Drug Metabolism in Drug Design and Development Basic Concepts and Practice

(nextflipdebug2) #1
  - 2.5.3 Monoamine Oxidase (MAO)
- 2.5.4 Aldehyde Oxidase and Xanthine Dehydrogenase
- 2.5.5 Peroxidases
- 2.5.6 Alcohol Dehydrogenases (ADH)
- 2.5.7 Aldehyde Dehydrogenases (ALDH)


  • 2.6 Reduction

    • 2.6.1 P450, ADH

    • 2.6.2 NADPH-P450 Reductase

    • 2.6.3 Aldo-Keto Reductases (AKR)

    • 2.6.4 Quinone Reductase (NQO)

    • 2.6.5 Glutathione Peroxidase (GPX)



  • 2.7 Hydrolysis

    • 2.7.1 Epoxide Hydrolase

    • 2.7.2 Esterases and Amidases



  • 2.8 Summary

    • References



  • 3 Conjugative Metabolism of Drugs

    • 3.1 UDP-Glucuronosyltransferases

      • 3.1.1 Location Within the Cell

      • 3.1.2 Endogenous Substrates

      • 3.1.3 Enzyme Multiplicity

      • 3.1.4 Inducibility

      • 3.1.5 Pharmacogenetics

      • 3.1.6 Experimental Considerations

      • 3.1.7 Enzyme Selective Substrates and Inhibitors

      • 3.1.8 Drug–Drug Interactions and Glucuronidation

      • 3.1.9 Summary



    • 3.2 Cytosolic Sulfotransferases

      • 3.2.1 Cellular Location and Tissue Expression

      • 3.2.2 The SULT Superfamily of Cytosolic Enzymes

      • 3.2.3 Inducibility

      • 3.2.4 SULT Pharmacogenetics

      • 3.2.5 Analytical Detection of Sulfonated Metabolites

      • 3.2.6 SULT Inhibitors (Pacifici and Coughtrie, 2005)

      • 3.2.7 Drug–Drug Interactions and Sulfonation

      • 3.2.8 Summary



    • 3.3 Glutathione-S-Transferases

      • 3.3.1 General Overview

      • 3.3.2 Classification of the GST Enzymes

      • 3.3.3 Localization and Expression

      • 3.3.4 Reactions Catalyzed by GSTs

      • 3.3.5 Regulation of GSTs

      • 3.3.6 GST Alpha Class

      • 3.3.7 GST Mu Class

      • 3.3.8 GST Pi Class

      • 3.3.9 GST Theta Class

      • 3.3.10 GST Zeta Class

      • 3.3.11 Incubation Conditions and Analytical Methods

        • Pathway) 3.3.12 Glutathione Conjugate Metabolism (Mercapturic Acid



      • References





  • 4 Enzyme Kinetics

    • 4.1 Introduction Timothy S. Tracy

    • 4.2 Enzyme Catalysis

    • 4.3 Michaelis–Menten Kinetics

      • 4.3.1 Meanings ofKm,Vmaxand Their Clinical Relevance



    • 4.4 Graphical Kinetic Plots

    • 4.5 Atypical Kinetics–Allosteric Effects

      • 4.5.1 Overview of Atypical Kinetic Phenomena

      • 4.5.2 Homotropic Cooperativity

      • 4.5.3 Heterotropic Cooperativity



    • 4.6 Graphical Analysis of Atypical Kinetic Data

    • 4.7 Enzyme Inhibition Kinetics

      • 4.7.1 Overview

      • 4.7.2 Competitive Inhibition

      • 4.7.3 Mixed Inhibition

      • 4.7.4 Noncompetitive Inhibition

      • 4.7.5 Uncompetitive Inhibition

        • Kinetic Parameters 4.7.6 Summary of Effects of Various Inhibition Types of



      • 4.7.7 Meanings of IC 50 andKiParameters



    • 4.8 Inhibition Kinetics Graphical Plots

    • 4.9 Mechanism-Based Enzyme Inactivation Kinetics

      • Acknowledgment

      • References





  • 5 Metabolism-Mediated Drug–Drug Interactions

    • 5.1 Introduction Hongjian Zhang, Michael W. Sinz, and A. David Rodrigues

    • 5.2 Enzyme Inhibition

      • 5.2.1 Types of Inhibition

      • 5.2.2 In vitroEvaluation of Inhibition

      • 5.2.3 Prediction of CYP Inhibition UsingIn vitroData

      • 5.2.4 Clinical Evaluation of Inhibition





  • 5.3 Enzyme Induction

    • 5.3.1 Enzyme and Pharmacokinetic Changes

    • 5.3.2 Mechanisms of Enzyme Induction

    • 5.3.3 Induction Models



  • 5.4 Reaction Phenotyping

    • 5.4.1 Experimental Considerations

    • 5.4.2 Data Interpretation and Integration

    • 5.4.3 Clinical Evaluation

    • References

    • and Drug Resistance 6 Drug Transporters in Drug Disposition, Drug Interactions,



  • 6.1 Introduction Cindy Q. Xia, Johnny J. Yang, and Suresh K. Balani

    • and Toxicity 6.2 Roles of Transporters in Drug Disposition

    • 6.2.1 Transporters in Drug Absorption

    • 6.2.2 Transporters in Drug Distribution

    • 6.2.3 Transporters in Drug Metabolism

    • 6.2.4 Transporters in Drug Excretion

    • 6.2.5 Transporters in Toxicity



  • 6.3 Transporters in Drug Resistance

  • 6.4 Polymorphism of Transporters and Interindividual Variation

  • 6.5 Transporters in Drug–Drug or Drug–Food Interactions

    • 6.5.1 Oral Absorption

    • 6.5.2 Brain Penetration

    • 6.5.3 Renal Excretion and Hepatic Clearance

    • 6.5.4 Food Effect

    • 6.5.5 Formulation Effect

    • 6.5.6 In vitro–In vivoCorrelation

    • or Inducer 6.6 Methods to Evaluate Transporter Substrate, Inhibitor,

    • 6.6.1 In vitroModels

    • 6.6.2 In situ/Ex vivoModels

    • 6.6.3 In vivoModels



  • 6.7 Conclusions and Perspectives

    • References

    • Drug Interaction Studies 7 Regulatory Considerations of Drug Metabolism and



  • 7.1 Introduction Xiaoxiong Wei and Mingshe Zhu

  • 7.2 Regulatory Guidances Relevant to Drug Metabolism

    • 7.2.1 Toxicokinetic Studies

    • 7.2.2 Use of Radiolabeled Materials

    • 7.2.3 Metabolite Safety Assessment

    • 7.2.4 Drug–Drug Interaction Studies

    • 7.2.5 Analytical Method Validation and Compliance

    • 7.2.6 Regulatory Submission Format and Content



  • 7.3 Metabolism Studies Relevant to Metabolite Safety Assessment

    • 7.3.1 Goals and General Strategies

    • 7.3.2 In vitroMetabolite Profiling Studies

    • 7.3.3 ADME Studies

    • 7.3.4 Analytical Methods for Metabolite Profiling

    • 7.3.5 Special Considerations



  • 7.4 Drug–Drug Interaction Studies

    • 7.4.1 General Strategies

    • 7.4.2 In vivoStudies

    • 7.4.3 Case Study



  • 7.5 Conclusions

    • Acknowledgment

    • References

      • PHARMACEUTICAL INDUSTRY PART II ROLE OF DRUG METABOLISM IN THE



    • Drug Discovery Process 8 Drug Metabolism Research as an Integral Part of the



  • 8.1 Introduction W. Griffith Humphreys

  • 8.2 Metabolic Clearance

    • 8.2.1 General

    • 8.2.2 Prediction of Human Clearance

    • 8.2.3 In vivoMethods to Study Metabolism

    • 8.2.4 Screening Strategies

    • 8.2.5 In silicoMethods to Study Metabolism



  • 8.3 Metabolite Profiling

  • 8.4 Reaction Phenotyping

  • 8.5 Assessment of Potential Toxicology of Metabolites

    • 8.5.1 Reactive Metabolite Studies—In vitro

    • 8.5.2 Reactive Metabolite Studies—In vivo

      • with Off-Target Receptors 8.5.3 Toxicology Mediated Through Metabolite Interaction





  • 8.6 Assessment of Potential for Active Metabolites

    • 8.6.1 Detection of Active Metabolites During Drug Discovery

      • Activity of Metabolites 8.6.2 Methods for Assessing and Evaluating the Biological



    • 8.6.3 Methods for Generation of Metabolites



  • 10.3 New Radiochromatography Techniques

    • 10.3.1 HPLC-MSC

    • 10.3.2 Stop-Flow HPLC-RFD

    • 10.3.3 Dynamic Flow HPLC-RFD

    • 10.3.4 UPLC-Radiodetection

    • 10.3.5 HPLC-AMS



  • 10.4 Radiochromatography in Conjunction with Mass Spectrometry

    • 10.4.1 LC-RFD-MS

    • 10.4.2 Stop-Flow and Dynamic Flow LC–RFD–MS

    • 10.4.3 LC-MSC-MS

      • Approach



    • Drug Metabolism Studies 10.5 Application of New Radiochromatography Techniques in

