Cover image for Predictive Toxicology : From Vision to Reality, Volume 64.
Predictive Toxicology : From Vision to Reality, Volume 64.
Title:
Predictive Toxicology : From Vision to Reality, Volume 64.
Author:
Mannhold, Raimund.
ISBN:
9783527674206
Personal Author:
Edition:
1st ed.
Physical Description:
1 online resource (483 pages)
Series:
Methods and Principles in Medicinal Chemistry Ser. ; v.64

Methods and Principles in Medicinal Chemistry Ser.
Contents:
Predictive Toxicology: From Vision to Reality -- Contents -- List of Contributors -- Preface -- A Personal Foreword -- 1 Introduction to Predictive Toxicology Tools and Methods -- 1.1 Computational Tools and Bioinformatics -- 1.1.1 In Silico Prediction Tools -- 1.1.2 Bioinformatics -- 1.2 Omics Technologies -- 1.2.1 Toxicogenomics (Transcriptomics) -- 1.2.2 Proteomics -- 1.2.3 Metabolomics -- 1.3 Data Interpretation and Knowledge Management -- 1.4 Biomarker Development -- 1.5 Advanced In Vitro Systems and Stem Cell Research -- 1.5.1 Advanced In Vitro Testing -- 1.5.2 Stem Cell Research -- 1.6 Immunogenicity -- 1.7 Integration and Validation -- 1.7.1 Use of Omics for Toxicology Testing -- 1.7.2 Integration of "New" Technologies into Risk Assessment -- 1.7.3 Use of Human-Derived Cellular Systems -- 1.7.4 "General" Acceptance - Translation into Guidelines -- 1.8 Research Initiative/Collaborations -- 1.9 Concluding Remarks -- References -- 2 In Silico Toxicology - Current Approaches and Future Perspectives to Predict Toxic Effects with Computational Tools -- 2.1 Introduction -- 2.2 Prediction of Hazard -- 2.2.1 Definition of Hazard and Its Use -- 2.2.2 Prediction of Mutagenicity -- 2.2.3 Prediction of Phospholipidosis -- 2.2.4 Prediction of Carcinogenicity -- 2.2.5 Prediction of Skin Sensitization -- 2.2.6 Prediction of Skin and Eye Irritation -- 2.2.7 Approaches to Systemic Toxicity Prediction -- 2.2.7.1 The Cramer Classes -- 2.2.7.2 Predicting Toxic Doses of Drugs -- 2.2.7.3 Predicting Organ Toxicity -- 2.2.7.4 Adverse Outcome Pathways and Potential for Prediction -- 2.3 Prediction of Risk -- 2.3.1 Risk Definition and Some Basic Considerations -- 2.3.2 Data Availability -- 2.3.3 Database Structure and Data Curation -- 2.3.4 Approaches to Model and Predict Risk -- 2.4 Thoughts on Validation -- 2.5 Conclusions and Outlook -- References.

3 In Silico Approaches: Data Management - Bioinformatics -- 3.1 Introduction -- 3.2 Experimental Setup and Statistical Power -- 3.3 Properties of Different Omics Data -- 3.3.1 Next-Generation Sequencing Data -- 3.3.2 DNA Methylation Data -- 3.3.3 miRNA Data -- 3.3.4 CNV and SNP Data -- 3.3.5 ChIP-seq Data -- 3.3.6 Gene Expression Microarray Data (Affymetrix) -- 3.3.7 Mass Spectrometry Data -- 3.3.8 Missing Values and Zero Values -- 3.3.9 Data Normalization -- 3.4 Statistical Methods -- 3.4.1 Data Overviews -- 3.4.2 Null Hypothesis/Type I and Type II Errors -- 3.4.3 Multiple Testing Methods -- 3.4.4 Statistical Tests -- 3.4.5 Linear Models and Linear Mixed Models -- 3.5 Prediction and Classification -- 3.5.1 Overview -- 3.5.2 Generating a Reference Compendium of Compounds -- 3.5.3 Cross-Validation -- 3.5.4 Selection Bias -- 3.6 Combining Different Omics Data and Biological Interpretations -- 3.7 Data Management -- References -- 4 Role of Modeling and Simulation in Toxicology Prediction -- 4.1 Introduction -- 4.2 The Need to Bring PK and PD in Predictive Models Together -- 4.2.1 Physiologically Based Pharmacokinetic Modeling -- 4.2.2 Mathematical (PBPK, PK/PD) Modeling -- 4.2.3 Predictive Tools -- 4.3 Methodological Aspects and Concepts -- 4.3.1 "Cascading" Drug Effects -- 4.3.2 Linking Exposure and Effect -- 4.3.3 Receptor Occupancy/Enzyme Inhibition -- 4.3.4 Transduction into In Vivo Response -- 4.3.4.1 Indirect Response Models -- 4.3.4.2 Transit Compartment Models -- 4.3.5 Disease Modeling -- 4.4 Application During Lead Optimization -- 4.4.1 Example 1: PK/PD Modeling for Identifying the Therapeutic Window between an Efficacy and a Safety Response -- 4.5 Application During Clinical Candidate Selection -- 4.5.1 Example 2: Translational PK/PD Modeling to Support Go/No Go Decisions -- 4.6 Entry-into-Human Preparation and Translational PK/PD Modeling.

