Comprehensive Survey of Consensus Docking for High-Throughput Virtual Screening
Abstract
:1. Introduction
2. Structural Data Determination
3. Computational Approaches Based on Structural Data: Protein Docking
3.1. Search Algorithms
3.2. Scoring Functions
4. Computational Approaches Based on Structural Data: Virtual Screening (VS)
5. Consensus Models of Docking
5.1. Consensus Methods
5.2. Datasets
5.3. Metric Validation
6. Computational Power
7. The Vanilloid Receptor TRPV1: A Case Study
8. Outlook
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Sample Availability
References
- Dhasmana, A.R.; Jahan, S.R.; Lohani, M.; Arif, J.M. Chapter 19—High-Throughput Virtual Screening (HTVS) of Natural Compounds and Exploration of Their Biomolecular Mechanisms: An In Silico Approach. In New Look to Phytomedicine; Ahmad, M.S., Khan, I.A., Chattopadhyay, D., Eds.; Academic Press: Cambridge, MA, USA, 2019; pp. 523–548. [Google Scholar]
- Arrowsmith, J.; Miller, P. Trial watch: Phase II and phase III attrition rates 2011-2012. Nat. Rev. Drug. Discov. 2013, 12, 569. [Google Scholar] [CrossRef] [PubMed]
- Smith, C. Drug target validation: Hitting the target. Nature 2003, 422, 341, 343, 345 passim. [Google Scholar] [CrossRef] [Green Version]
- Kontoyianni, M. Docking and Virtual Screening in Drug Discovery. Methods Mol. Biol. 2017, 1647, 255–266. [Google Scholar] [PubMed]
- Parker, C.N.; Bajorath, J. Towards Unified Compound Screening Strategies: A Critical Evaluation of Error Sources in Experimental and Virtual High-Throughput Screening. Qsar. Comb. Sci. 2006, 25, 1153–1161. [Google Scholar] [CrossRef]
- Tomar, V.; Mazumder, M.; Chandra, R.; Yang, J.; Sakharkar, M.K. Small molecule drug design. In Encyclopedia of Bioinformatics and Computational Biology: ABC of Bioinformatics; Ranganathan, S., Nakai, M.G.K., Schönbach, C.B., Eds.; Academic Press: Oxford, UK, 2018; Volume 1–3, pp. 741–760. [Google Scholar]
- Abdolmaleki, A.; Ghasemi, J.B.; Ghasemi, F. Computer Aided Drug Design for Multi-Target Drug Design: SAR /QSAR, Molecular Docking and Pharmacophore Methods. Curr. Drug. Targets. 2017, 18, 556–575. [Google Scholar] [CrossRef] [PubMed]
- Srinivasarao, M.; Low, P.S. Ligand-Targeted Drug Delivery. Chem. Rev. 2017, 117, 12133–12164. [Google Scholar] [CrossRef]
- Acharya, C.; Coop, A.; Polli, J.E.; Mackerell, A.D., Jr. Recent advances in ligand-based drug design: Relevance and utility of the conformationally sampled pharmacophore approach. Curr. Comput. Aided. Drug. Des. 2011, 7, 10–22. [Google Scholar] [CrossRef] [Green Version]
- Lionta, E.; Spyrou, G.; Vassilatis, D.K.; Cournia, Z. Structure-based virtual screening for drug discovery: Principles, applications and recent advances. Curr. Top. Med. Chem. 2014, 14, 1923–1938. [Google Scholar] [CrossRef]
- Aminpour, M.; Montemagno, C.; Tuszynski, J.A. An Overview of Molecular Modeling for Drug Discovery with Specific Illustrative Examples of Applications. Molecules 2019, 24, 1693. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Wang, K. Exploiting the Diversity of Ion Channels: Modulation of Ion Channels for Therapeutic Indications. Handb. Exp. Pharm. 2019, 260, 187–205. [Google Scholar]
- Fernandez-Ballester, G.; Fernandez-Carvajal, A.; Ferrer-Montiel, A. Targeting thermoTRP ion channels: In silico preclinical approaches and opportunities. Expert. Opin. Targets 2020, 24, 1079–1097. [Google Scholar] [CrossRef] [PubMed]
- Benjin, X.; Ling, L. Developments, applications, and prospects of cryo-electron microscopy. Protein. Sci. 2020, 29, 872–882. [Google Scholar] [CrossRef] [PubMed]
- Denisov, I.G.; Sligar, S.G. Nanodiscs in Membrane Biochemistry and Biophysics. Chem. Rev. 2017, 117, 4669–4713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Callaway, E. The revolution will not be crystallized: A new method sweeps through structural biology. Nature 2015, 525, 172–174. [Google Scholar] [CrossRef] [Green Version]
- Fernandez-Ballester, G.; Serrano, L. Prediction of protein-protein interaction based on structure. Methods Mol. Biol. 2006, 340, 207–234. [Google Scholar]
- Evers, A.; Gohlke, H.; Klebe, G. Ligand-supported homology modelling of protein binding-sites using knowledge-based potentials. J. Mol. Biol. 2003, 334, 327–345. [Google Scholar] [CrossRef]
- Enkavi, G.; Javanainen, M.; Kulig, W.; Rog, T.; Vattulainen, I. Multiscale Simulations of Biological Membranes: The Challenge To Understand Biological Phenomena in a Living Substance. Chem. Rev. 2019, 119, 5607–5774. [Google Scholar] [CrossRef] [Green Version]
- Corradi, V.; Sejdiu, B.I.; Mesa-Galloso, H.; Abdizadeh, H.; Noskov, S.Y.; Marrink, S.J.; Tieleman, D.P. Emerging Diversity in Lipid-Protein Interactions. Chem. Rev. 2019, 119, 5775–5848. [Google Scholar] [CrossRef] [Green Version]
- Humphrey, W.; Dalke, A.; Schulten, K. VMD: Visual molecular dynamics. J. Mol. Graph. 1996, 14, 33–38. [Google Scholar] [CrossRef]
- Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Zidek, A.; Potapenko, A.; et al. Highly accurate protein structure prediction with AlphaFold. Nature 2021, 596, 583–589. [Google Scholar] [CrossRef]
- Skolnick, J.; Gao, M.; Zhou, H.; Singh, S. AlphaFold 2: Why It Works and Its Implications for Understanding the Relationships of Protein Sequence, Structure, and Function. J. Chem. Inf. Model 2021, 61, 4827–4831. [Google Scholar] [CrossRef] [PubMed]
- Baek, M.; DiMaio, F.; Anishchenko, I.; Dauparas, J.; Ovchinnikov, S.; Lee, G.R.; Wang, J.; Cong, Q.; Kinch, L.N.; Schaeffer, R.D.; et al. Accurate prediction of protein structures and interactions using a three-track neural network. Science 2021, 373, 871–876. [Google Scholar] [CrossRef] [PubMed]
