leachables in the drug product above the AET
- 19.8. Conclusions
- Chapter 20. Microbiology methods
- 20.1. Introduction
- 20.2. General microbiology practices
- 20.3. Microbial testing for non-sterile products
- 20.4. Testing for sterile products
- 20.5. Particulate matter in injection products
- 20.6. Commonly used USP microbiology informational chapters
- 20.7. Alternative microbiological methods
- 20.8. Conclusion
- Chapter 21. Solid-state characterization methods: An overview
- 21.1. Introduction
- 21.2. Relevance of solid-state characterization
- 21.3. ICH guidelines and potentially controlled solid-state parameters
- 21.4. Timing of solid-state characterization development
- 21.5. Methods overview
- 21.6. Method selection for crystalline/amorphous quantification
- 21.7. Conclusions
- Chapter 22. Solid-state characterization – Method development and validation
- 22.1. Introduction
- 22.2. Method validation
- 22.3. Sample preparation
- 22.4. Sample selection
- 22.5. Data normalization
- 22.6. Feasibility testing
- 22.7. Identification tests
- 22.8. Limit tests
- 22.9. Quantitative methods
- 22.10. System suitability and goodness-of-fit
- 22.11. Conclusions
- Chapter 23. Chiral methods
- 23.1. Introduction
- 23.2. Implementation of health authority guidelines to chiral drug development
- 23.3. Chiral LC
- 23.4. Other chiral separation methods
- 23.5. Non-separation techniques
- 23.6. Chiral method validation
- 23.7. Conclusions
- Chapter 24. Water determination
- 24.1. Introduction
- 24.2. Loss on drying (LOD)
- 24.3. Karl Fischer Titration (KFT)
- 24.4. Gas chromatographic methods
- 24.5. Near-infrared spectroscopy (NIRS)
- 24.6. Dynamic vapor sorption
- 24.7. Sampling and sample preparation
- 24.8. Conclusions
- Section IV. Dosage forms and product types
- Chapter 25. Orally administered dosage forms
- 25.1. Introduction
- 25.2. Universal test methods
- 25.3. Specific test methods
- 25.4. Conclusions
- Chapter 26. Drug release: Topical products
- 26.1. Introduction
- 26.2. In vitro release test
- 26.3. Method development
- 26.4. IVRT method validation
- 26.5. Common sources of variability in IVRT methods
- 26.6. Regulatory guidelines
- 26.7. Conclusions
- Chapter 27. Transdermal products
- 27.1. Introduction
- 27.2. Product quality document relationships to test method development
- 27.3. Drug substance
- 27.4. Adhesive raw material
- 27.5. Excipient and component raw materials
- 27.6. Drug product
- 27.7. Chemical methods
- 27.8. Functional performance methods
- 27.9. Physical property methods
- 27.10. Microbiological methods
- 27.11. In-process controls
- 27.12. Extractables and leachables
- 27.13. Laboratory support operations
- 27.14. Conclusions
- Chapter 28. Inhalation products
- 28.1. Introduction
- 28.2. Drug substance for use in inhalation
- 28.3. Drug products for inhalation
- 28.4. Conclusions
- Chapter 29. Ophthalmic products
- 29.1. Introduction
- 29.2. Specifications and methods for ophthalmic products
- 29.3. Universal specifications and test methods
- 29.4. Specific test methods and specifications
- 29.5. Intraocular ophthalmic products
- 29.6. Conclusions
- Chapter 30. General analytical considerations for parenteral products
- 30.1. Introduction
- 30.2. Quality target product profile for parenteral products
- 30.3. Critical quality attributes for parenteral products
- 30.4. Quality specifications for parenteral products
- 30.5. Critical aspects related to registration of parenteral drug products
- Chapter 31. Specification of biotechnology products including cell and gene therapy
- 31.1. Introduction
- 31.2. Regulatory considerations
- 31.3. Specification of drug substance and drug product