4.6.1 Selection of Safe and Pharmacologically Active Dose for Anticancer Drugs -- 4.6.1.1 Example 3 -- 4.6.1.2 Example 4 -- 4.6.2 PK/PD for Toxicology Study Design and Evaluation -- 4.6.2.1 Example 5 -- 4.6.2.2 Example 6 -- 4.6.2.3 Example 7 -- 4.7 Justification of Starting Dose, Calculation of Safety Margins, and Support of Phase I Clinical Trial Design -- 4.8 Outlook and Conclusions -- References -- 5 Genomic Applications for Assessing Toxicities of Liver and Kidney Injury -- 5.1 Introduction -- 5.1.1 Toxicogenomics in Drug Development -- 5.2 Toxicogenomic Approaches -- 5.2.1 High-Throughput Expression Profiles and DNA Microarrays -- 5.2.2 Data Analysis -- 5.3 Specific Applications of Toxicogenomics -- 5.3.1 Mechanistic Toxicogenomics and Risk Assessment -- 5.3.2 Toxicogenomic Profiling of Hepatotoxicity -- 5.3.2.1 Hepatotoxicity in Drug Development -- 5.3.3 Functional and Structural Properties of the Liver -- 5.3.4 Liver Morphology -- 5.3.5 Cell Types -- 5.3.6 Functional Gradients -- 5.4 Toxicogenomic Applications for the Better Understanding of Hepatotoxicity -- 5.4.1 Mechanistic Toxicology -- 5.4.2 Class Identification -- 5.4.3 Predictive Toxicology -- 5.4.4 In Vitro Classifiers of Hepatotoxicity -- 5.4.5 Biomarker Identification -- 5.5 Toxicogenomic Profiling of Nephrotoxicity -- 5.5.1 Toxicogenomic Approaches in Nephrotoxicity -- 5.5.2 Finding Genes that Matter in AKI -- 5.5.3 Searching for New Biomarkers of Kidney Injury -- 5.6 Limitations of Toxicogenomics -- 5.6.1 Idiosyncrasies -- 5.6.2 Epigenetics -- 5.7 Conclusions -- References -- 6 Use of Toxicogenomics for Mechanistic Characterization of Hepatocarcinogens in Shorter Term Studies -- 6.1 Introduction -- 6.1.1 Rodent Carcinogenicity Testing -- 6.1.2 Classes of Carcinogens -- 6.2 Toxicogenomics.

6.2.1 Mechanistic Toxicogenomic Analysis after Short-Term Treatment with Rodent Hepatocarcinogens -- 6.2.2 Approaches for Prediction of Potential Hepatocarcinogens Based on Gene Expression Profiling -- 6.2.3 Recent Developments: Transcriptional Benchmark Dose Modeling Based on Functional Analyses -- 6.2.4 Recent Opportunities: Publicly Available Data -- 6.3 Conclusions and Outlook -- References -- 7 Discovery and Application of Novel Biomarkers -- 7.1 Introduction -- 7.1.1 New Technologies Give Rise to Novel Opportunities for Biomarker Discovery -- 7.2 Novel RNA Biomarkers -- 7.2.1 The Complex RNA Biomarker in Cancer -- 7.2.2 The Complex RNA Biomarker in Toxicology -- 7.2.3 Connectivity Mapping with the Complex RNA Biomarker for Hazard Identification -- 7.2.4 miRNA Biomarkers -- 7.3 DNA as a Biomarker -- 7.3.1 DNA Polymorphisms as Future Biomarkers of Disease and Xenobiotic Susceptibility -- 7.3.2 DNA and Protein Adduct Biomarkers -- 7.3.3 Epigenetic Biomarkers -- 7.4 Novel Biomarkers: Beyond Nucleotide-Based Discovery -- 7.5 Summary and Outlook -- References -- 8 Predictive Toxicology: Genetics, Genomics, Epigenetics, and Next-Generation Sequencing in Toxicology -- 8.1 Introduction -- 8.2 Technological Advances -- 8.3 Applications in Toxicology -- 8.3.1 Genome Sequencing and Sequence Level Comparisons -- 8.3.2 Genotype and Metabolism -- 8.3.3 Mechanistic Toxicology and Toxicogenomics -- 8.3.4 Epigenetic Changes and miRNAs -- 8.4 Summary and Outlook -- References -- 9 Biomarkers as Tools for Predictive Safety Assessment: Novel Markers of Drug-Induced Kidney Injury -- 9.1 Need and Search for Novel Biomarkers of Kidney Injury -- 9.2 Urinary Biomarkers of Drug-Induced Kidney Injury -- 9.2.1 Structure and Function of Novel Urinary Biomarkers -- 9.2.1.1 Kidney Injury Molecule-1 -- 9.2.1.2 Clusterin -- 9.2.1.3 Cystatin C -- 9.2.1.4 β2-Microglobulin.