- Carlsson, J.; Coleman, R.G.; Setola, V.; Irwin, J.J.; Fan, H.; Schlessinger, A.; Sali, A.; Roth, B.L.; Shoichet, B.K. Ligand discovery from a dopamine D3 receptor homology model and crystal structure. Nat. Chem. Biol. 2011, 7, 769–778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lim, V.J.Y.; Du, W.; Chen, Y.Z.; Fan, H. A benchmarking study on virtual ligand screening against homology models of human GPCRs. Proteins 2018, 86, 978–989. [Google Scholar] [CrossRef]
- Wong, F.; Krishnan, A.; Zheng, E.J.; Stark, H.; Manson, A.L.; Earl, A.M.; Jaakkola, T.; Collins, J.J. Benchmarking AlphaFold-enabled molecular docking predictions for antibiotic discovery. Mol. Syst. Biol. 2022, 18, e11081. [Google Scholar] [CrossRef]
- Weng, Y.; Pan, C.; Shen, Z.; Chen, S.; Xu, L.; Dong, X.; Chen, J. Identification of Potential WSB1 Inhibitors by AlphaFold Modeling, Virtual Screening, and Molecular Dynamics Simulation Studies. Evid. Based. Complement. Altern. Med. 2022, 2022, 4629392. [Google Scholar] [CrossRef]
- Lee, A.C.; Harris, J.L.; Khanna, K.K.; Hong, J.H. A Comprehensive Review on Current Advances in Peptide Drug Development and Design. Int. J. Mol. Sci. 2019, 20, 2383. [Google Scholar] [CrossRef] [Green Version]
- Sousa, S.F.; Fernandes, P.A.; Ramos, M.J. Protein-ligand docking: Current status and future challenges. Proteins 2006, 65, 15–26. [Google Scholar] [CrossRef]
- Pinzi, L.; Rastelli, G. Molecular Docking: Shifting Paradigms in Drug Discovery. Int. J. Mol. Sci. 2019, 20, 4331. [Google Scholar] [CrossRef] [Green Version]
- Tovchigrechko, A.; Vakser, I.A. GRAMM-X public web server for protein-protein docking. Nucleic. Acids. Res. 2006, 34, W310–W314. [Google Scholar] [CrossRef]
- Wang, J.; Dokholyan, N.V. MedusaDock 2.0: Efficient and Accurate Protein-Ligand Docking with Constraints. J. Chem. Inf. Model. 2019, 59, 2509–2515. [Google Scholar] [CrossRef] [PubMed]
- Yadava, U. Search algorithms and scoring methods in protein-ligand docking. Endocrinol. Int. J. 2018, 6, 359–367. [Google Scholar] [CrossRef]
- Kearsley, S.K.; Underwood, D.J.; Sheridan, R.P.; Miller, M.D. Flexibases: A way to enhance the use of molecular docking methods. J. Comput. Aided. Mol. Des. 1994, 8, 565–582. [Google Scholar] [CrossRef] [PubMed]
- Huang, S.Y.; Zou, X. Advances and challenges in protein-ligand docking. Int. J. Mol. Sci. 2010, 11, 3016–3034. [Google Scholar] [CrossRef] [Green Version]
- Hart, T.N.; Read, R.J. A multiple-start Monte Carlo docking method. Proteins 1992, 13, 206–222. [Google Scholar] [CrossRef]
- Morris, G.M.; Goodsell, D.S.; Halliday, R.S.; Huey, R.; Hart, W.E.; Belew, R.K.; Olson, A.J. Automated docking using a Lamarckian genetic algorithm and an empirical binding free energy function. J. Comput. Chem. 1998, 19, 1639–1662. [Google Scholar] [CrossRef]
- Arun Prasad, P.; Gautham, N. A new peptide docking strategy using a mean field technique with mutually orthogonal Latin square sampling. J. Comput. Aided. Mol. Des. 2008, 22, 815–829. [Google Scholar] [CrossRef]
- Yang, C.; Chen, E.A.; Zhang, Y. Protein-Ligand Docking in the Machine-Learning Era. Molecules 2022, 27, 4568. [Google Scholar] [CrossRef]
- Liu, J.; Wang, R. Classification of current scoring functions. J. Chem. Inf. Model. 2015, 55, 475–482. [Google Scholar] [CrossRef]
- Goodsell, D.S.; Morris, G.M.; Olson, A.J. Automated docking of flexible ligands: Applications of AutoDock. J. Mol. Recognit. 1996, 9, 1–5. [Google Scholar] [CrossRef]
- Pu, C.; Yan, G.; Shi, J.; Li, R. Assessing the performance of docking scoring function, FEP, MM-GBSA, and QM/MM-GBSA approaches on a series of PLK1 inhibitors. Medchemcomm 2017, 8, 1452–1458. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Su, M.; Liu, Z.; Li, J.; Liu, J.; Han, L.; Wang, R. Assessing protein-ligand interaction scoring functions with the CASF-2013 benchmark. Nat. Protoc. 2018, 13, 666–680. [Google Scholar] [CrossRef] [PubMed]
- Huang, S.Y.; Zou, X. An iterative knowledge-based scoring function to predict protein-ligand interactions: II. Validation of the scoring function. J. Comput. Chem. 2006, 27, 1876–1882. [Google Scholar] [CrossRef] [PubMed]
- Cortes, C.; Vapnik, V. Support-vector networks. Mach. Learn. 1995, 20, 273–297. [Google Scholar] [CrossRef]
- Schmidhuber, J. Deep learning in neural networks: An overview. Neural. Netw. 2015, 61, 85–117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liaw, A.; Wiener, M. Classification and regression by randomForest. R. News. 2002, 2, 18–22. [Google Scholar]
- Vieira, T.; Magalhaes, R.; Sousa, S. Tailoring specialized scoring functions for more efficient virtual screening. Frontiers 2019, 2, 1–4. [Google Scholar]
- Shen, C.; Weng, G.; Zhang, X.; Leung, E.L.; Yao, X.; Pang, J.; Chai, X.; Li, D.; Wang, E.; Cao, D.; et al. Accuracy or novelty: What can we gain from target-specific machine-learning-based scoring functions in virtual screening? Brief. Bioinform. 2021, 22, bbaa410. [Google Scholar] [CrossRef]
- Wallach, I.; Heifets, A. Most Ligand-Based Classification Benchmarks Reward Memorization Rather than Generalization. J. Chem. Inf. Model 2018, 58, 916–932. [Google Scholar] [CrossRef] [Green Version]
- Maia, E.H.B.; Assis, L.C.; de Oliveira, T.A.; da Silva, A.M.; Taranto, A.G. Structure-Based Virtual Screening: From Classical to Artificial Intelligence. Front. Chem. 2020, 8, 343. [Google Scholar] [CrossRef]
