Computational Chemistry on a Budget - Supporting Drug Discovery with Limited Resources.

An increasing number of new drugs have their origin in small biotech or academia. In contrast to big pharma, these environments are often more limited in terms of resources and this necessitates different approaches to the drug discovery process. In this perspective, we outline how computational methods can help advance drug discovery in a setting with more limited resources and we share what, based on our experience, are the best practices for these methods.

[1]  C L Verlinde,et al.  Structure-based drug design: progress, results and challenges. , 1994, Structure.

[2]  B. Shoichet,et al.  Information decay in molecular docking screens against holo, apo, and modeled conformations of enzymes. , 2003, Journal of medicinal chemistry.

[3]  C. Lipinski Lead- and drug-like compounds: the rule-of-five revolution. , 2004, Drug discovery today. Technologies.

[4]  A. Hopkins,et al.  Ligand efficiency: a useful metric for lead selection. , 2004, Drug discovery today.

[5]  Brian K. Shoichet,et al.  Virtual screening of chemical libraries , 2004, Nature.

[6]  Jens Sadowski,et al.  Structure Modification in Chemical Databases , 2005 .

[7]  E. Perola Minimizing false positives in kinase virtual screens , 2006, Proteins.

[8]  K. Wanner,et al.  Methods and Principles in Medicinal Chemistry , 2007 .

[9]  Anders Karlén,et al.  Is it possible to increase hit rates in structure-based virtual screening by pharmacophore filtering? An investigation of the advantages and pitfalls of post-filtering. , 2008, Journal of molecular graphics & modelling.

[10]  Daniel James,et al.  Lessons Learnt from Assembling Screening Libraries for Drug Discovery for Neglected Diseases , 2007, ChemMedChem.

[11]  Thomas J. Crisman,et al.  Which aspects of HTS are empirically correlated with downstream success? , 2008, Current opinion in drug discovery & development.

[12]  A. Pierce,et al.  Docking study yields four novel inhibitors of the protooncogene Pim-1 kinase. , 2008, Journal of medicinal chemistry.

[13]  Gerard J Kleywegt,et al.  Limitations and lessons in the use of X-ray structural information in drug design , 2008, Drug Discovery Today.

[14]  Max Kuhn,et al.  Building Predictive Models in R Using the caret Package , 2008 .

[15]  Sean Ekins,et al.  Novel web-based tools combining chemistry informatics, biology and social networks for drug discovery. , 2009, Drug discovery today.

[16]  Mark Whittaker,et al.  The multiple roles of computational chemistry in fragment-based drug design , 2009, J. Comput. Aided Mol. Des..

[17]  Gerhard Klebe,et al.  Molecular Docking Screens Using Comparative Models of Proteins , 2009, J. Chem. Inf. Model..

[18]  Simona Distinto,et al.  How To Optimize Shape-Based Virtual Screening: Choosing the Right Query and Including Chemical Information , 2009, J. Chem. Inf. Model..

[19]  Alexander Tropsha,et al.  Best Practices for QSAR Model Development, Validation, and Exploitation , 2010, Molecular informatics.

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

[21]  Santosh A. Khedkar,et al.  Successful applications of computer aided drug discovery: moving drugs from concept to the clinic. , 2010, Current topics in medicinal chemistry.

[22]  Richard A. Lewis,et al.  Three-dimensional pharmacophore methods in drug discovery. , 2010, Journal of medicinal chemistry.

[23]  P. Wyatt,et al.  Drug discovery in academia: the third way? , 2010, Expert opinion on drug discovery.

[24]  David Rogers,et al.  Extended-Connectivity Fingerprints , 2010, J. Chem. Inf. Model..

[25]  J. Bajorath,et al.  Quo vadis, virtual screening? A comprehensive survey of prospective applications. , 2010, Journal of medicinal chemistry.

[26]  D. Bojanic,et al.  Impact of high-throughput screening in biomedical research , 2011, Nature Reviews Drug Discovery.

[27]  M. Mezei,et al.  Molecular docking: a powerful approach for structure-based drug discovery. , 2011, Current computer-aided drug design.

[28]  Stephen Frye,et al.  US academic drug discovery , 2011, Nature Reviews Drug Discovery.