- 31.4. Specification of drug substance
- 31.5. Specification of drug product
- 31.6. Specification of cell and gene therapy (CGT)
- 31.7. Justification of specification
- 31.8. Challenges and issues in specification development
- 31.9. Conclusions
- Chapter 32. Biotechnology products: Validation of analytical methods
- 32.1. Introduction
- 32.2. Method design and the analytical target profile
- 32.3. Method validation
- 32.4. Compendial method verification
- 32.5. Continuous method usage
- 32.6. Prior knowledge
- 32.7. Guidelines
- 32.8. Conclusions
- Chapter 33. Biosimilars
- 33.1. Introduction
- 33.2. Analytical method development
- 33.3. Analytical methods: demonstration of fit-for-purpose and validation
- 33.4. Analytical similarity
- 33.5. Reference materials
- 33.6. Pharmacopeial monographs for biosimilars
- 33.7. Specification of biosimilars
- 33.8. Conclusions
- Chapter 34. Antibody-drug conjugates
- 34.1. Introduction
- 34.2. Structure of an ADC
- 34.3. Overall control strategy for ADC drugs
- 34.4. Critical aspects for the characterization of ADCs
- Chapter 35. Oligonucleotides
- 35.1. Introduction
- 35.2. Impurities
- 35.3. Analytical methods
- 35.4. Conclusions
- Chapter 36. Specifications for vaccines
- 36.1. Overview of different vaccine modalities
- 36.2. General recommendations and justifications for vaccine specifications
- 36.3. Reflections on clinical relevance for vaccine specifications
- 36.4. Release and shelf-life specifications
- 36.5. Illustrative examples of different vaccine modalities, including COVID learnings
- 36.6. Challenges and opportunities for global regulatory harmonization of specifications
- Chapter 37. Nanoparticle suspension dosage forms by nanomilling
- 37.1. Introduction
- 37.2. Universal test methods
- 37.3. Specific test methods
- 37.4. Conclusions
- Chapter 38. Development of connected drug delivery systems
- 38.1. Introduction
- 38.2. Development of connected drug delivery systems
- 38.3. Deliverables
- 38.4. Design verification
- 38.5. Design validation
- 38.6. Development of mechanical parts
- 38.7. Development of embedded or stand-alone software
- 38.8. Conclusions
- Section V. Biological fluids
- Chapter 39. Bioanalysis of biological matrix samples using liquid chromatography-tandem mass spectrometry detection
- 39.1. Introduction
- 39.2. Technologies for the bioanalysis of biological matrix samples
- 39.3. Validation of LC-MS bioanalytical methods
- 39.4. Bioanalytical methods for proteins
- 39.5. Conclusion
- Chapter 40. Bioanalysis of oligonucleotides
- 40.1. Introduction
- 40.2. Overview of bioanalytical methods for oligonucleotides
- 40.3. LCMS assays for the bioanalysis of oligonucleotides
- 40.4. Ligand binding assays for the bioanalysis of oligonucleotides
- 40.5. qPCR assays for the bioanalysis of oligonucleotides
- 40.6. Conclusions
- Index
- No. of pages: 1190
- Language: English
- Edition: 3
- Published: September 22, 2024
- Imprint: Elsevier
- Paperback ISBN: 9780443134661
- eBook ISBN: 9780443134678
CR
Christopher M. Riley
Dr. Christopher Riley is the President of Riley and Rabel Consulting Services. He received a bachelor’s degree in Pharmacy (1977) and PhD degree in Pharmaceutical Chemistry (1980) from the University of Bath, UK. He taught Pharmaceutical Chemistry, at the Universities of Florida (1983-6) and the University of Kansas (1986-94), and was a Vice President at DuPont Merck (1994-2001) and ALZA (a division of J&J) (2001-7). He has coauthored more than 140 book chapters and papers in peer-reviewed journals, as well as 5 books. He has extensive experience in CMC regulatory affairs and the development of all types of dosage forms.
KN