    • 10.5.1 Profiling of Radiolabeled Metabolites in Plasma

      • Using Radiolabeled Cofactors or Trapping Agents 10.5.2 Analysis of Metabolites of Nonradiolabeled Drugs

      • of Sequential Metabolites 10.5.3 Determination of Structures and Formation Pathways



    • 10.5.4 Enzyme Kinetic Studies



  • 10.6 Summary

    • References

    • for Metabolite Identification 11 Application of Liquid Chromatography/Mass Spectrometry



  • 11.1 Introduction Shuguang Ma and Swapan K. Chowdhury

  • 11.2 LC/MS Instrumentation

    • 11.2.1 High Performance Liquid Chromatography (HPLC)

    • 11.2.2 Atmospheric Pressure Ionization Methods

    • 11.2.3 Mass Analyzers

    • 11.2.4 Tandem Mass Spectrometry



  • 11.3 Metabolite Identification––Role of LC/MS

    • 11.3.1 Metabolite Characterization in Drug Discovery

      • Clinical Development 11.3.2 Metabolite Identification in Preclinical and



    • Identification 11.4 Techniques for Improving Metabolite Detection and

    • 11.4.1 Chemical Derivatization

    • 11.4.2 Stable Isotope Labeling

    • 11.4.3 Hydrogen/deuterium (H/D) Exchange MS

    • 11.4.4 Accurate Mass Measurement

      • Metabolite Identification 11.4.5 Nanospray Ionization (NSI) MS for





  • 11.5 Software-Assisted Metabolite Identification

    • 11.5.1 Data-Dependent Acquisition (DDA)

    • 11.5.2 Mass Defect Filter (MDF)

    • Identification 11.6 Additional MS-Related Techniques for Metabolite

    • 11.6.1 LC/NMR/MS

    • 11.6.2 LC/ICPMS



  • 11.7 Characterization of Unstable Metabolites

    • 11.7.1 Glucuronides

    • 11.7.2 N-Oxides

      • Ions by the Presence of Alkali Adducts 11.7.3 Differentiation of Molecular Ions from In-Source Fragment



    • Intermediates 11.8 Detection and Characterization of Reactive Metabolites and

    • 11.8.1 Trapping Reactive Metabolites

    • 11.8.2 Screening for Glutathione Conjugates



  • 11.9 Conclusions and Future Directions

    • Acknowledgments

    • References

    • Structure Determination 12 Introduction to NMR and Its Application in Metabolite



  • 12.1 Introduction and Vikram Roongta

  • 12.2 Theory

  • 12.3 NMR Hardware

  • 12.4 NMR Observables

    • 12.4.1 Chemical Shifts

    • 12.4.2 Coupling Constants

    • 12.4.3 Integration



  • 12.5 Sample Requirements for NMR

  • 12.6 Most Commonly Used NMR Experiments and Techniques

    • 12.6.1 1D NMR Experiments

    • 12.6.2 2D NMR Experiments

    • 12.6.3 Solvent Suppression Techniques

    • 12.6.4 Hyphenated NMR Methods

    • or Metabolites 12.7 General protocol for NMR Analysis of Unknown Compounds

    • Known Biotransformations 12.8 Examples of Metabolite Structure Determination from

    • References

      • Method in Human or Rat Liver Microsomes—A Semiautomated



    • 14.2.5 Protocol ForIn vivoCovalent Protein Binding in Rats

    • 14.2.6 Notes

    • Concentrations in Hepatocytes 14.3 Protocol for Measurement of Intracellular GSH and GSSG

