Menu Expand
Computational Systems Pharmacology and Toxicology

Computational Systems Pharmacology and Toxicology

Rudy J Richardson | Dale E Johnson

(2017)

Additional Information

Book Details

Abstract

The network approaches of systems pharmacology and toxicology serve as early predictors of the most relevant screening approach to pursue both in drug discovery and development and ecotoxicological assessments. Computational approaches have the potential to improve toxicological experimental design, enable more rapid drug efficacy and safety testing and also reduce the number of animals used in experimentation. Rapid advances in availability of computing technology hold tremendous promise for advancing applied and basic science and increasing the efficiency of risk assessment.
This book provides an understanding of the basic principles of computational toxicology and the current methods of predictive toxicology using chemical structures, toxicity-related databases, in silico chemical-protein docking, and biological pathway tools. The book begins with an introduction to systems pharmacology and toxicology and computational tools followed by a section exploring modelling adverse outcomes and events. The second part of the book covers the discovery of protein targets and the characterisation of toxicant-protein interactions. Final chapters include case studies and additionally discuss interactions between phytochemicals and Western therapeutics.
This book will be useful for scientists involved in environmental research and risk assessment. It will be a valuable resource for postgraduate students and researchers wishing to learn about key methods used in studying biological targets both from a toxicity and pharmacological activity standpoint.


Dr Dale E. Johnson is currently an Adjunct Professor in Molecular Toxicology at UC Berkeley and an Adjunct Professor in Environmental Health Sciences at the University of Michigan. He teaches computational toxicology, drug discovery & development, and lectures in pharmacogenomics. He is also President and CEO of Emiliem, Inc. and CEO of Elara Bioscience LLC, a software company. He also consults for biopharmaceutical companies and investment firms. Dr Johnson has over 30 years’ experience as a research and development scientist, manager, executive, and entrepreneur in the biopharmaceutical field working in large pharmaceutical and biotech companies and several start-ups. Prior to Emiliem, he served as VP Drug Assessment & Development and VP Preclinical Development at Chiron Corporation and previously worked at Hoechst-Roussel Pharmaceuticals, International Research and Development Corp., Medical Research Division of American Cyanamid, Eos Biotechnology and Ddplatform. Dr Rudy J. Richardson is currently the Dow Professor of Toxicology at the University of Michigan. He also holds a joint appointment in the Neurology Department. Sabbatical leaves at Warner-Lambert/Parke-Davis (now Pfizer) in Ann Arbor and the University of Padua in Italy. Dr Richardson has served several times as the Director of the Toxicology Program, and currently he serves as Graduate Chair of the EHS Department. His research interests include computational toxicology, biomarkers and biosensors of exposure and disease, mechanisms of neurodegenerative disease, chemistry and toxicology of organophosphorus and organic nitro compounds, and interactions of toxicants with target macromolecules.