- Sledz, P.; Caflisch, A. Protein structure-based drug design: From docking to molecular dynamics. Curr. Opin. Struct. Biol. 2018, 48, 93–102. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Lu, G.; Sze, K.H.; Su, X.; Chan, W.Y.; Leung, K.S. Machine-learning scoring functions trained on complexes dissimilar to the test set already outperform classical counterparts on a blind benchmark. Brief. Bioinform. 2021, 22, bbab225. [Google Scholar] [CrossRef] [PubMed]
- Ohue, M.; Aoyama, K.; Akiyama, Y. High-Performance Cloud Computing for Exhaustive Protein–Protein Docking. In Advances in Parallel & Distributed Processing, and Applications; Springer: Berlin/Heidelberg, Germany, 2021; pp. 737–746. [Google Scholar]
- Fernandez-Ballester, G.; Fernandez-Carvajal, A.; Gonzalez-Ros, J.M.; Ferrer-Montiel, A. Ionic channels as targets for drug design: A review on computational methods. Pharmaceutics 2011, 3, 932–953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oakes, V.; Domene, C. Combining Structural Data with Computational Methodologies to Investigate Structure-Function Relationships in TRP Channels. Methods Mol. Biol. 2019, 1987, 65–82. [Google Scholar]
- Cavasotto, C.N.; Abagyan, R.A. Protein flexibility in ligand docking and virtual screening to protein kinases. J. Mol. Biol. 2004, 337, 209–225. [Google Scholar] [CrossRef]
- Cavasotto, C.N.; Orry, A.J.; Abagyan, R.A. The challenge of considering receptor flexibility in ligand docking and virtual screening. Curr. Comput.-Aided. Drug. Des. 2005, 1, 423–440. [Google Scholar] [CrossRef]
- Tian, S.; Sun, H.; Pan, P.; Li, D.; Zhen, X.; Li, Y.; Hou, T. Assessing an ensemble docking-based virtual screening strategy for kinase targets by considering protein flexibility. J. Chem. Inf. Model 2014, 54, 2664–2679. [Google Scholar] [CrossRef]
- Korb, O.; Olsson, T.S.; Bowden, S.J.; Hall, R.J.; Verdonk, M.L.; Liebeschuetz, J.W.; Cole, J.C. Potential and limitations of ensemble docking. J. Chem. Inf. Model 2012, 52, 1262–1274. [Google Scholar] [CrossRef]
- Amaro, R.E.; Baudry, J.; Chodera, J.; Demir, O.; McCammon, J.A.; Miao, Y.; Smith, J.C. Ensemble Docking in Drug Discovery. Biophys. J. 2018, 114, 2271–2278. [Google Scholar] [CrossRef] [Green Version]
- Du, X.; Li, Y.; Xia, Y.L.; Ai, S.M.; Liang, J.; Sang, P.; Ji, X.L.; Liu, S.Q. Insights into Protein-Ligand Interactions: Mechanisms, Models, and Methods. Int. J. Mol. Sci. 2016, 17, 144. [Google Scholar] [CrossRef] [Green Version]
- Wong, C.F. Flexible receptor docking for drug discovery. Expert. Opin. Drug. Discov. 2015, 10, 1189–1200. [Google Scholar] [CrossRef] [PubMed]
- Mohammadi, S.; Narimani, Z.; Ashouri, M.; Firouzi, R.; Karimi-Jafari, M.H. Ensemble learning from ensemble docking: Revisiting the optimum ensemble size problem. Sci. Rep. 2022, 12, 410. [Google Scholar] [CrossRef] [PubMed]
- Bender, B.J.; Gahbauer, S.; Luttens, A.; Lyu, J.; Webb, C.M.; Stein, R.M.; Fink, E.A.; Balius, T.E.; Carlsson, J.; Irwin, J.J.; et al. A practical guide to large-scale docking. Nat. Protoc. 2021, 16, 4799–4832. [Google Scholar] [CrossRef] [PubMed]
- Stafford, K.A.; Anderson, B.M.; Sorenson, J.; van den Bedem, H. AtomNet PoseRanker: Enriching Ligand Pose Quality for Dynamic Proteins in Virtual High-Throughput Screens. J. Chem. Inf. Model. 2022, 62, 1178–1189. [Google Scholar] [CrossRef] [PubMed]
- Ghersi, D.; Sanchez, R. Beyond structural genomics: Computational approaches for the identification of ligand binding sites in protein structures. J. Struct. Funct. Genom. 2011, 12, 109–117. [Google Scholar] [CrossRef]
- Kufareva, I.; Ilatovskiy, A.V.; Abagyan, R. Pocketome: An encyclopedia of small-molecule binding sites in 4D. Nucleic Acids. Res. 2012, 40, D535–D540. [Google Scholar] [CrossRef] [Green Version]
- Zhao, J.; Cao, Y.; Zhang, L. Exploring the computational methods for protein-ligand binding site prediction. Comput. Struct. Biotechnol. J. 2020, 18, 417–426. [Google Scholar] [CrossRef]
- Zhang, C.; Freddolino, P.L.; Zhang, Y. COFACTOR: Improved protein function prediction by combining structure, sequence and protein-protein interaction information. Nucleic Acids Res. 2017, 45, W291–W299. [Google Scholar] [CrossRef] [Green Version]
- Schmidtke, P.; Le Guilloux, V.; Maupetit, J.; Tuffery, P. fpocket: Online tools for protein ensemble pocket detection and tracking. Nucleic Acids. Res. 2010, 38, W582–W589. [Google Scholar] [CrossRef] [Green Version]
- Schmidtke, P.; Bidon-Chanal, A.; Luque, F.J.; Barril, X. MDpocket: Open-source cavity detection and characterization on molecular dynamics trajectories. Bioinformatics 2011, 27, 3276–3285. [Google Scholar] [CrossRef] [Green Version]
- Halgren, T.A. Identifying and characterizing binding sites and assessing druggability. J. Chem. Inf. Model 2009, 49, 377–389. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Grimm, M.; Dai, W.T.; Hou, M.C.; Xiao, Z.X.; Cao, Y. CB-Dock: A web server for cavity detection-guided protein-ligand blind docking. Acta Pharm. Sin. 2020, 41, 138–144. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.; Bell, E.W.; Yin, M.; Zhang, Y. EDock: Blind protein-ligand docking by replica-exchange monte carlo simulation. J. Cheminform. 2020, 12, 37. [Google Scholar] [CrossRef] [PubMed]
- Vajda, S.; Beglov, D.; Wakefield, A.E.; Egbert, M.; Whitty, A. Cryptic binding sites on proteins: Definition, detection, and druggability. Curr. Opin. Chem. Biol. 2018, 44, 1–8. [Google Scholar] [CrossRef]
- Lu, S.; Ji, M.; Ni, D.; Zhang, J. Discovery of hidden allosteric sites as novel targets for allosteric drug design. Drug. Discov. Today 2018, 23, 359–365. [Google Scholar] [CrossRef]
- Cimermancic, P.; Weinkam, P.; Rettenmaier, T.J.; Bichmann, L.; Keedy, D.A.; Woldeyes, R.A.; Schneidman-Duhovny, D.; Demerdash, O.N.; Mitchell, J.C.; Wells, J.A.; et al. CryptoSite: Expanding the Druggable Proteome by Characterization and Prediction of Cryptic Binding Sites. J. Mol. Biol. 2016, 428, 709–719. [Google Scholar] [CrossRef] [Green Version]
