Frequent hitters: nuisance artifacts in high-throughput screening.

One of the major challenges in early drug discovery is the recognition of frequent hitters (FHs), that is, compounds that nonspecifically bind to a range of macromolecular targets or false positives caused by various types of assay interference. In this review, we survey the mechanisms underlying different types of FH, including aggregators, spectroscopic interference compounds (i.e., luciferase inhibitors and fluorescent compounds), reactive compounds, and promiscuous compounds. We also review commonly used experimental detection techniques and computational prediction models for FH identification. In addition, the rational applications of these computational filters are discussed. It is believed that, with the rational use of FH filters, the efficiency of drug discovery will be significantly improved.

[1]  Renaldo Mendoza,et al.  ALARM NMR: a rapid and robust experimental method to detect reactive false positives in biochemical screens. , 2005, Journal of the American Chemical Society.

[2]  R. Solé,et al.  Data completeness—the Achilles heel of drug-target networks , 2008, Nature Biotechnology.

[3]  Balaguru Ravikumar,et al.  Improving the efficacy-safety balance of polypharmacology in multi-target drug discovery , 2018, Expert opinion on drug discovery.

[4]  Igor V. Tetko,et al.  Identification of Small-Molecule Frequent Hitters from AlphaScreen High-Throughput Screens , 2014, Journal of biomolecular screening.

[5]  B. Shoichet,et al.  A common mechanism underlying promiscuous inhibitors from virtual and high-throughput screening. , 2002, Journal of medicinal chemistry.

[6]  Anne Mai Wassermann,et al.  Dark chemical matter as a promising starting point for drug lead discovery. , 2015, Nature chemical biology.

[7]  Paul J Hergenrother,et al.  Deoxynyboquinones as NQO1-Activated Cancer Therapeutics. , 2015, Accounts of chemical research.

[8]  Scott B Ficarro,et al.  A structure-guided approach to creating covalent FGFR inhibitors. , 2010, Chemistry & biology.

[9]  F. Nan,et al.  Isoquinoline-1,3,4-trione Derivatives Inactivate Caspase-3 by Generation of Reactive Oxygen Species* , 2008, Journal of Biological Chemistry.

[10]  Luhua Lai,et al.  LigBuilder 2: A Practical de Novo Drug Design Approach , 2011, J. Chem. Inf. Model..

[11]  Anton Simeonov,et al.  Firefly luciferase in chemical biology: a compendium of inhibitors, mechanistic evaluation of chemotypes, and suggested use as a reporter. , 2012, Chemistry & biology.

[12]  Jürgen Bajorath,et al.  Data structures for computational compound promiscuity analysis and exemplary applications to inhibitors of the human kinome , 2019, Journal of Computer-Aided Molecular Design.

[13]  B. Shoichet,et al.  Identification and prediction of promiscuous aggregating inhibitors among known drugs. , 2003, Journal of medicinal chemistry.

[14]  Robert W. Bryant,et al.  Evaluation of Fluorescent Compound Interference in 4 Fluorescence Polarization Assays: 2 Kinases, 1 Protease, and 1 Phosphatase , 2003, Journal of biomolecular screening.

[15]  Dan Li,et al.  Prediction of luciferase inhibitors by the high-performance MIEC-GBDT approach based on interaction energetic patterns. , 2017, Physical chemistry chemical physics : PCCP.

[16]  Feng-Xu Wu,et al.  ACFIS: a web server for fragment-based drug discovery , 2016, Nucleic Acids Res..

[17]  Adrian Whitty,et al.  The resurgence of covalent drugs , 2011, Nature Reviews Drug Discovery.

[18]  T. Hunter,et al.  The Protein Kinase Complement of the Human Genome , 2002, Science.

[19]  Olivier Sperandio,et al.  FAF-Drugs3: a web server for compound property calculation and chemical library design , 2015, Nucleic Acids Res..

[20]  J Stables,et al.  Development of a dual glow-signal firefly and Renilla luciferase assay reagent for the analysis of G-protein coupled receptor signalling. , 1999, Journal of receptor and signal transduction research.