Table of Contents

Section Title Page Action Price
Cover Cover
Computational Systems Pharmacology and Toxicology i
Preface vii
Contents ix
Chapter 1 - Systems Biology Approaches in Pharmacology and Toxicology 1
1.1 Introduction 1
1.2 Systems Toxicology 2
1.3 Chemical Toxicities 3
1.3.1 Single-Target Toxicity Concepts 3
1.3.2 Toxicological Profiling for Potential Adverse Reactions 5
1.3.3 Toxicological Concepts for Safer Chemical Design 6
1.3.4 Biomarkers 8
1.4 Environmental Toxicology 9
1.4.1 Adverse Outcome Pathway 9
1.4.2 Expanding Exposure Concepts 10
1.4.3 Exposome 11
1.5 Systems and Network Pharmacology 12
1.5.1 Secondary Pharmacology and Off-Target Effects 13
1.5.2 Prediction of Potential Adverse Effects 14
1.6 Conclusions 14
References 14
Chapter 2 - Databases Facilitating Systems Biology Approaches in Toxicology 19
2.1 Introduction 19
2.2 Categorized Lists of Databases for Systems Toxicology 21
2.2.1 TOXNET Databases (Including Those with Direct Links from TOXNET) 21
2.2.2 US EPA Chemical Toxicity Databases 24
2.2.3 National Toxicology Program Databases 24
2.2.4 Additional Toxicity Databases 25
2.2.5 Chemical–Gene–Protein Databases 26
2.2.6 Pathway-Network Databases 27
2.2.7 Chemistry, Structural Alert, and QSAR Databases and Tools 28
2.2.8 Drug and Drug Target Databases 30
2.3 Websites with Extensive Links to Databases and Tools 31
2.4 Conclusions 31
References 32
Chapter 3 - Tools for Green Molecular Design to Reduce Toxicological Risk 36
3.1 Introduction 37
3.2 Physiochemical, Genotoxicity, and Blood–Brain Barrier Passage Properties of Chemicals 38
3.3 Tools for Green Molecular Design 39
3.3.1 Expert Systems 39
3.3.2 Decision Trees 44
3.3.3 QSAR Tools 44
3.3.4 Representative Tools 45
3.3.4.1 ACD Percepta (www.acdlabs.com) 45
3.3.4.2 ADMET Predictor (www.simulations-plus.com) 46
3.3.4.3 Medchem Designer 46
3.3.4.4 Derek and Meteor Nexus from Lhasa Limited (www.lhasalimited.org) 47
3.3.4.5 Qikprop (www.schrodinger.com/QikProp) 48
3.3.4.6 OECD QSAR Toolbox 48
3.3.4.7 Toxtree (http://toxtree.sourceforge.net/) 48
3.3.4.8 Chemaxon Suite (Marvin Sketch and Metabolizer) (www.chemaxon.com/) 49
3.3.4.9 Chemicalize (www.chemicalize.com) 49
3.3.4.10 AIM (Analog Identification Methodology) (http://www.epa.gov/tsca-screening-tools/analog-identification-methodology-aim-t... 49
3.3.4.11 Chemspider (www.chemspider.com) 49
3.3.4.12 Mobyle@RPBS (http://mobyle.rpbs.univ-paris-diderot.fr) 49
3.4 Case Study 50
3.5 The Design of Ideal Tools for Chemists 51
3.6 Conclusions 54
References 54
Chapter 4 - Linking Environmental Exposure to Toxicity† 60
4.1 Introduction 60
4.2 The AOP Framework: An Organizing Principle for Toxicological Data 64
4.2.1 AOP Knowledge Management 67
4.2.2 Phases of AOP Development 68
4.2.3 Data Resources for AOP Development 70
4.3 Environmental Exposure and Pharmacokinetic Considerations for Adverse Outcome Development 73
4.4 The AEP Framework: An Organizing Principle for Exposure Data 75
4.4.1 Data resources for AEP development 79
4.5 AEP–AOP Integration for Linking Toxicity to Exposure: Applications of the AOP and AEP Frameworks for Risk Assessments and Che... 80
4.6 Conclusions and Future Directions 82
References 83
Chapter 5 - Linking Drug or Phytochemical Exposure to Toxicity 89
5.1 Introduction 89
5.2 Pharmacokinetic and Toxicokinetic Models 91
5.2.1 Structural Models 91
5.2.1.1 Compartmental Models 92
5.2.1.2 Physiological Models 96
5.2.2 Variance Models 98
5.2.2.1 Other Modeling Methodologies 101
5.3 PK/PD Relationships 102
5.3.1 Mathematical Description of Pharmacodynamic Effects 104
5.3.1.1 Direct Effects and Effect Compartments 106
5.3.1.2 Indirect Effects 107
5.3.2 Combined PK/PD and TK/TD Modeling 109
5.3.3 Modeling Pharmacodynamics in the Absence of Pharmacokinetic Data: K-PD Models 110
5.4 Modeling Drug Interactions with Phytochemicals 111