- Zhavoronkov, A.; Ivanenkov, Y.A.; Aliper, A.; Veselov, M.S.; Aladinskiy, V.A.; Aladinskaya, A.V.; Terentiev, V.A.; Polykovskiy, D.A.; Kuznetsov, M.D.; Asadulaev, A.; et al. Deep learning enables rapid identification of potent DDR1 kinase inhibitors. Nat. Biotechnol. 2019, 37, 1038–1040. [Google Scholar] [CrossRef]
- Sorokina, M.; Merseburger, P.; Rajan, K.; Yirik, M.A.; Steinbeck, C. COCONUT online: Collection of Open Natural Products database. J. Cheminform. 2021, 13, 2. [Google Scholar] [CrossRef]
- Sterling, T.; Irwin, J.J. ZINC 15—Ligand Discovery for Everyone. J. Chem. Inf. Model. 2015, 55, 2324–2337. [Google Scholar] [CrossRef]
- Gaulton, A.; Hersey, A.; Nowotka, M.; Bento, A.P.; Chambers, J.; Mendez, D.; Mutowo, P.; Atkinson, F.; Bellis, L.J.; Cibrian-Uhalte, E.; et al. The ChEMBL database in 2017. Nucleic Acids Res. 2017, 45, D945–D954. [Google Scholar] [CrossRef]
- Wishart, D.S.; Feunang, Y.D.; Guo, A.C.; Lo, E.J.; Marcu, A.; Grant, J.R.; Sajed, T.; Johnson, D.; Li, C.; Sayeeda, Z.; et al. DrugBank 5.0: A major update to the DrugBank database for 2018. Nucleic Acids Res. 2018, 46, D1074–D1082. [Google Scholar] [CrossRef] [PubMed]
- Wishart, D.S.; Guo, A.; Oler, E.; Wang, F.; Anjum, A.; Peters, H.; Dizon, R.; Sayeeda, Z.; Tian, S.; Lee, B.L.; et al. HMDB 5.0: The Human Metabolome Database for 2022. Nucleic Acids Res. 2022, 50, D622–D631. [Google Scholar] [CrossRef] [PubMed]
- Frye, L.; Bhat, S.; Akinsanya, K.; Abel, R. From computer-aided drug discovery to computer-driven drug discovery. Drug. Discov. Today Technol. 2021, 39, 111–117. [Google Scholar] [CrossRef] [PubMed]
- Pagadala, N.S.; Syed, K.; Tuszynski, J. Software for molecular docking: A review. Biophys. Rev. 2017, 9, 91–102. [Google Scholar] [CrossRef] [PubMed]
- Miranda, W.E.; Ngo, V.A.; Perissinotti, L.L.; Noskov, S.Y. Computational membrane biophysics: From ion channel interactions with drugs to cellular function. Biochim. Biophys. Acta Proteins Proteom. 2017, 1865, 1643–1653. [Google Scholar] [CrossRef] [PubMed]
- Nikolaeva Koleva, M.; Fernandez-Ballester, G. In Silico Approaches for TRP Channel Modulation. Methods Mol. Biol. 2019, 1987, 187–206. [Google Scholar] [PubMed]
- Wang, G.; Zhu, W. Molecular docking for drug discovery and development: A widely used approach but far from perfect. Future Med. Chem. 2016, 8, 1707–1710. [Google Scholar] [CrossRef] [Green Version]
- Morris, G.M.; Huey, R.; Lindstrom, W.; Sanner, M.F.; Belew, R.K.; Goodsell, D.S.; Olson, A.J. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J. Comput. Chem. 2009, 30, 2785–2791. [Google Scholar] [CrossRef] [Green Version]
- Allen, W.J.; Balius, T.E.; Mukherjee, S.; Brozell, S.R.; Moustakas, D.T.; Lang, P.T.; Case, D.A.; Kuntz, I.D.; Rizzo, R.C. DOCK 6: Impact of new features and current docking performance. J. Comput. Chem. 2015, 36, 1132–1156. [Google Scholar] [CrossRef] [Green Version]
- Trott, O.; Olson, A.J. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 2010, 31, 455–461. [Google Scholar] [CrossRef] [Green Version]
- Korb, O.; Stützle, T.; Exner, T.E. PLANTS: Application of ant colony optimization to structure-based drug design. In International Workshop on Ant Colony Optimization and Swarm Intelligence, 2006; Springer: Berlin/Heidelberg, Germany, 2006; pp. 247–258. [Google Scholar]
- Korb, O.; Stutzle, T.; Exner, T.E. Empirical scoring functions for advanced protein-ligand docking with PLANTS. J. Chem. Inf. Model. 2009, 49, 84–96. [Google Scholar] [CrossRef] [PubMed]
- Ruiz-Carmona, S.; Alvarez-Garcia, D.; Foloppe, N.; Garmendia-Doval, A.B.; Juhos, S.; Schmidtke, P.; Barril, X.; Hubbard, R.E.; Morley, S.D. rDock: A fast, versatile and open source program for docking ligands to proteins and nucleic acids. PLoS. Comput. Biol. 2014, 10, e1003571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vistoli, G.; Pedretti, A.; Mazzolari, A.; Testa, B. In silico prediction of human carboxylesterase-1 (hCES1) metabolism combining docking analyses and MD simulations. Bioorg. Med. Chem. 2010, 18, 320–329. [Google Scholar] [CrossRef] [PubMed]
- Vistoli, G.; Mazzolari, A.; Testa, B.; Pedretti, A. Binding Space Concept: A New Approach to Enhance the Reliability of Docking Scores and Its Application to Predicting Butyrylcholinesterase Hydrolytic Activity. J. Chem. Inf. Model 2017, 57, 1691–1702. [Google Scholar] [CrossRef] [PubMed]
- Jurrus, E.; Engel, D.; Star, K.; Monson, K.; Brandi, J.; Felberg, L.E.; Brookes, D.H.; Wilson, L.; Chen, J.; Liles, K.; et al. Improvements to the APBS biomolecular solvation software suite. Protein Sci. 2018, 27, 112–128. [Google Scholar] [CrossRef]
- Wang, R.; Lai, L.; Wang, S. Further development and validation of empirical scoring functions for structure-based binding affinity prediction. J. Comput. Aided. Mol. Des. 2002, 16, 11–26. [Google Scholar] [CrossRef]
- Obiol-Pardo, C.; Rubio-Martinez, J. Comparative evaluation of MMPBSA and XSCORE to compute binding free energy in XIAP-peptide complexes. J. Chem. Inf. Model 2007, 47, 134–142. [Google Scholar] [CrossRef]
- Neudert, G.; Klebe, G. DSX: A knowledge-based scoring function for the assessment of protein-ligand complexes. J. Chem. Inf. Model 2011, 51, 2731–2745. [Google Scholar] [CrossRef]
- Tran-Nguyen, V.K.; Bret, G.; Rognan, D. True Accuracy of Fast Scoring Functions to Predict High-Throughput Screening Data from Docking Poses: The Simpler the Better. J. Chem. Inf. Model 2021, 61, 2788–2797. [Google Scholar] [CrossRef]