[29]  Evan Bolton,et al.  PubChem's BioAssay Database , 2011, Nucleic Acids Res..

[30]  Andreas Bender,et al.  Recognizing Pitfalls in Virtual Screening: A Critical Review , 2012, J. Chem. Inf. Model..

[31]  John P. Overington,et al.  ChEMBL: a large-scale bioactivity database for drug discovery , 2011, Nucleic Acids Res..

[32]  Frank Neese,et al.  The ORCA program system , 2012 .

[33]  Fredrik Svensson,et al.  Virtual Screening Data Fusion Using Both Structure- and Ligand-Based Methods , 2012, J. Chem. Inf. Model..

[34]  Tian Zhu,et al.  Hit identification and optimization in virtual screening: practical recommendations based on a critical literature analysis. , 2013, Journal of medicinal chemistry.

[35]  Paul Czodrowski,et al.  hERG Me Out , 2013, J. Chem. Inf. Model..

[36]  Woody Sherman,et al.  Protein and ligand preparation: parameters, protocols, and influence on virtual screening enrichments , 2013, Journal of Computer-Aided Molecular Design.

[37]  S. Muresan,et al.  Chemical predictive modelling to improve compound quality , 2013, Nature Reviews Drug Discovery.

[38]  Caroline M. R. Low,et al.  UK academic drug discovery , 2013, Nature Reviews Drug Discovery.

[39]  A. Cavalli,et al.  Predicting the reactivity of nitrile-carrying compounds with cysteine: a combined computational and experimental study. , 2014, ACS medicinal chemistry letters.

[40]  Nathan Brown,et al.  Best of Both Worlds: On the Complementarity of Ligand-Based and Structure-Based Virtual Screening , 2014, J. Chem. Inf. Model..

[41]  R. Ketteler,et al.  Academic drug discovery within the United Kingdom: a reassessment , 2015, Nature Reviews Drug Discovery.

[42]  Jessica Holien,et al.  Improvements, trends, and new ideas in molecular docking: 2012–2013 in review , 2015, Journal of molecular recognition : JMR.

[43]  Hanoch Senderowitz,et al.  A reliable computational workflow for the selection of optimal screening libraries , 2015, Journal of Cheminformatics.

[44]  Stefano Forli,et al.  Charting a Path to Success in Virtual Screening , 2015, Molecules.

[45]  Gang Fu,et al.  PubChem Substance and Compound databases , 2015, Nucleic Acids Res..

[46]  Cathy H. Wu,et al.  UniProt: the universal protein knowledgebase , 2016, Nucleic Acids Research.

[47]  E. Benfenati In Silico Methods for Predicting Drug Toxicity , 2016, Methods in Molecular Biology.

[48]  W. Guba,et al.  A Real-World Perspective on Molecular Design. , 2016, Journal of medicinal chemistry.

[49]  Anabella Villalobos,et al.  Central Nervous System Multiparameter Optimization Desirability: Application in Drug Discovery. , 2016, ACS chemical neuroscience.

[50]  Emilio Benfenati,et al.  In Silico Methods for Carcinogenicity Assessment. , 2016, Methods in molecular biology.

[51]  A. Bender,et al.  Analysis of Iterative Screening with Stepwise Compound Selection Based on Novartis In-house HTS Data. , 2016, ACS chemical biology.

[52]  George Papadatos,et al.  SureChEMBL: a large-scale, chemically annotated patent document database , 2015, Nucleic Acids Res..

[53]  Richard D. Smith,et al.  CSAR 2014: A Benchmark Exercise Using Unpublished Data from Pharma , 2016, J. Chem. Inf. Model..

[54]  Nicholas A Meanwell,et al.  Improving Drug Design: An Update on Recent Applications of Efficiency Metrics, Strategies for Replacing Problematic Elements, and Compounds in Nontraditional Drug Space. , 2016, Chemical research in toxicology.

[55]  Theodora Katsila,et al.  Computational approaches in target identification and drug discovery , 2016, Computational and structural biotechnology journal.

[56]  Shuai Liu,et al.  D3R Grand Challenge 2: blind prediction of protein–ligand poses, affinity rankings, and relative binding free energies , 2017, Journal of Computer-Aided Molecular Design.

