An Introduction to Bioanalysis of Biopharmaceuticals

This document was uploaded by one of our users. The uploader already confirmed that they had the permission to publish it. If you are author/publisher or own the copyright of this documents, please report to us by using this DMCA report form.

Simply click on the Download Book button.

Yes, Book downloads on Ebookily are 100% Free.

Sometimes the book is free on Amazon As well, so go ahead and hit "Search on Amazon"

The book provides a comprehensive review of the fundamental and practical aspects of bioanalytical support and the integral role it plays in the development of safe and efficacious biopharmaceutical drugs with speed and cost-effectiveness. The book focuses on a broad range of conventional and emerging biopharmaceutical modalities including monoclonal antibody-based therapeutics, gene therapy, cell therapy, peptides and oligonucleotides.

 

The book starts with an introductory overview of bioanalysis showcasing the integral role it plays in understanding the drug disposition (pharmacokinetics/pharmacodynamics and immunogenicity) and the progression of bioanalytical strategy as the drug progresses through discovery and development stages of the program, taking into consideration the continually evolving regulatory landscape.

 

The book further diversifies into individual biopharmaceutical modalities - monoclonal antibodies, antibody-drug conjugates, bispecifics, Fc-fusion proteins, gene therapies, cell therapies, peptides and oligonucleotides. The individual chapters focus on modality-specific bioanalytical assay strategies, critical reagents, assay formats, analytical platforms, associated bioanalytical challenges and mitigation strategies, industry best practices, and the latest understanding of regulatory guidance as applicable to the fast-growing biopharmaceutical landscape.

Author(s): Seema Kumar
Series: AAPS Advances in the Pharmaceutical Sciences Series, 57
Publisher: Springer
Year: 2022