[21]  Peter Wipf,et al.  Profiling the NIH Small Molecule Repository for compounds that generate H2O2 by redox cycling in reducing environments. , 2010, Assay and drug development technologies.

[22]  Alexander Tropsha,et al.  Phantom PAINS: Problems with the Utility of Alerts for Pan-Assay INterference CompoundS , 2017, J. Chem. Inf. Model..

[23]  Brian K Shoichet,et al.  Interpreting steep dose-response curves in early inhibitor discovery. , 2006, Journal of medicinal chemistry.

[24]  Yanli Wang,et al.  A novel method for mining highly imbalanced high-throughput screening data in PubChem , 2009, Bioinform..

[25]  Kristin E. D. Coan,et al.  Stoichiometry and physical chemistry of promiscuous aggregate-based inhibitors. , 2008, Journal of the American Chemical Society.

[26]  Jayme L. Dahlin,et al.  Assay Interference by Chemical Reactivity , 2015 .

[27]  Adam Yasgar,et al.  Quantitative high-throughput screening: a titration-based approach that efficiently identifies biological activities in large chemical libraries. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[28]  Nils-Ole Friedrich,et al.  Hit Dexter: A Machine‐Learning Model for the Prediction of Frequent Hitters , 2018, ChemMedChem.

[29]  Christopher P Austin,et al.  Characterization of chemical libraries for luciferase inhibitory activity. , 2008, Journal of medicinal chemistry.

[30]  G. Rishton Reactive compounds and in vitro false positives in HTS , 1997 .

[31]  Brian K Shoichet,et al.  A detergent-based assay for the detection of promiscuous inhibitors , 2006, Nature Protocols.

[32]  Jürgen Bajorath,et al.  How Frequently Are Pan-Assay Interference Compounds Active? Large-Scale Analysis of Screening Data Reveals Diverse Activity Profiles, Low Global Hit Frequency, and Many Consistently Inactive Compounds. , 2017, Journal of medicinal chemistry.

[33]  Brian K. Shoichet,et al.  Colloidal Drug Formulations Can Explain “Bell-Shaped” Concentration–Response Curves , 2014, ACS chemical biology.

[34]  D. Auld,et al.  Illuminating insights into firefly luciferase and other bioluminescent reporters used in chemical biology. , 2010, Chemistry & biology.

[35]  Johannes Kirchmair,et al.  Hit Dexter 2.0: Machine-Learning Models for the Prediction of Frequent Hitters , 2019, J. Chem. Inf. Model..

[36]  P Schneider,et al.  Spotting and designing promiscuous ligands for drug discovery. , 2016, Chemical communications.

[37]  Wolfgang Guba,et al.  Development of a virtual screening method for identification of "frequent hitters" in compound libraries. , 2002, Journal of medicinal chemistry.

[38]  Evan Bolton,et al.  PubChem 2019 update: improved access to chemical data , 2018, Nucleic Acids Res..

[39]  D. Boschelli,et al.  4-anilino-3-quinolinecarbonitriles: an emerging class of kinase inhibitors. , 2002, Current topics in medicinal chemistry.

[40]  Tudor I. Oprea,et al.  Badapple: promiscuity patterns from noisy evidence , 2016, Journal of Cheminformatics.

[41]  Ian A. Watson,et al.  Rules for identifying potentially reactive or promiscuous compounds. , 2012, Journal of medicinal chemistry.

[42]  Peter Caprioli,et al.  Evaluation of an antibody-free ADP detection assay: ADP-Glo. , 2009, Assay and drug development technologies.

[43]  R. Solé,et al.  The topology of drug-target interaction networks: implicit dependence on drug properties and target families. , 2009, Molecular bioSystems.

[44]  B. Shoichet,et al.  High-throughput assays for promiscuous inhibitors , 2005, Nature chemical biology.

[45]  Tudor I. Oprea,et al.  An Overview of the Challenges in Designing, Integrating, and Delivering BARD , 2014, Journal of biomolecular screening.