[57]  Jianyou Shi,et al.  Assessing the performance of docking scoring function, FEP, MM-GBSA, and QM/MM-GBSA approaches on a series of PLK1 inhibitors. , 2017, MedChemComm.

[58]  Christian Tyrchan,et al.  Matched Molecular Pair Analysis in Short: Algorithms, Applications and Limitations , 2016, Computational and structural biotechnology journal.

[59]  Huikun Zhang,et al.  Machine Learning Consensus Scoring Improves Performance Across Targets in Structure-Based Virtual Screening , 2017, J. Chem. Inf. Model..

[60]  Daniel Svozil,et al.  Probes &Drugs portal: an interactive, open data resource for chemical biology , 2017, Nature Methods.

[61]  Carl Petersson,et al.  In Silico Absorption, Distribution, Metabolism, Excretion, and Pharmacokinetics (ADME-PK): Utility and Best Practices. An Industry Perspective from the International Consortium for Innovation through Quality in Pharmaceutical Development. , 2017, Journal of medicinal chemistry.

[62]  Gautier Koscielny,et al.  Open Targets: a platform for therapeutic target identification and validation , 2016, Nucleic Acids Res..

[63]  Paul C D Hawkins,et al.  Ligand-Based Methods in GPCR Computer-Aided Drug Design. , 2018, Methods in molecular biology.

[64]  Lewis R. Vidler,et al.  Investigating the Behavior of Published PAINS Alerts Using a Pharmaceutical Company Data Set , 2018, ACS medicinal chemistry letters.

[65]  Thomas Blaschke,et al.  The rise of deep learning in drug discovery. , 2018, Drug discovery today.

[66]  J Willem M Nissink,et al.  Seven Year Itch: Pan-Assay Interference Compounds (PAINS) in 2017—Utility and Limitations , 2017, ACS chemical biology.

[67]  Yuemin Bian,et al.  Deep Learning for Drug Design: an Artificial Intelligence Paradigm for Drug Discovery in the Big Data Era , 2018, The AAPS Journal.

[68]  L. McNamee,et al.  Contribution of NIH funding to new drug approvals 2010–2016 , 2018, Proceedings of the National Academy of Sciences.

[69]  Gerard J. P. van Westen,et al.  Data-driven approaches used for compound library design, hit triage and bioactivity modeling in high-throughput screening , 2016, Briefings Bioinform..

[70]  Charlie Hodgman,et al.  Gsmodutils: a python based framework for test-driven genome scale metabolic model development , 2018, bioRxiv.

[71]  Fresh from the biotech pipeline—2018 , 2019, Nature Biotechnology.

[72]  Emanuel J. V. Gonçalves,et al.  Prioritization of cancer therapeutic targets using CRISPR–Cas9 screens , 2019, Nature.

[73]  Ren Jun,et al.  In silico approaches and tools for the prediction of drug metabolism and fate: A review , 2019, Comput. Biol. Medicine.

[74]  Fredrik Svensson,et al.  LightGBM: An Effective and Scalable Algorithm for Prediction of Chemical Toxicity-Application to the Tox21 and Mutagenicity Data Sets , 2019, J. Chem. Inf. Model..

[75]  Yan Li,et al.  Comparative Assessment of Scoring Functions: The CASF-2016 Update , 2018, J. Chem. Inf. Model..

[76]  John J. M. Wiener,et al.  Accelerating Lead Identification by High Throughput Virtual Screening: Prospective Case Studies from the Pharmaceutical Industry , 2019, J. Chem. Inf. Model..

[77]  Paul N. Mortenson,et al.  Structure-Activity and Structure-Conformation Relationships of Aryl Propionic Acid Inhibitors of the Kelch-like ECH-Associated Protein 1/Nuclear Factor Erythroid 2-Related Factor 2 (KEAP1/NRF2) Protein-Protein Interaction. , 2019, Journal of medicinal chemistry.

[78]  Bin Li,et al.  Applications of machine learning in drug discovery and development , 2019, Nature Reviews Drug Discovery.

[79]  F. Svensson,et al.  Scaffold-hopping identifies furano[2,3-d]pyrimidine amides as potent Notum inhibitors , 2019, Bioorganic & medicinal chemistry letters.