- Singh, N.; Chaput, L.; Villoutreix, B.O. Virtual screening web servers: Designing chemical probes and drug candidates in the cyberspace. Brief. Bioinform. 2021, 22, 1790–1818. [Google Scholar] [CrossRef] [Green Version]
- Glaser, J.; Vermaas, J.V.; Rogers, D.M.; Larkin, J.; LeGrand, S.; Boehm, S.; Baker, M.B.; Scheinberg, A.; Tillack, A.F.; Thavappiragasam, M. High-throughput virtual laboratory for drug discovery using massive datasets. Int. J. High. Perform. Comput. Appl. 2021, 35, 452–468. [Google Scholar] [CrossRef]
- Talarico, C.; Gervasoni, S.; Manelfi, C.; Pedretti, A.; Vistoli, G.; Beccari, A.R. Combining Molecular Dynamics and Docking Simulations to Develop Targeted Protocols for Performing Optimized Virtual Screening Campaigns on the hTRPM8 Channel. Int. J. Mol. Sci. 2020, 21, 2265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mobley, D.L.; Klimovich, P.V. Perspective: Alchemical free energy calculations for drug discovery. J. Chem. Phys. 2012, 137, 230901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Steinbrecher, T.; Labahn, A. Towards accurate free energy calculations in ligand protein-binding studies. Curr. Med. Chem. 2010, 17, 767–785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marcou, G.; Rognan, D. Optimizing fragment and scaffold docking by use of molecular interaction fingerprints. J. Chem. Inf. Model. 2007, 47, 195–207. [Google Scholar] [CrossRef]
- Mazzolari, A.; Gervasoni, S.; Pedretti, A.; Fumagalli, L.; Matucci, R.; Vistoli, G. Repositioning Dequalinium as Potent Muscarinic Allosteric Ligand by Combining Virtual Screening Campaigns and Experimental Binding Assays. Int. J. Mol. Sci. 2020, 21, 5961. [Google Scholar] [CrossRef]
- Pedretti, A.; Mazzolari, A.; Gervasoni, S.; Vistoli, G. Rescoring and Linearly Combining: A Highly Effective Consensus Strategy for Virtual Screening Campaigns. Int. J. Mol. Sci. 2019, 20, 2060. [Google Scholar] [CrossRef] [Green Version]
- Palacio-Rodriguez, K.; Lans, I.; Cavasotto, C.N.; Cossio, P. Exponential consensus ranking improves the outcome in docking and receptor ensemble docking. Sci. Rep. 2019, 9, 5142. [Google Scholar] [CrossRef] [Green Version]
- Chilingaryan, G.; Abelyan, N.; Sargsyan, A.; Nazaryan, K.; Serobian, A.; Zakaryan, H. Combination of consensus and ensemble docking strategies for the discovery of human dihydroorotate dehydrogenase inhibitors. Sci. Rep. 2021, 11, 11417. [Google Scholar] [CrossRef]
- Wang, Z.; Sun, H.; Yao, X.; Li, D.; Xu, L.; Li, Y.; Tian, S.; Hou, T. Comprehensive evaluation of ten docking programs on a diverse set of protein-ligand complexes: The prediction accuracy of sampling power and scoring power. Phys. Chem. Chem. Phys. 2016, 18, 12964–12975. [Google Scholar] [CrossRef]
- Xu, W.; Lucke, A.J.; Fairlie, D.P. Comparing sixteen scoring functions for predicting biological activities of ligands for protein targets. J. Mol. Graph. Model. 2015, 57, 76–88. [Google Scholar] [CrossRef] [Green Version]
- Huang, S.Y. Exploring the potential of global protein-protein docking: An overview and critical assessment of current programs for automatic ab initio docking. Drug Discov. Today 2015, 20, 969–977. [Google Scholar] [CrossRef] [PubMed]
- Wang, R.; Wang, S. How does consensus scoring work for virtual library screening? An idealized computer experiment. J. Chem. Inf. Comput. Sci. 2001, 41, 1422–1426. [Google Scholar] [CrossRef] [PubMed]
- Clark, R.D.; Strizhev, A.; Leonard, J.M.; Blake, J.F.; Matthew, J.B. Consensus scoring for ligand/protein interactions. J. Mol. Graph. Model. 2002, 20, 281–295. [Google Scholar] [CrossRef] [PubMed]
- Ericksen, S.S.; Wu, H.; Zhang, H.; Michael, L.A.; Newton, M.A.; Hoffmann, F.M.; Wildman, S.A. Machine Learning Consensus Scoring Improves Performance Across Targets in Structure-Based Virtual Screening. J. Chem. Inf. Model 2017, 57, 1579–1590. [Google Scholar] [CrossRef] [PubMed]
- Llanos, M.A.; Enrique, N.; Sbaraglini, M.L.; Garofalo, F.M.; Talevi, A.; Gavernet, L.; Martin, P. Structure-Based Virtual Screening Identifies Novobiocin, Montelukast, and Cinnarizine as TRPV1 Modulators with Anticonvulsant Activity In Vivo. J. Chem. Inf. Model 2022, 62, 3008–3022. [Google Scholar] [CrossRef] [PubMed]
- McGann, M.; Nicholls, A.; Enyedy, I. The statistics of virtual screening and lead optimization. J. Comput. Aided Mol. Des. 2015, 29, 923–926. [Google Scholar] [CrossRef]
- Manelfi, C.; Gossen, J.; Gervasoni, S.; Talarico, C.; Albani, S.; Philipp, B.J.; Musiani, F.; Vistoli, G.; Rossetti, G.; Beccari, A.R.; et al. Combining Different Docking Engines and Consensus Strategies to Design and Validate Optimized Virtual Screening Protocols for the SARS-CoV-2 3CL Protease. Molecules 2021, 26, 797. [Google Scholar] [CrossRef]
- Feher, M. Consensus scoring for protein-ligand interactions. Drug Discov. Today 2006, 11, 421–428. [Google Scholar] [CrossRef]
- Houston, D.R.; Walkinshaw, M.D. Consensus docking: Improving the reliability of docking in a virtual screening context. J. Chem. Inf. Model 2013, 53, 384–390. [Google Scholar] [CrossRef]
- Kukol, A. Consensus virtual screening approaches to predict protein ligands. Eur. J. Med. Chem. 2011, 46, 4661–4664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ren, X.; Shi, Y.S.; Zhang, Y.; Liu, B.; Zhang, L.H.; Peng, Y.B.; Zeng, R. Novel Consensus Docking Strategy to Improve Ligand Pose Prediction. J. Chem. Inf. Model 2018, 58, 1662–1668. [Google Scholar] [CrossRef] [PubMed]