5.4.1 Inhibition of Metabolism 112
5.4.2 Induction of Metabolism 113
5.4.3 Enhancement of Absorption 114
5.4.4 Inhibition of Absorption 115
5.4.5 Modeling of Pharmacodynamic Interactions 115
5.5 Conclusions 116
References 116
Chapter 6 - Chemical Similarity, Shape Matching and QSAR 120
6.1 Introduction 120
6.2 Molecular Similarity, Chemical Spaces and Activity Landscapes 121
6.2.1 Molecular Similarity: Concept and Definitions 121
6.2.1.1 Molecular Similarity Concept 121
6.2.1.2 Applicability of Molecular Similarity Measures 122
6.2.1.3 Structure Representations for Molecular Similarity Analysis 122
6.2.1.4 Molecular Similarity Functions 125
6.2.2 Chemical Spaces and Activity Landscapes 131
6.2.3 Applications of Molecular Similarity Analysis 138
6.2.3.1 Similarity-Based Virtual Screening 138
6.2.3.2 Activity Prediction 139
6.2.3.3 Clustering, Networks and Diversity 141
6.3 Quantitative Structure–Activity/Property Relationships (QSAR/QSPR) 142
6.3.1 Congeneric Series and Consistent Mechanisms 143
6.3.2 Diverse Series and Big Data 148
6.3.2.1 Prediction of ADMET Properties 150
6.3.2.2 Prediction of Potential Drug Targets 152
6.3.2.3 Prediction of Activity Towards Individual Targets 152
6.3.2.4 Prediction of Physico-Chemical Properties 153
6.3.2.5 Open Web-Based QSAR/QSPR Services 153
6.4 Conclusion 153
Acknowledgements 154
References 154
Chapter 7 - In silico Chemical–Protein Docking and Molecular Dynamics 174
7.1 Introduction 174
7.2 Molecular Docking: Overview and Applications 175
7.2.1 Genetic Algorithms 177
7.2.2 Monte Carlo Procedure 177
7.2.3 Matching Algorithms 177
7.3 Scoring Ligand Poses 178
7.4 Inverse Docking 179
7.5 Case Study: Using In silico Docking to Investigate Interactions of 1,3-Dinitrobenzene with Adenosine Deaminase 179
7.6 Case Study: Using In silico Docking to Assess Binding of Bisphenol-A to Estrogen-Related Receptor-γ 180
7.7 Molecular Dynamics 182
7.7.1 Running MD Simulations 182
7.7.2 Analysis of MD Trajectories 183
7.7.3 Case Study: Gaining Insights into the Conformational Dynamics of Human Neuropathy Target Esterase via MD Simulations of its... 185
References 187
Chapter 8 - Computational Tools for Chemical Toxicity Testing and Risk Assessment Under the Framework of Adverse Outcome Pathways 191
8.1 Introduction 191
8.2 The AOP Concept 193
8.3 Quantitative Methods in Traditional Apical Endpoints Testing 194
8.4 PBPK Modeling and In vitro to In vivo Extrapolation 196
8.5 SAR Modeling 197
8.6 Computational Modeling of Toxicity Pathways 198
8.6.1 Concept of Toxicity Pathways 198
8.6.2 Purpose of Modeling Toxicity Pathways 199
8.6.3 How to Model Toxicity Pathways 200
8.6.4 Case Studies 203
8.6.5 Education on Computational Toxicology 205
8.6.6 Pathway Modeling Software Tools 205
Acknowledgements 206
References 206
Chapter 9 - In silico Toxicology: An Overview of Toxicity Databases, Prediction Methodologies, and Expert Review 209
9.1 Introduction 209
9.2 Toxicity Databases 215
9.2.1 Overview 215
9.2.2 Database Organization 216
9.2.3 Genetic Toxicity and Carcinogenicity 217
9.2.4 Reproductive and Developmental Toxicity 219
9.2.5 Acute and Repeated Dose Toxicity 220
9.3 In silico Methodologies 221
9.3.1 Overview 221
9.3.2 Expert Alerts 221
9.3.3 QSARs 223
9.3.4 Read-Across 226
9.4 Expert Reviews 227
9.4.1 Assessing Experimental Data 227
9.4.2 Drawing Conclusions from Multiple Systems 228
9.4.3 Reviews Accepting or Refuting An In silico Result 230
9.4.4 Documenting In silico Results 233
9.5 Conclusions 233
Acknowledgements 236
References 236
Chapter 10 - Data Sources for Herbal and Traditional Medicines 243
10.1 Introduction 243
10.2 TCM Databases 244
10.2.1 Chem-TCM (Chemical Database of Traditional Chinese Medicine) 244
10.2.2 HIT (Linking Herbal Active Ingredients to Targets) 244