[46]  Bo-Han Su,et al.  Rule-Based Classification Models of Molecular Autofluorescence , 2015, J. Chem. Inf. Model..

[47]  George Papadatos,et al.  The ChEMBL database in 2017 , 2016, Nucleic Acids Res..

[48]  Jie Dong,et al.  TargetNet: a web service for predicting potential drug–target interaction profiling via multi-target SAR models , 2016, Journal of Computer-Aided Molecular Design.

[49]  Jürgen Bajorath,et al.  Matched molecular pair analysis of small molecule microarray data identifies promiscuity cliffs and reveals molecular origins of extreme compound promiscuity. , 2012, Journal of medicinal chemistry.

[50]  Jürgen Bajorath,et al.  Systematic computational identification of promiscuity cliff pathways formed by inhibitors of the human kinome , 2019, Journal of Computer-Aided Molecular Design.

[51]  J Willem M Nissink,et al.  Promiscuous 2-aminothiazoles (PrATs): a frequent hitting scaffold. , 2015, Journal of medicinal chemistry.

[52]  Taraneh Mirzadegan,et al.  Structural Basis of Small-Molecule Aggregate Induced Inhibition of a Protein-Protein Interaction. , 2017, Journal of medicinal chemistry.

[53]  Michael Cory,et al.  Expression, preparation, and high-throughput screening of caspase-8: discovery of redox-based and steroid diacid inhibition. , 2002, Archives of biochemistry and biophysics.

[54]  Michael J. Keiser,et al.  Predicting new molecular targets for known drugs , 2009, Nature.

[55]  Anton Simeonov,et al.  Interference with Fluorescence and Absorbance , 2015 .

[56]  Ruili Huang,et al.  Fluorescence spectroscopic profiling of compound libraries. , 2008, Journal of medicinal chemistry.

[57]  Michael J. Keiser,et al.  Relating protein pharmacology by ligand chemistry , 2007, Nature Biotechnology.

[58]  Ronald T Raines,et al.  Bright ideas for chemical biology. , 2008, ACS chemical biology.

[59]  Olivier Sperandio,et al.  FAF-Drugs2: Free ADME/tox filtering tool to assist drug discovery and chemical biology projects , 2008, BMC Bioinformatics.

[60]  Aurélien Grosdidier,et al.  SwissTargetPrediction: a web server for target prediction of bioactive small molecules , 2014, Nucleic Acids Res..

[61]  J. Jesús Naveja,et al.  HitPickV2: a web server to predict targets of chemical compounds , 2018, Bioinform..

[62]  Maria Paola Costi,et al.  Comprehensive mechanistic analysis of hits from high-throughput and docking screens against beta-lactamase. , 2008, Journal of medicinal chemistry.

[63]  J. Peters Polypharmacology - foe or friend? , 2013, Journal of medicinal chemistry.

[64]  Brian K. Shoichet,et al.  Protein stability effects in aggregate-based enzyme inhibition. , 2019, Journal of medicinal chemistry.

[65]  Igor V Tetko,et al.  Identification of Small-Molecule Frequent Hitters of Glutathione S-Transferase–Glutathione Interaction , 2016, Journal of biomolecular screening.

[66]  Jeffrey R. Huth,et al.  Enhancement of chemical rules for predicting compound reactivity towards protein thiol groups , 2007, J. Comput. Aided Mol. Des..

[67]  Michael Sattler,et al.  Luciferase Advisor: High-Accuracy Model To Flag False Positive Hits in Luciferase HTS Assays , 2018, J. Chem. Inf. Model..

[68]  Stefan Vasile,et al.  The oxidative mechanism of action of ortho-quinone inhibitors of protein-tyrosine phosphatase alpha is mediated by hydrogen peroxide. , 2004, Archives of biochemistry and biophysics.

[69]  Michal Heger,et al.  Don't discount all curcumin trial data , 2017, Nature.

[70]  J. Baell,et al.  New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. , 2010, Journal of medicinal chemistry.

[71]  Ge-Fei Hao,et al.  Computational discovery of picomolar Q(o) site inhibitors of cytochrome bc1 complex. , 2012, Journal of the American Chemical Society.

[72]  Jürgen Bajorath,et al.  Identification of Compounds That Interfere with High‐Throughput Screening Assay Technologies , 2019, Chemmedchem.

[73]  Christopher P Austin,et al.  Quantitative analyses of aggregation, autofluorescence, and reactivity artifacts in a screen for inhibitors of a thiol protease. , 2010, Journal of medicinal chemistry.

[74]  G. Rishton Nonleadlikeness and leadlikeness in biochemical screening. , 2003, Drug discovery today.

[75]  David Lagorce,et al.  FAF‐Drugs4: free ADME‐tox filtering computations for chemical biology and early stages drug discovery , 2017, Bioinform..

[76]  Ajay Goel,et al.  Multi-targeted therapy by curcumin: how spicy is it? , 2008, Molecular nutrition & food research.

[77]  Dong-Sheng Cao,et al.  Structural Analysis and Identification of Colloidal Aggregators in Drug Discovery , 2019, J. Chem. Inf. Model..

[78]  Pierre Tufféry,et al.  FAF-Drugs: free ADME/tox filtering of compound collections , 2006, Nucleic Acids Res..

[79]  Michael J. Keiser,et al.  Complementarity Between a Docking and a High-Throughput Screen in Discovering New Cruzain Inhibitors† , 2010, Journal of medicinal chemistry.

[80]  Maria F. Sassano,et al.  Automated design of ligands to polypharmacological profiles , 2012, Nature.

[81]  Anton Simeonov,et al.  Molecular basis for the high-affinity binding and stabilization of firefly luciferase by PTC124 , 2010, Proceedings of the National Academy of Sciences.

[82]  Adel Bakhtiarova,et al.  Resveratrol inhibits firefly luciferase. , 2006, Biochemical and biophysical research communications.

[83]  J. Baell,et al.  Chemistry: Chemical con artists foil drug discovery , 2014, Nature.

[84]  Yanli Wang,et al.  PubChem BioAssay: 2017 update , 2016, Nucleic Acids Res..

[85]  Jean-Louis Reymond,et al.  The polypharmacology browser: a web-based multi-fingerprint target prediction tool using ChEMBL bioactivity data , 2017, Journal of Cheminformatics.

[86]  Igor V. Tetko,et al.  Online chemical modeling environment (OCHEM): web platform for data storage, model development and publishing of chemical information , 2011, J. Comput. Aided Mol. Des..

[87]  Jean-Louis Reymond,et al.  Polypharmacology Browser PPB2: Target Prediction Combining Nearest Neighbors with Machine Learning , 2018, J. Chem. Inf. Model..

[88]  J. Irwin,et al.  An Aggregation Advisor for Ligand Discovery. , 2015, Journal of medicinal chemistry.

[89]  Daniel Reker,et al.  Computational advances in combating colloidal aggregation in drug discovery , 2019, Nature Chemistry.

[90]  James Inglese,et al.  Interferences with Luciferase Reporter Enzymes , 2016 .

[91]  Hanbing Rao,et al.  Identification of small molecule aggregators from large compound libraries by support vector machines , 2009, J. Comput. Chem..

[92]  Jing-Fang Yang,et al.  PADFrag: A Database Built for the Exploration of Bioactive Fragment Space for Drug Discovery , 2018, J. Chem. Inf. Model..

[93]  Feng-Xu Wu,et al.  Crystal Structure of 4-Hydroxyphenylpyruvate Dioxygenase in Complex with Substrate Reveals a New Starting Point for Herbicide Discovery , 2019, Research.

[94]  Brian K Shoichet,et al.  Colloidal aggregation: from screening nuisance to formulation nuance. , 2018, Nano today.

[95]  Peter Wipf,et al.  Redox Regulation of Cdc25B by Cell-Active Quinolinediones , 2005, Molecular Pharmacology.