Language: English
Pages: 165
City: Cham

Preface
Contents
About the Editor
Chapter 1: Introduction
1.1 Biopharmaceutical Diversity
1.2 Small-Molecule Vs Large-Molecule Biotherapeutics Bioanalysis
1.3 Biotherapeutics Drug Disposition
1.3.1 Absorption/Distribution
1.3.2 Metabolism/Elimination
1.3.3 Excretion
1.4 Immunogenicity
1.5 Bioanalytical Strategies for Biotherapeutics
1.5.1 Ligand Binding Assays for Biotherapeutics
1.5.2 Detection Platforms
1.5.3 Direct and Indirect Measurement of Biotherapeutics Using LC-MS
1.5.3.1 Signature Peptides
1.5.3.2 Top-Down or Intact Measurement
1.5.3.3 Sample Preparation for Signature Peptides (Bottom-Up Quantification)
1.6 Biotherapeutics Bioanalytical Challenges and Future Directions
References
Chapter 2: An Introduction to Bioanalysis of Monoclonal Antibodies
2.1 Introduction
2.2 Pharmacokinetic/Pharmacodynamic Properties
2.3 Pharmacokinetic Assessment
2.3.1 PK Quantitation by Immunoassay
2.3.2 PK Quantitation by LC-MS/MS
2.3.2.1 Sample Preparation
Protein Precipitation (Pellet Precipitation)
2.3.2.2 Protein Immunocapture
Generic Immunocapture
Nonclinical Studies
Clinical Studies
Selective Immunocapture
2.3.2.3 Protein Digestion
2.3.2.4 Surrogate Peptide Selection
Universal Surrogate Peptides
Signature Surrogate Peptides
2.3.2.5 Internal Standard
Stable Isotope Labeled Peptide Internal Standard
Stable Isotope Labeled Monoclonal Antibody Internal Standard
2.4 Immunogenicity
2.4.1 Immunogenicity Assessment by Immunoassays
2.4.1.1 Screening Assay
Screening Cut Point
2.4.1.2 Confirmatory/Specificity Assay
Confirmatory/Specificity Cut Point
2.4.1.3 Titer Assay
2.4.1.4 Isotyping Assays
2.4.1.5 Neutralizing Antibody Assay
Direct Assay
Indirect Assay
2.4.2 Analytical Considerations for ADA Assays
2.4.2.1 Sensitivity and Positive Control
2.4.2.2 Drug Tolerance
2.4.2.3 Preexisting Antibodies
2.4.2.4 Target Interference and Rheumatoid Factor
2.4.3 Immunogenicity Assessment by LC-MS/MS
2.4.3.1 LC-MS Strategies for ADA Quantification
2.4.3.2 LC-MS Strategies with Limited Applicability
2.4.3.3 Universal LC-MS Strategies with More Applicability
2.5 Future Developments and Challenges
References
Chapter 3: An Introduction to Bioanalysis of Antibody-Drug Conjugates
3.1 Introduction
3.2 Chemistry of Antibody-Drug Conjugates
3.2.1 Antibody
3.2.2 Payload
3.2.3 Linker
3.2.3.1 Cleavable Linker
3.2.3.2 Non-cleavable Linker
3.2.4 ADC Conjugation Chemistry
3.3 ADC Mechanism of Action
3.4 PK Considerations for Pre-Clinical and Clinical Development of ADC
3.5 Bioanalysis of ADC to Support their PK Assessment
3.5.1 Bioanalytical Platforms for ADC
3.5.1.1 Ligand Binding Assays for ADC Bioanalysis
Total Antibody by LBA
Total ADC (Conjugated Antibody) by LBA
3.5.1.2 Mass Spectrometry for ADC Bioanalysis
Hybrid LC-MS/MS
Hybrid-LC-MS/MS for Total Antibody Assay Quantitation
Hybrid-LC-MS/MS for Total ADC (Conjugated-Antibody) Quantitation
Hybrid-LC-MS/MS for Total ADC (Conjugated-Payload) Quantitation
LC-MS/MS-Based ADC Bioanalysis
Unconjugated Payload Quantitation
3.5.2 In Vivo DAR Characterization
3.6 Immunogenicity Assays for ADC
3.6.1 Immunogenicity Testing Strategy for ADC
3.6.2 Screening Assay for ADC
3.6.3 Confirmatory Assay for ADC
3.6.4 Titer Assay for ADC
3.6.5 Neutralizing Antibody Assay for ADC
3.6.6 Domain Characterization Assay for ADC
3.7 Future Perspective
References
Chapter 4: An Introduction to Bioanalysis of Bispecific and Fusion Proteins
4.1 Introduction: Structural Overview, Comparisons, and History of bsAb and Fusion Proteins
4.1.1 Bispecific Antibodies
4.1.2 Fusion Proteins
4.1.3 ADME
4.2 Bioanalytical Method Development and Scope
4.2.1 Pharmacokinetic (PK) Approach
4.2.1.1 Bioanalytical Strategy
4.2.1.2 Critical Reagent Generation
4.2.1.3 Platforms
4.2.1.4 Clinical Vs. Nonclinical
4.2.1.5 Challenges in PK Bioanalysis
4.2.2 Immunogenicity and Anti-Drug Antibody (ADA) Approach
4.2.2.1 Bioanalytical Strategy
4.2.2.2 Critical Reagent Generation
4.2.2.3 Platforms
4.2.2.4 Clinical Vs. Nonclinical Bioanalytical Strategies
4.2.2.5 Challenges in ADA Bioanalysis
4.3 Regulatory Guidelines
4.3.1 Nonclinical and Clinical Considerations
4.4 Emerging and Novel Trends
References
Chapter 5: Gene Therapy and Cell Therapy: Bioanalytical Challenges and Practical Solutions
5.1 Introduction
5.2 What Is Gene and Cell Therapy?
5.3 Bioanalysis
5.4 Immunogenicity Evaluation
5.4.1 Binding Antibody Assays
5.4.1.1 Strategy
5.4.1.2 Challenges and Solutions
5.4.2 Neutralizing Antibody (NAb) Assay
5.4.2.1 Strategy
5.4.2.2 Challenges and Solutions
5.4.3 Cellular Immunity
5.4.3.1 Strategy
5.4.3.2 Challenges and Solutions
5.4.4 Regulatory Perspective on Immunogenicity Assessment for GTx and CTx
5.5 Pharmacokinetic Evaluations
5.5.1 PK Assessment Strategies for GTx
5.5.2 Strategic Considerations for CTx
5.5.2.1 Somatic Cell Therapies
5.5.2.2 Cell Therapies with Ex Vivo Modification Using Viral Technologies
5.5.2.3 Immortalized CTx
5.5.3 Bioanalytical Methods for PK Evaluations of GTx and CTx Therapies
5.5.4 PK Assay Development Strategies
5.5.5 Discussion
References
Chapter 6: Peptides and Oligonucleotide-Based Therapy: Bioanalytical Challenges and Practical Solutions
6.1 Introduction
6.1.1 Peptide Drugs and Modifications
6.1.2 Oligonucleotide Drugs and Modifications
6.2 Peptides Bioanalysis for Pharmacokinetic Evaluations
6.2.1 Assay Development on LC-MS/MS Platform
6.2.2 Challenges in Sample Handling for Peptide Bioanalysis
6.2.2.1 Adsorption
6.2.2.2 Stability
6.2.3 Sample Preparation for Peptide Bioanalysis by LC-MS/MS
6.2.4 Instrumentation for Peptide Bioanalysis by LC-MS/MS
6.3 Oligonucleotide Bioanalysis for Pharmacokinetic Evaluations
6.3.1 Assay Development Using LC-MS Platform
6.3.2 Assay Development Using LBA Platform
6.3.3 Assay Development Using qPCR Platform
6.3.4 Regulatory Requirements for Bioanalysis of Oligonucleotides
6.4 Immunogenicity Risk Assessment for Peptides and Oligonucleotide Therapeutics
6.4.1 Product-Specific Factors
6.4.1.1 Amino Acid or Nucleotide Modifications
6.4.1.2 Product Aggregation
6.4.1.3 Impurities
6.4.1.4 Formulation
6.4.2 Immunogenicity Assessment for Peptide Therapeutics
6.4.2.1 In Silico Immunogenicity Analysis
6.4.2.2 MHC-Peptide Binding Assay
6.4.2.3 Transient Cell-Line-Based Reporter Assays
6.4.2.4 Functional in Vitro Cell-Based Assay
6.5 ADA Assay for Peptides and Oligonucleotide Therapeutics
6.6 Regulatory Perspective
6.7 Future Perspective
References
Index