10.2.3 TCMSP (Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform) 246
10.2.4 TCMGeneDIT (A Database for Associated TCM, Gene and Disease Information Using Text Mining) 248
10.2.5 TCMID (Traditional Chinese Medicine Integrative Database for Herb Molecular Mechanism Analysis) 249
10.2.6 TTD (Therapeutic Target Database) 251
10.3 Omics Data in TCM 253
10.3.1 Genomics in TCM 253
10.3.2 Transcriptomics in TCM 254
10.3.3 Proteomics in TCM 256
10.3.4 Metabonomics in TCM 256
10.4 Summary 257
Acknowledgements 257
References 257
Chapter 11 - Network Pharmacology Research Approaches for Chinese Herbal Medicines 261
11.1 Introduction 261
11.1.1 Modernization of TCM 263
11.1.2 Concept of Network Pharmacology 263
11.2 Network Pharmacology in TCM Research 265
11.3 Network Pharmacology in the Understanding of Herb–Drug Interactions 268
11.4 Pharmacogenomics in TCM 269
11.5 TCMs in Clinical Trials 271
11.6 The Future of Network Pharmacology in Traditional Medicine 273
11.7 Conclusion 274
References 274
Chapter 12 - Chemical–Disease Category Linkage (CDCL): Computational Methods Linking Traditional Chinese Medicines and Western Therapeutics 279
12.1 Introduction 279
12.1.1 Databases for CDCL Information and Study 281
12.1.1.1 TCM and Chemical Constituents 282
12.1.1.2 TCM Classification and Systems Approach 282
12.1.1.3 Western Therapeutics 283
12.1.1.4 Therapeutic Targets and Protein Interactions 283
12.1.1.5 Pathway Analysis 283
12.1.2 TCM Classifications 283
12.1.3 Active Ingredients in Herbs 286
12.1.3.1 Wind Cold 286
12.1.3.2 Heat (Blood) 286
12.1.3.3 Tonify Qi 287
12.1.3.4 Tonify Blood 287
12.2 Open Access Tools for CDCL Informatics 287
12.3 Computational CDCL Studies with Commercial Tools 289
12.4 Herb–Drug Interactions 292
12.4.1 Pharmacokinetic Interactions 292
12.4.2 Pharmacogenomic-Related Interactions 294
12.5 Combination Therapies and Future Directions 294
12.6 Conclusions 295
References 296
Chapter 13 - Educational Programs for Computational Toxicology and Pharmacology 300
13.1 Introduction 300
13.2 Historical Context: Computational Toxicology 301
13.2.1 Background 301
13.2.2 Programs at University of California Berkeley and University of Michigan 302
13.3 Inquiry-Based Science Courses 303
13.4 Current Computational Toxicology Courses 304
13.4.1 Toxicology Tutorials 305
13.4.2 Course Concepts 305
13.4.3 Case Studies 306
13.4.3.1 Chemical Structural Features Determine Biological Effects 306
Case Study Example 1 306
Case Study Example 2.There is wide agreement that chemicals of great concern are those that: persist (P), bioaccumulate (B), and... 307
Case Study Example 3 307
Case Study Example 4 308
Typical Report for Case Studies (One Per Team if Applicable) 308
13.4.3.2 Herbal Traditional Medicines 308
Case Study Example 5 308
Case Study Example 6 309
13.4.3.3 Environmental Chemicals and Health Relationships 309
Case Study Example 7.A current topic is selected that recently appeared in the scientific literature or the news media. This top... 309
13.5 Course Projects 310
13.5.1 Starting Projects 310
13.5.2 Typical Project Categories 310
13.5.3 Therapeutics vs. Environmental Chemicals 311
13.5.4 Challenges in Computational Toxicology 311
13.5.4.1 Hazard-Based Information Gathering 311
13.6 Sample Project: The Chemical of Concern Question 312
13.6.1 Health Effects Inquiry 313
13.6.2 Endpoints for Breast Cancer 313
13.6.3 Project Question 314
13.7 Course Projects Presented and Published 314
13.7.1 Projects Presented at National Scientific Meetings 314
13.7.1.1 Society of Environmental Toxicology and Chemistry (SETAC) 314
13.7.1.2 National Society of Toxicology Through 2015 314
13.7.2 Projects from the Courses Published in Journals 316
13.8 Computational Pharmacology as Part of the Principles of Drug Action 316
13.9 Conclusion 318
References 319
Subject Index 324