    • 14.3.1 Introduction

      • Hepatocytes 14.3.2 Measurement of Intracellular GSH/GSSG in



    • 14.4 Perspectives

      • Acknowledgments

      • References





  • 15 Reaction Phenotyping

    • 15.1 Introduction Susan Hurst, J. Andrew Williams, and Steven Hansel

    • 15.2 Cytochrome P450 Reaction Phenotyping

    • 15.3 Noncytochrome P450 Reaction Phenotyping

      • 15.3.1 Flavin-Containing Monooxygenases

        • MAO-B) 15.3.2 Monoamine Oxidases A and B (MAO-A and



      • 15.3.3 Esterases



    • 15.4 Conjugation Phenotyping

      • 15.4.1 UGT Reaction Phenotyping

      • 15.4.2 N-Acetylation Reaction Phenotyping

      • 15.4.3 Sulfation Reaction Phenotyping



    • 15.5 Transporter Phenotyping

    • 15.6 Nonradiolabeled Reaction Phenotyping

      • 15.6.1 Objective

      • 15.6.2 Selection of Appropriate Experimental Systems

      • 15.6.3 Experimental Approach Considerations

      • 15.6.4 Selection of Appropriate Experimental Designs

        • Enzyme Systems 15.6.5 Quantitative Reaction Phenotyping: Expressed or Purified





    • 15.7 Radiolabeled Reaction Phenotyping

      • Phenotyping Studies 15.7.1 QuantitativeIn vitroRadiolabeled Reaction

      • 15.7.2 In vivoQuantitative ADME Studies

      • 15.7.3 Drug–Drug Interaction Potential

      • 15.7.4 Specialized Clinical Studies



    • 15.8 Summary and Future Directions

      • Acknowledgments

      • References



    • Enzyme Incubation Method Sheet Appendix A: Reaction Phenotyping—Expressed cDNA

    • Chemical Inhibition Appendix B: Reaction Phenotyping—Microsomal

      • Discovery and Development 16 Analysis ofIn vitroCytochrome P450 Inhibition in Drug



    • 16.1 Introduction Magang Shou and Renke Dai

    • 16.2 Reversible Inhibition

      • 16.2.1 Materials and Reagents

      • 16.2.2 Instrument

      • 16.2.3 Optimization of Kinetic Reaction

      • 16.2.4 LC/MS/MS Analysis

      • 16.2.5 Automated Sample Preparation and Incubation

      • 16.2.6 Data Analysis



    • 16.3 Irreversible Inhibition

      • 16.3.1 Kinetic Model for Mechanism-Based Inhibition

      • 16.3.2 Measurements of Kinetic Parameters

      • 16.3.3 General Incubation Procedure and Sample Preparation

      • 16.3.4 Data Analysis



    • 16.4 Fluorescent Assay

      • CYP Inhibition Data 16.5 Prediction of Human Drug–Drug Interactions fromIn vitro

      • 16.5.1 Reversible CYP Inhibition

        • from Mechanism-Based CYP Inhibition 16.5.2 Prediction of Human Drug–Drug Interactions



      • 16.5.3 Factors Affecting the Prediction of Drug–Drug Interactions



    • 16.6 Conclusion

      • Acknowledgment

      • References





  • 17 Testing Drug Candidates for CYP3A4 Induction

    • 17.1 Introduction Gang Luo, Liang-Shang Gan, and Thomas M. Guenthner

    • 17.2 Assessments

      • Animal Models 17.2.1 Assessment of Induction Potential Using Intact

      • 17.2.2 Assessment of Induction Potential UsingIn vitroModels

        • in Humans 17.2.3 Direct Assessment of CYP3A4 InductionIn vivo





    • 17.3 Final Comments

      • References

      • Metabolite Profiling and Identification, and Data Presentation 18 ADME Studies in Animals and Humans: Experimental Design,



    • 18.1 Objectives, Rational, and Regulatory Compliance Donglu Zhang and S. Nilgun Comezoglu

    • 18.2 Study Designs

      • 18.2.1 Choice of Radiolabel

      • 18.2.2 Preparation of Animals and Human Subjects

      • 18.2.3 Dose Selection, Formulation, and Administration

        • Sample Collection/Pooling 18.2.4 In-Life Studies in Animals and Humans and





    • 18.3 Sample Analysis

      • 18.3.1 Sample Preparation: Plasma, Urine, Bile, and Feces

      • 18.3.2 Radioactivity Determination

      • 18.3.3 LC/MS/MS Quantification and Pharmacokinetic Analysis

      • 18.3.4 Metabolite Profiling

      • 18.3.5 Metabolite Identification

        • and Generation in Bioreactors 18.3.6 Metabolite Isolation fromIn vivoSamples



      • an Example 18.4 Data Presentation Using Metabolism of [^14 C]Muraglitazar as

        • Radioactivity 18.4.1 Pharmacokinetic Results and Excretion of



      • 18.4.2 Metabolite Profiling in Plasma, Urine, Bile, and Feces

      • 18.4.3 Metabolite Identification by LC/MS/MS



    • 18.5 Conclusions and Path Forward

      • Acknowledgments

      • Appendix A: Rat Tissue Distribution and Dosimetry Calculation

      • References





  • Index

Free download pdf