- Oda, A.; Tsuchida, K.; Takakura, T.; Yamaotsu, N.; Hirono, S. Comparison of consensus scoring strategies for evaluating computational models of protein-ligand complexes. J. Chem. Inf. Model 2006, 46, 380–391. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Fu, R.; Zhou, L.H.; Chen, S.P. Application of consensus scoring and principal component analysis for virtual screening against beta-secretase (BACE-1). PLoS ONE 2012, 7, e38086. [Google Scholar]
- Reau, M.; Langenfeld, F.; Zagury, J.F.; Lagarde, N.; Montes, M. Decoys Selection in Benchmarking Datasets: Overview and Perspectives. Front. Pharm. 2018, 9, 11. [Google Scholar] [CrossRef] [PubMed]
- Good, A.C.; Oprea, T.I. Optimization of CAMD techniques 3. Virtual screening enrichment studies: A help or hindrance in tool selection? J. Comput. Aided Mol. Des. 2008, 22, 169–178. [Google Scholar] [CrossRef]
- Stumpfe, D.; Bajorath, J. Applied virtual screening: Strategies, recommendations, and caveats. In Virtual Screening: Principles, Challenges, and Practical Guidelines; Wiley Online Library: Hoboken, NJ, USA, 2011; pp. 291–318. [Google Scholar]
- Bauer, M.R.; Ibrahim, T.M.; Vogel, S.M.; Boeckler, F.M. Evaluation and optimization of virtual screening workflows with DEKOIS 2.0--a public library of challenging docking benchmark sets. J. Chem. Inf. Model 2013, 53, 1447–1462. [Google Scholar] [CrossRef]
- Mysinger, M.M.; Carchia, M.; Irwin, J.J.; Shoichet, B.K. Directory of useful decoys, enhanced (DUD-E): Better ligands and decoys for better benchmarking. J. Med. Chem. 2012, 55, 6582–6594. [Google Scholar] [CrossRef]
- Rohrer, S.G.; Baumann, K. Maximum unbiased validation (MUV) data sets for virtual screening based on PubChem bioactivity data. J. Chem. Inf. Model 2009, 49, 169–184. [Google Scholar] [CrossRef]
- Imrie, F.; Bradley, A.R.; Deane, C.M. Generating Property-Matched Decoy Molecules Using Deep Learning. Bioinformatics 2021, 37, 2134–2141. [Google Scholar] [CrossRef]
- Zhang, X.; Shen, C.; Liao, B.; Jiang, D.; Wang, J.; Wu, Z.; Du, H.; Wang, T.; Huo, W.; Xu, L.; et al. TocoDecoy: A New Approach to Design Unbiased Datasets for Training and Benchmarking Machine-Learning Scoring Functions. J. Med. Chem. 2022, 65, 7918–7932. [Google Scholar] [CrossRef] [PubMed]
- Stein, R.M.; Yang, Y.; Balius, T.E.; O'Meara, M.J.; Lyu, J.; Young, J.; Tang, K.; Shoichet, B.K.; Irwin, J.J. Property-Unmatched Decoys in Docking Benchmarks. J. Chem. Inf. Model 2021, 61, 699–714. [Google Scholar] [CrossRef] [PubMed]
- Sheridan, R.P.; Singh, S.B.; Fluder, E.M.; Kearsley, S.K. Protocols for bridging the peptide to nonpeptide gap in topological similarity searches. J. Chem. Inf. Comput. Sci. 2001, 41, 1395–1406. [Google Scholar] [CrossRef] [PubMed]
- Truchon, J.F.; Bayly, C.I. Evaluating virtual screening methods: Good and bad metrics for the “early recognition” problem. J. Chem. Inf. Model 2007, 47, 488–508. [Google Scholar] [CrossRef]
- Di Stefano, M.; Galati, S.; Ortore, G.; Caligiuri, I.; Rizzolio, F.; Ceni, C.; Bertini, S.; Bononi, G.; Granchi, C.; Macchia, M.; et al. Machine Learning-Based Virtual Screening for the Identification of Cdk5 Inhibitors. Int. J. Mol. Sci. 2022, 23, 10653. [Google Scholar] [CrossRef]
- Gimeno, A.; Mestres-Truyol, J.; Ojeda-Montes, M.J.; Macip, G.; Saldivar-Espinoza, B.; Cereto-Massague, A.; Pujadas, G.; Garcia-Vallve, S. Prediction of Novel Inhibitors of the Main Protease (M-pro) of SARS-CoV-2 through Consensus Docking and Drug Reposition. Int. J. Mol. Sci. 2020, 21, 3793. [Google Scholar] [CrossRef]
- Ochoa, R.; Palacio-Rodriguez, K.; Clemente, C.M.; Adler, N.S. dockECR: Open consensus docking and ranking protocol for virtual screening of small molecules. J. Mol. Graph. Model 2021, 109, 108023. [Google Scholar] [CrossRef]
- Preto, J.; Gentile, F. Assessing and improving the performance of consensus docking strategies using the DockBox package. J. Comput. Aided Mol. Des. 2019, 33, 817–829. [Google Scholar] [CrossRef]
- Tuccinardi, T.; Poli, G.; Romboli, V.; Giordano, A.; Martinelli, A. Extensive consensus docking evaluation for ligand pose prediction and virtual screening studies. J. Chem. Inf. Model 2014, 54, 2980–2986. [Google Scholar] [CrossRef]
- Liu, B.; Qiu, W.; Jiang, L.; Gong, Z. Software pipelining for graphic processing unit acceleration: Partition, scheduling and granularity. Int. J. High. Perform. Comput. Appl. 2016, 30, 169–185. [Google Scholar] [CrossRef]
- Korb, O.; Finn, P.W.; Jones, G. The cloud and other new computational methods to improve molecular modelling. Expert. Opin. Drug Discov. 2014, 9, 1121–1131. [Google Scholar] [CrossRef] [PubMed]
- Ebejer, J.P.; Fulle, S.; Morris, G.M.; Finn, P.W. The emerging role of cloud computing in molecular modelling. J. Mol. Graph. Model 2013, 44, 177–187. [Google Scholar] [CrossRef] [PubMed]
- Santos-Martins, D.; Solis-Vasquez, L.; Tillack, A.F.; Sanner, M.F.; Koch, A.; Forli, S. Accelerating AutoDock4 with GPUs and gradient-based local search. J. Chem. Theory Comput. 2021, 17, 1060–1073. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Li, H.; Yu, K.; Jin, Z. Molecular docking-based computational platform for high-throughput virtual screening. CCF Trans. High Perform. Comput. 2022, 4, 63–74. [Google Scholar] [CrossRef]
- Liao, M.; Cao, E.; Julius, D.; Cheng, Y. Structure of the TRPV1 ion channel determined by electron cryo-microscopy. Nature 2013, 504, 107–112. [Google Scholar] [CrossRef] [Green Version]
- Gao, Y.; Cao, E.; Julius, D.; Cheng, Y. TRPV1 structures in nanodiscs reveal mechanisms of ligand and lipid action. Nature 2016, 534, 347–351. [Google Scholar] [CrossRef]
- Tominaga, M.; Tominaga, T. Structure and function of TRPV1. Pflug. Arch. 2005, 451, 143–150. [Google Scholar] [CrossRef]
- Arora, V.; Campbell, J.N.; Chung, M.K. Fight fire with fire: Neurobiology of capsaicin-induced analgesia for chronic pain. Pharmacol. Ther. 2021, 220, 107743. [Google Scholar] [CrossRef]