9.2.1.5 Liver-Type Fatty Acid Binding Protein -- 9.2.1.6 Neutrophil Gelatinase-Associated Lipocalin -- 9.2.1.7 Others -- 9.2.2 Experimental and Clinical Support for the Use of Novel Urinary Biomarkers for the Detection and Prediction of Acute Kidney Injury -- 9.2.2.1 Performance of Novel Urinary Biomarkers in Preclinical Models of Renal Injury -- 9.2.2.2 Clinical Support for Novel Urinary Kidney Injury Biomarkers -- 9.3 Genomic Biomarkers -- 9.3.1 Individual Genes -- 9.3.2 Biomarker Panels and Gene Signatures -- 9.3.3 MicroRNAs -- 9.4 Qualification and Use of Novel Kidney Injury Biomarkers in Preclinical Safety Assessment -- 9.4.1 Biomarker Qualification and Regulatory Acceptance -- 9.4.2 Application of Novel Renal Safety Markers to Preclinical Decision Making -- 9.4.3 Technological Aspects -- 9.5 Summary and Perspectives -- References -- 10 The Use of Renal Cell Culture for Nephrotoxicity Investigations -- 10.1 Introduction -- 10.2 In Vitro Renal Models -- 10.2.1 Characterization -- 10.2.2 Immortalization of Primary Cells -- 10.2.3 Available Podocyte and Proximal Tubule Cell Lines -- 10.3 Stem Cells -- 10.4 Optimal Cell Culture Conditions -- 10.5 In Vitro Nephrotoxicity Assessment -- 10.6 Outlook -- References -- 11 The Zebrafish Model in Toxicology -- 11.1 The Need for a Physiologically Relevant Organ Model in Drug Toxicity Testing -- 11.2 Extensive Knowledge about Genetics, Development, and Physiology of D. rerio -- 11.3 Studies of Specific Organ Toxicities in Zebrafish Embryos and Larvae -- 11.3.1 Cardiotoxicity -- 11.3.2 Neurotoxicity -- 11.3.3 Hepatotoxicity -- 11.3.4 Teratogenicity -- 11.3.5 Future Directions: ADME Studies and Future Explorative Research -- 11.3.5.1 Absorption and Distribution -- 11.3.5.2 Metabolism -- 11.3.5.3 Harmonization and Validation -- 11.3.5.4 Future Explorative Research -- References.

12 Predictive Method Development: Challenges for Cosmetics and Genotoxicity as a Case Study.
Abstract:
Tailored to the needs of scientists developing drugs, chemicals, cosmetics and other products this one-stop reference for medicinal chemists covers all the latest developments in the field of predictive toxicology and its applications in safety assessment. With a keen emphasis on novel approaches, the topics have been tackled by selected expert scientists, who are familiar with the theoretical scientific background as well as with the practical application of current methods. Emerging technologies in toxicity assessment are introduced and evaluated in terms of their predictive power, with separate sections on computer predictions and simulation methods, novel in vitro systems including those employing stem cells, toxicogenomics and novel biomarkers. In each case, the most promising methods are discussed and compared to classical in vitro and in vivo toxicology assays. Finally, an outlook section discusses such forward-looking topics as immunotoxicology assessment and novel regulatory requirements. With its wealth of methodological knowledge and its critical evaluation of modern approaches, this is a valuable guide for toxicologists working in pharmaceutical development, as well as in safety assessment and the regulation of drugs and chemicals.
Local Note:
Electronic reproduction. Ann Arbor, Michigan : ProQuest Ebook Central, 2017. Available via World Wide Web. Access may be limited to ProQuest Ebook Central affiliated libraries.
Electronic Access:
Click to View
Holds: Copies: