Menu Expand
High Throughput Screening Methods

High Throughput Screening Methods

Joshua A Bittker | Nathan T Ross

(2016)

Abstract

High throughput screening remains a key part of early stage drug and tool compound discovery, and methods and technologies have seen many fundamental improvements and innovations over the past 20 years. This comprehensive book provides a historical survey of the field up to the current state-of-the-art. In addition to the specific methods, this book also considers cultural and organizational questions that represent opportunities for future success.
Following thought-provoking foreword and introduction from Professor Stuart Schreiber and the editors, chapters from leading experts across academia and industry cover initial considerations for screening, methods appropriate for different goals in small molecule discovery, newer technologies that provide alternative approaches to traditional miniaturization procedures, and practical aspects such as cost and resourcing. Within the context of their historical development, authors explain common pitfalls and their solutions.
This book will serve as both a practical reference and a thoughtful guide to the philosophy underlying technological change in such a fast-moving area for postgraduates and researchers in academia and industry, particularly in the areas of chemical biology, pharmacology, structural biology and assay development.


Table of Contents

Section Title Page Action Price
Cover Cover
Contents xi
Foreword: Transforming Medicine by Innovating the Science of Therapeutics v
Preface vii
Chapter 1 HTS Methods: Assay Design and Optimisation 1
1.1 Introduction 1
1.2 HTS at AstraZeneca 2
1.2.1 Criteria and Acceptance 3
1.2.2 Robustness/Reliability 4
1.2.3 Analysing Data to Define Robustness/Reliability 8
1.2.4 As Simple to Run as Possible 9
1.2.5 Assay Validation 10
1.3 Summary 14
References 14
Chapter 2 Considerations Related to Small-molecule Screening Collections 16
2.1 Introduction 16
2.2 General Considerations Related to HTS Compound Collections 17
2.2.1 Determination of Screening Objectives 18
2.2.2 Size of HTS Compound Collections 18
2.2.3 Chemical Diversity in Compound Collections 19
2.2.4 Quality of Compounds in Screening Collections 19
2.3 Sources of Compounds in Screening Collections 20
2.3.1 Natural Products in Screening Collections 20
2.3.2 Synthetic Drug-like Compounds 23
2.3.3 Diverse Collections 26
2.4 Performance of Compounds in Screening Collections 29
2.4.1 Background 29
2.4.2 Performance of Compounds from Different Sources 29
2.4.3 Performance Diversity of Compound Collections 31
2.4.4 Pan Assay Interference Compounds 32
2.4.5 Dark Chemical Matter 33
2.5 Conclusions and Discussion 33
References 34
Chapter 3 Combination Screening 37
3.1 Introduction 37
3.2 Measures of Synergy 38
3.2.1 Bliss Independence Model of Synergy 39
3.2.2 Loewe Additivity 40
3.2.3 Other Measures 43
3.2.4 Reconciling Measures of Synergy 46
3.3 Design of Combination Experiments 47
3.4 Statistical Inference of Combination Effects 49
3.4.1 The Error Distribution 50
3.4.2 Bootstrap Intervals 51
3.4.3 Intervals for Bliss Independence 52
3.4.4 Intervals for Loewe Additivity Interaction Index 53
3.5 Null Hypothesis Significance Testing 54
3.5.1 Significance Testing of Bliss 54
3.5.2 Significance Test of Loewe Additivity 55
3.6 Concluding Remarks 56
References 56
Chapter 4 Modern Biophysical Methods for Screening and Drug Discovery 58
4.1 Introduction 58
4.2 Physicochemical Properties and High Concentration Screening 59
4.2.1 Physicochemical Properties of Chemical Libraries 59
4.2.2 High Concentration Screening 63
4.3 Differential Scanning Fluorimetry 64
4.4 Surface Plasmon Resonance 65
4.5 Mass Spectrometry Techniques 67
4.5.1 Affinity Selection MS 68
4.5.2 Affinity Chromatography Methods 69
4.5.3 Protein MS 70
4.6 NMR Spectroscopy 70
4.6.1 Protein NMR 72
4.6.2 Ligand Observed NMR 73
4.7 Calorimetric Methods 75
4.7.1 Differential Scanning Calorimetry 76
4.7.2 Isothermal Titration Calorimetry 77
4.8 X-Ray Crystallography 77
4.9 Newer Methods on the Horizon 78
4.10 Summary and Recommendations 79
Acknowledgments 80
References 80
Chapter 5 Genetic Perturbation Methods, from the ‘Awesome Power' of Yeast Genetics to the CRISPR Revolution 87
5.1 Introduction 87
5.2 Genetic Methodologies 88
5.2.1 Random Mutagenesis 88
5.2.2 Targeted Genome-wide Deletions (Homozygous/Heterozygous) 90
5.2.3 Random Genome-wide Deletions (Homozygous/Heterozygous) 91
5.2.4 RNA Interference 92
5.2.5 CRISPR 95
5.2.6 Overexpression 104
5.2.7 Synthetic Biology 104
5.3 Concluding Remarks 105
Acknowledgments 105
References 105
Chapter 6 Understanding Luminescence Based Screens 117
6.1 Why Luminescence? An Introduction to Bioluminescent and Chemiluminescent Assays 117
6.1.1 Overview of Common Luminescent Enzymes Employed in Assays 119
6.1.2 Chemiluminescence in HTS Assays 122
6.2 Considerations and Applications of Bioluminescent Assays 123
6.2.1 Prevalence of Luciferase Inhibitors in Compound Libraries 123
6.2.2 Mechanisms of Luciferase Inhibition 124
6.2.3 Ligand Based Stabilization of Luciferases: Impact on RGA Results 129
6.2.4 Methods to Mitigate Luciferase Inhibitors in RGAs: Counter-Screens and Orthogonal Assay Formats 131
6.2.5 Luciferases as Post-translational Sensors 133
6.2.6 Use of Luciferases in Biochemical Applications 134
6.3 Considerations and Applications of Amplified Luminescent Proximity Homogenous Assays: AlphaScreen and AlphaLISA 135
6.3.1 Example Protocols and Key Experiments 135
6.3.2 Interferences with ALPHA Technology 138
6.4 Conclusion 138
References 139
Chapter 7 High Throughput Screening Compatible Methods for Quantifying Protein Interactions in Living Cells 143
7.1 Introduction 143
7.2 Analysis of PPIs in Intact Cells 145
7.2.1 Two-hybrid Systems 145
7.2.2 Protein Fragment Complementation Technologies 147
7.2.3 FRET for Analysis of PPIs 150
7.3 Measuring Target Engagement in Cells 153
7.3.1 Target Engagement by Protein Stability Analysis 153
7.3.2 Real Time, Quantitative Analysis of Target Engagement via FRET 155
7.4 Outlook 159
References 159
Chapter 8 Approaches to High Content Imaging and Multi-feature Analysis 162
8.1 Introduction 162
8.2 Imaging Hardware 164
8.3 Image Analysis 169
8.4 Quality Control of Image Acquisition and Well Level Data 170
8.4.1 Quality Control of Image Acquisition 170
8.4.2 Quality Control of Well Level Data 171
8.5 Single Cell Analysis 171
8.6 Analysis of Multiparametric Data 173
8.6.1 Feature Selection and Dimensional Reduction 174
8.6.2 Distance and Similarity 174
8.7 Machine Learning: Supervised and Unsupervised Methods 176
8.7.1 Supervised Learning (Classification) 176
8.7.2 Unsupervised Learning (Clustering) 178
8.8 Conclusion 178
References 178
Chapter 9 Pharmacological and Genetic Screening of Molecularly Characterized Cell Lines 181
9.1 Introduction 181
9.2 Cell Lines 182
9.2.1 Applications in Cancer 182
9.2.2 Mistaken Identities 183
9.2.3 Authentication 184
9.2.4 Molecular Characterizations 184
9.3 In vitro Drug Screening towards Precision Medicine 185
9.3.1 Drug Sensitivity Studies 185
9.3.2 Molecular Predictors of Drug Response 190
9.3.3 Datasets 192
9.3.4 Drug Perturbation Studies 196
9.3.5 Genetic Perturbation 200
9.4 Conclusions and Outlook 204
References 205
Chapter 10 Multidimensional Profile Based Screening: Understanding Biology through Cellular Response Signatures 214
10.1 Introduction 214
10.2 Multidimensional Profiling through Multi-feature Measurement in a Single System 216
10.2.1 LINCS Concept 216
10.2.2 Gene Expression Profiling: CMap 217
10.2.3 Proteomics Profiling and Other Measurements 218
10.2.4 Cell Morphology Profiling: Cell Painting 220
10.3 Multidimensional Profiling through Single Feature Measurement in Multiple Systems 222
10.3.1 Expanding on the NCI-60: Building a Comprehensive Cell Viability Profile Matrix 223
10.3.2 Current Logistical Constraints in Screening: Large Numbers of Cell Lines or Large Numbers of Compounds? 223
10.3.3 PRISM: Multiplexed Cancer Cell Line HTS 224
10.4 Analysis Approaches for using Cell Based Profiles to Identify Mechanism of Action 225
10.4.1 NCI-60 COMPARE 227
10.4.2 CMap: Computational Aspects 227
10.5 Comprehensive Cell Line Profiling Requires Comprehensive Reference Annotations 231
10.5.1 Importance of Compound Annotations 231
10.5.2 Importance of Cell Line Annotations and Quality Control 232
10.6 Summary and Needs for the Future 234
10.6.1 Increasing the Number of Cell Lines for Screening and Call for Common Quality Control 234
10.6.2 Increasing the Perturbagen Dimension to include Genetic Perturbagens 235
10.7 Conclusion 235
Acknowledgments 236
References 236
Chapter 11 3D Cell Culture and Dish Based Organogenesis: Optimizing In vitro Cellular Physiology 239
11.1 Introduction 239
11.2 3D Culture Gels, Scaffolds and Bioprinting 240
11.2.1 Hydrogels 240
11.2.2 Scaffolds 241
11.2.3 3D Bioprinting 243
11.3 3D Culture and Drug Discovery 244
11.4 Organs-on-Chips 246
11.5 3D Cell Culture Limitations 248
11.6 Conclusion 249
References 249
Chapter 12 Small-molecule-mediated Targeted Protein Degradation for Drug Discovery 252
12.1 Introduction 252
12.2 E3-ligase-targeting Heterobifunctional Degraders 253
12.2.1 The State of the Art 253
12.2.2 CRBN-recruiting Heterobifunctional Degraders 254
12.2.3 VHL-recruiting Heterobifunctional Degraders 257
12.2.4 The Generality of the Bifunctional Degrader Approach 259
12.2.5 Towards the Mechanism of Action of Bifunctional Degraders 260
12.2.6 Towards a Therapeutic 261
12.2.7 Scope and Limitations 262
12.3 Alternative Approaches to Small-molecule-mediated Targeted Protein Degradation 266
12.3.1 Hydrophobic Tag 267
12.3.2 Chaperone Mediated Autophagy 267
12.3.3 Molecular Glue Degraders 268
12.4 Identifying Suitable Targets for Small-molecule-mediated Protein Degradation 269
12.5 Conclusion and Outlook 269
References 270
Chapter 13 Phenotypic Screens with Model Organisms 275
13.1 Introduction to Large-scale Screens 275
13.2 What Animal Models to Choose for a Screen? 276
13.3 Problems and Questions Addressed with Phenotypic Screens 282
13.3.1 Genetic Screens 282
13.3.2 Small Bioactive Molecule Screens 283
13.3.3 Genome Editing Assisted Targeted Screens 284
13.3.4 Summary 285
13.4 The Output of Large Scale Screens 286
13.4.1 Features of Different Screen Models 286
13.4.2 Behavioral Readout: Motion Index versus Complex Behavior 286
13.4.3 Physiological Readout: Electrophysiology and Imaging 288
13.5 Criteria, Sensitivity, Performance and Visualization of the Screen 290
13.5.1 Defining a ‘‘Hit\": Variance and Dimensionality 290
13.5.2 Assessing, Visualizing and Ranking Performance 291
13.5.3 Current Limitations and the Future 294
Acknowledgments 294
References 294
Chapter 14 Encoded Compound Libraries to Accelerate Small-molecule Therapeutic Discovery 303
14.1 Introduction 303
14.2 Combinatorial Chemistry 304
14.3 Encoding of Compound Libraries 307
14.3.1 First-generation Encoding 308
14.3.2 Second-generation Encoding 310
14.3.3 DNA Encoded Small-molecule Libraries 311
14.3.4 Associated Synthesis and Encoding 312
14.3.5 Disassociated Synthesis and Encoding 313
14.4 Screening of DNA Encoded Compound Libraries 315
14.5 Data Processing and Analysis 318
14.6 Conclusion 320
References 321
Chapter 15 Research Data Management 324
15.1 Introduction 324
15.2 Evolution of Research Data Management 326
15.3 Principles for Data Management in the 21st Century 328
15.4 Assembling Data from the Data Ecosystem 331
15.5 Annotating, Curating and Describing Data Derived from the Data Ecosystem 332
15.6 Predictive Analytics 335
15.7 Towards Understanding: Moving from Prediction to Action 338
15.8 Lessons Learned 340
15.9 Conclusion 340
References 340
Chapter 16 Small-molecule Bioactivity Databases 344
16.1 Introduction 344
16.2 Public Bioactivity Databases 345
16.2.1 BindingDB 347
16.2.2 PubChem 347
16.2.3 ChEMBL 351
16.2.4 GtoPdb 354
16.2.5 Public Data in the CDD Vault 354
16.3 Data Quality 360
16.4 Conclusions 364
Acknowledgments 365
References 365
Chapter 17 ‘‘So You Want to Run a High-throughput Screen: Do You Know How Much That Costs?\"; Costs of High Throughput Screens and How to Fund Them 372
17.1 Introduction: Planning for High Throughput Screening 372
17.2 Costs Associated with HTS 373
17.2.1 Do not Forget the Overheads 373
17.2.2 Example of a Budget for a High Throughput Screen 374
17.2.3 Beyond the HTS: Secondary and Tertiary Assays 377
17.2.4 Improving Cost Efficiency of HTS Execution 377
17.3 Organizational Models 379
17.3.1 Academic Core Facilities 379
17.3.2 Pharmaceutical Screening Facilities 382
17.3.3 Contract Research Organizations 383
17.3.4 Balancing Innovation and Economies of Scale 384
17.4 Funding Opportunities 384
17.4.1 Government Grants for Screening 385
17.4.2 Philanthropic Societies 385
17.4.3 Industrial Collaborations 386
17.4.4 Crowdsourcing 387
17.5 Future Outlook for HTS Funding 387
Acknowledgments 388
References 388
Subject Index 390