- Fernandez-Carvajal, A.; Fernandez-Ballester, G.; Ferrer-Montiel, A. TRPV1 in chronic pruritus and pain: Soft modulation as a therapeutic strategy. Front. Mol. Neurosci. 2022, 15, 930964. [Google Scholar] [CrossRef]
- Wong, G.Y.; Gavva, N.R. Therapeutic potential of vanilloid receptor TRPV1 agonists and antagonists as analgesics: Recent advances and setbacks. Brain Res. Rev. 2009, 60, 267–277. [Google Scholar] [CrossRef]
- Trevisani, M.; Gatti, R. TRPV1 antagonists as analgesic agents. Open Pain J. 2013, 6, 108–118. [Google Scholar] [CrossRef] [Green Version]
- Jardin, I.; Lopez, J.J.; Diez, R.; Sanchez-Collado, J.; Cantonero, C.; Albarran, L.; Woodard, G.E.; Redondo, P.C.; Salido, G.M.; Smani, T.; et al. TRPs in Pain Sensation. Front. Physiol. 2017, 8, 392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fernandez-Carvajal, A.; Gonzalez-Muniz, R.; Fernandez-Ballester, G.; Ferrer-Montiel, A. Investigational drugs in early phase clinical trials targeting thermotransient receptor potential (thermoTRP) channels. Expert. Opin. Investig. Drugs. 2020, 29, 1209–1222. [Google Scholar] [CrossRef] [PubMed]
- Nadezhdin, K.D.; Neuberger, A.; Nikolaev, Y.A.; Murphy, L.A.; Gracheva, E.O.; Bagriantsev, S.N.; Sobolevsky, A.I. Extracellular cap domain is an essential component of the TRPV1 gating mechanism. Nat. Commun. 2021, 12, 2154. [Google Scholar] [CrossRef] [PubMed]
- Cao, E.; Liao, M.; Cheng, Y.; Julius, D. TRPV1 structures in distinct conformations reveal activation mechanisms. Nature 2013, 504, 113–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- van Goor, M.K.; de Jager, L.; Cheng, Y.; van der Wijst, J. High-resolution structures of transient receptor potential vanilloid channels: Unveiling a functionally diverse group of ion channels. Protein Sci. 2020, 29, 1569–1580. [Google Scholar] [CrossRef] [PubMed]
- Elokely, K.M.; Doerksen, R.J. Docking challenge: Protein sampling and molecular docking performance. J. Chem. Inf. Model 2013, 53, 1934–1945. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.-C. Beware of docking! Trends Pharmacol. Sci. 2015, 36, 78–95. [Google Scholar] [CrossRef]
- Rueda, M.; Bottegoni, G.; Abagyan, R. Recipes for the selection of experimental protein conformations for virtual screening. J. Chem. Inf. Model 2010, 50, 186–193. [Google Scholar] [CrossRef] [Green Version]
- Kovacs, J.A.; Cavasotto, C.N.; Abagyan, R. Conformational sampling of protein flexibility in generalized coordinates: Application to ligand docking. J. Comput. Theor. Nanosci. 2005, 2, 354–361. [Google Scholar] [CrossRef]
- McCammon, J.A. Target flexibility in molecular recognition. Biochim. Biophys Acta 2005, 1754, 221–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leong, M.K.; Syu, R.-G.; Ding, Y.-L.; Weng, C.-F. Prediction of N-methyl-D-aspartate receptor GluN1-ligand binding affinity by a novel SVM-pose/SVM-score combinatorial ensemble docking scheme. Sci. Rep. 2017, 7, 40053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Acharya, A.; Agarwal, R.; Baker, M.B.; Baudry, J.; Bhowmik, D.; Boehm, S.; Byler, K.G.; Chen, S.Y.; Coates, L.; Cooper, C.J.; et al. Supercomputer-Based Ensemble Docking Drug Discovery Pipeline with Application to COVID-19. J. Chem. Inf. Model 2020, 60, 5832–5852. [Google Scholar] [CrossRef] [PubMed]
- Vogel, S.M.; Bauer, M.R.; Boeckler, F.M. DEKOIS: Demanding evaluation kits for objective in silico screening—A versatile tool for benchmarking docking programs and scoring functions. J. Chem. Inf. Model 2011, 51, 2650–2665. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Han, L.; Liu, Z.; Wang, R. Comparative assessment of scoring functions on an updated benchmark: 2. Evaluation methods and general results. J. Chem. Inf. Model 2014, 54, 1717–1736. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Liu, Z.; Li, J.; Han, L.; Liu, J.; Zhao, Z.; Wang, R. Comparative assessment of scoring functions on an updated benchmark: 1. Compilation of the test set. J. Chem. Inf. Model 2014, 54, 1700–1716. [Google Scholar] [CrossRef] [PubMed]
- Zev, S.; Raz, K.; Schwartz, R.; Tarabeh, R.; Gupta, P.K.; Major, D.T. Benchmarking the Ability of Common Docking Programs to Correctly Reproduce and Score Binding Modes in SARS-CoV-2 Protease Mpro. J. Chem. Inf. Model 2021, 61, 2957–2966. [Google Scholar] [CrossRef]
- Weng, G.; Gao, J.; Wang, Z.; Wang, E.; Hu, X.; Yao, X.; Cao, D.; Hou, T. Comprehensive Evaluation of Fourteen Docking Programs on Protein-Peptide Complexes. J. Chem. Theory Comput. 2020, 16, 3959–3969. [Google Scholar] [CrossRef] [PubMed]
- Gioia, D.; Bertazzo, M.; Recanatini, M.; Masetti, M.; Cavalli, A. Dynamic Docking: A Paradigm Shift in Computational Drug Discovery. Molecules 2017, 22, 2029. [Google Scholar] [CrossRef] [Green Version]
- Spitaleri, A.; Decherchi, S.; Cavalli, A.; Rocchia, W. Fast Dynamic Docking Guided by Adaptive Electrostatic Bias: The MD-Binding Approach. J. Chem. Theory. Comput. 2018, 14, 1727–1736. [Google Scholar] [CrossRef]
- Arute, F.; Arya, K.; Babbush, R.; Bacon, D.; Bardin, J.C.; Barends, R.; Biswas, R.; Boixo, S.; Brandao, F.; Buell, D.A.; et al. Quantum supremacy using a programmable superconducting processor. Nature 2019, 574, 505–510. [Google Scholar] [CrossRef] [PubMed]
Metric | Advantages | Disadvantages |
---|---|---|
ROC | 1. Simple graphical representation and exact measure of the accuracy of a test. 2. Performs equally well on both classes in balanced datasets. 3. The AUC is used as a simple numeric rating of diagnostic test accuracy. | 1. Actual decision thresholds are usually not displayed. 2. As the sample size decreases, the plot becomes irregular. 3. Not considered a good indicator for early enrichment of true active samples. |
PR | 1. Points out the efficiency of the model. 2. Shows how much the data are biased towards one class. 3. Helps understand whether the model is performing well in imbalanced datasets. | 1. It does not deal with all the cells of the confusion matrix. True negatives are never considered. 2. Focuses only on positive class. 3. Only suited for binary classification. |
Stage | Resource | Description |
---|---|---|
Target structure | RCSB Protein Data Bank (PDB) | PDB is the data center for the global Protein Data Bank (PDB) of 3D structure data for large biological molecules. https://www.rcsb.org; accessed 2 September 2022. |
Ligand structures | ChEMBL | ChEMBL is a database of bioactive molecules with drug-like properties. https://www.ebi.ac.uk/chembl/; accessed 2 September 2022. |
Decoys from DUD-E | DUD-E is designed to help benchmark molecular docking programs by providing challenging decoys. http://dude.docking.org; accessed 2 September 2022. | |
Target preparation | YASARA 22.5.22 | YASARA is a molecular modeling and simulation program for structure validation and prediction tools. It is used to rebuild missing side chains and loops. http://www.yasara.org; accessed 1 September 2022. |
Ligand preparation | Openbabel 2.4.1 | Openbabel. Addition of MMFF94 partial charges, salts removing, protonation at pH 7.4, conversion 2D-3D. https://openbabel.org/docs/dev/Command-line_tools/babel.html; accessed 3 October 2022. |
RDKit 2020.09.1.0 | RDKit (Chem package from RDKit). http://www.rdkit.org; accessed 3 October 2022. | |
Marvin 6.0 | Marvin (molconvert). https://chemaxon.com/marvin; accessed 3 October 2022. | |
Ligand optimization | RDKit | RDKit (package AllChem). http://www.rdkit.org; accessed 3 October 2022. |
YASARA | YASARA (NOVA force field and energy minimization steps). | |
ADMET descriptors | Marvin 6 | Marvin. ChemAxon’s calculator (cxcalc) is a command line program that performs chemical calculations using calculator plugins. https://chemaxon.com/marvin; accessed 3 October 2022. |
XLOGP3 | XLOGP3 is an optimized atom-additive method for the fast calculation of logP. http://www.sioc-ccbg.ac.cn/skins/ccbgwebsite/software/xlogp3/; accessed 6 September 2022. | |
RDKit | RDKit is used to obtain molecular descriptors. http://www.rdkit.org; accessed 3 October 2022. | |
FILTER-IT | FILTER-IT obtains some molecular descriptors and filters out molecules with unwanted properties. https://github.com/silicos-it/filter-it; accessed 6 September 2022. | |
UCSF Chimera 1.15 | UCSF Chimera is used for calculations of some molecular descriptors such as SASA and SESA (surf tool). https://www.cgl.ucsf.edu/chimera/; accessed 6 September 2022. | |
AMSOL 7.1 | AMSOL is used for calculating the free energies of solvation of molecules and ions in solution and partial atomic charges. https://comp.chem.umn.edu/amsol/; accessed 6 September 2022. | |
Docking | UCSF DOCK6.7 | UCSF DOCK6 identifies potential binding geometries and interactions of a molecule to a target using the anchor-and-grow search algorithm. https://dock.compbio.ucsf.edu/DOCK_6/index.htm; accessed 1 September 2022. |
AutoDock4 | AutoDock4 performs the docking of the ligands to a set of grids describing the target protein and pre-calculates these grids. https://autodock.scripps.edu; accessed 1 September 2022. | |
YASARA | YASARA is used to run macro executing VINA docking algorithms. | |
PLANTS | PLANTS is based on ant colony optimization employed to find a minimum energy conformation of the ligand in the protein’s binding site. https://github.com/discoverdata/parallel-PLANTS; accessed 1 September 2022. | |
RxDock | RxDock is designed for high-throughput virtual screening campaigns and binding mode prediction studies. https://rxdock.gitlab.io; accessed 1 September 2022. | |
XScore | XScore is an empirical scoring function which computes the binding affinities of the given ligand molecules to their target protein. https://www.ics.uci.edu/~dock/manuals/xscore1.1_manual/intro.html; accessed 1 September 2022. | |
DSX | DSX is a knowledge-based scoring function that consists of distance-dependent pair potentials, novel torsion angel potentials, and newly defined solvent accessible surface-dependent potentials. | |
Hits identification (Score-based consensus strategies) | NSR | NSR: Normalized score ratio |
ECR | ECR: Exponential Consensus Ranking | |
RBR | RBR: Rank-by-rank | |
RBV | RBV: Rank-by-vote | |
RBN | RBN: Rank-by-number | |
AASS | AASS: Average of auto-scaled score | |
Z-Score | Z-Score |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Blanes-Mira, C.; Fernández-Aguado, P.; de Andrés-López, J.; Fernández-Carvajal, A.; Ferrer-Montiel, A.; Fernández-Ballester, G. Comprehensive Survey of Consensus Docking for High-Throughput Virtual Screening. Molecules 2023, 28, 175. https://doi.org/10.3390/molecules28010175
Blanes-Mira C, Fernández-Aguado P, de Andrés-López J, Fernández-Carvajal A, Ferrer-Montiel A, Fernández-Ballester G. Comprehensive Survey of Consensus Docking for High-Throughput Virtual Screening. Molecules. 2023; 28(1):175. https://doi.org/10.3390/molecules28010175
Chicago/Turabian StyleBlanes-Mira, Clara, Pilar Fernández-Aguado, Jorge de Andrés-López, Asia Fernández-Carvajal, Antonio Ferrer-Montiel, and Gregorio Fernández-Ballester. 2023. "Comprehensive Survey of Consensus Docking for High-Throughput Virtual Screening" Molecules 28, no. 1: 175. https://doi.org/10.3390/molecules28010175