1
|
Correy GJ, Rachman MM, Togo T, Gahbauer S, Doruk YU, Stevens MGV, Jaishankar P, Kelley B, Goldman B, Schmidt M, Kramer T, Radchenko DS, Moroz YS, Ashworth A, Riley P, Shoichet BK, Renslo AR, Walters WP, Fraser JS. Exploration of structure-activity relationships for the SARS-CoV-2 macrodomain from shape-based fragment linking and active learning. SCIENCE ADVANCES 2025; 11:eads7187. [PMID: 40435250 PMCID: PMC12118597 DOI: 10.1126/sciadv.ads7187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 04/22/2025] [Indexed: 06/01/2025]
Abstract
The macrodomain of severe acute respiratory syndrome coronavirus 2 nonstructural protein 3 is required for viral pathogenesis and is an emerging antiviral target. We previously performed an x-ray crystallography-based fragment screen and found submicromolar inhibitors by fragment linking. However, these compounds had poor membrane permeability and liabilities that complicated optimization. Here, we developed a shape-based virtual screening pipeline-FrankenROCS. We screened the Enamine high-throughput collection of 2.1 million compounds, selecting 39 compounds for testing, with the most potent binding with a 130 μM median inhibitory concentration (IC50). We then paired FrankenROCS with an active learning algorithm (Thompson sampling) to efficiently search the Enamine REAL database of 22 billion molecules, testing 32 compounds with the most potent binding with a 220 μM IC50. Further optimization led to analogs with IC50 values better than 10 μM. This lead series has improved membrane permeability and is poised for optimization. FrankenROCS is a scalable method for fragment linking to exploit synthesis-on-demand libraries.
Collapse
Affiliation(s)
- Galen J. Correy
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Moira M. Rachman
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Takaya Togo
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Stefan Gahbauer
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Yagmur U. Doruk
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA
| | - Maisie G. V. Stevens
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA
| | - Priyadarshini Jaishankar
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | | | | | | | | | | | - Yurii S. Moroz
- Enamine Ltd., Kyiv, Ukraine
- Chemspace LLC, Kyiv, Ukraine
- Department of Chemistry, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA
| | | | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Adam R. Renslo
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | | | - James S. Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
2
|
López Pérez K, Huddleston K, Jung V, Miranda-Quintana RA. BitBIRCH Clustering Refinement Strategies. J Chem Inf Model 2025. [PMID: 40425525 DOI: 10.1021/acs.jcim.5c00627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
Chemical libraries are becoming not only increasingly bigger, but they are doing so at an accelerated pace. Keeping up with this explosion in chemical data demands more than just hardware upgrades; we need dramatically more efficient algorithms as well. We have been working in this direction, with the introduction of the instant similarity (iSIM) framework, which uses n-ary similarity to speed up the processing of very large sets. Recently, we showed how to use this technique to cluster billions of molecules with unprecedented efficiency through the BitBIRCH algorithm. In this Application Note, we present a package fully dedicated to expanding on the BitBIRCH method, including multiple options that give the user appreciable control over the tree structure, while dramatically improving the quality of the final partitions. Remarkably, this is achieved without compromising the efficiency of the original method. We also present new postprocessing tools that help dissect the clustering information, as well as ample examples showcasing the new functionalities. BitBIRCH is publicly available at: https://github.com/mqcomplab/bitbirch.
Collapse
Affiliation(s)
- Kenneth López Pérez
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, Florida 32603, United States
| | - Kate Huddleston
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, Florida 32603, United States
| | - Vicky Jung
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, Florida 32603, United States
| | - Ramón Alain Miranda-Quintana
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, Florida 32603, United States
| |
Collapse
|
3
|
Sindt F, Bret G, Rognan D. On the Difficulty to Rescore Hits from Ultralarge Docking Screens. J Chem Inf Model 2025. [PMID: 40401777 DOI: 10.1021/acs.jcim.5c00730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Docking-based virtual screening tools customized to mine ultralarge chemical spaces are consistently reported to yield both higher hit rates and more potent ligands than that achieved by conventional docking of smaller million-sized compound libraries. This remarkable achievement is however counterbalanced by the absolute necessity to design an efficient postprocessing of the millions of potential virtual hits for selecting a few chemically diverse compounds for synthesis and biological evaluation. We here retrospectively analyzed ten successful ultralarge virtual screening hit lists that underwent in vitro binding assays, for binding affinity prediction using eight rescoring methods including simple empirical scoring functions, machine learning, molecular-mechanics and quantum-mechanics approaches. Although the best predictions usually rely on the most sophisticated methods, none of the tested rescoring methods could robustly distinguish known binders from inactive compounds, across all assays. Energy refinement of protein-ligand complexes, prior to rescoring, marginally helped molecular mechanics and quantum mechanics approaches but deteriorates predictions from empirical and machine learning scoring functions.
Collapse
Affiliation(s)
- François Sindt
- Laboratoire d'innovation thérapeutique, UMR7200 CNRS-Université de Strasbourg, F-67400 Illkirch, France
| | - Guillaume Bret
- Laboratoire d'innovation thérapeutique, UMR7200 CNRS-Université de Strasbourg, F-67400 Illkirch, France
| | - Didier Rognan
- Laboratoire d'innovation thérapeutique, UMR7200 CNRS-Université de Strasbourg, F-67400 Illkirch, France
| |
Collapse
|
4
|
Gkeka P, Svensson F, Magadán CR, de Groot MJ, Jerome SV. Computational Hit Finding: An Industry Perspective. J Med Chem 2025. [PMID: 40392533 DOI: 10.1021/acs.jmedchem.4c03087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Computational hit finding, particularly virtual screening, has been a mainstay of drug discovery campaigns for decades, providing a cost-efficient complement to wet experiments. Innovation in this space slowed considerably as these approaches converged around mature software programs and stock chemical libraries up to ∼10 million in size. Recently, however, powered by massive increases in computational power, the emergence of ultralarge make-on-demand virtual libraries, the development of large capacity neural networks, the expansion of the domain of applicability of free energy calculations, and advances in protein structure prediction, the virtual screening field is currently seeing major change. We present a guide from industry practitioners summarizing key aspects on the changing computational hit finding landscape including practical recommendations for building a performant end-to-end screening workflow, strategies to mitigate risk by avoiding common pitfalls, determining success criteria, and a brief discussion of emerging technologies likely to impact drug discovery in the near future.
Collapse
Affiliation(s)
- Paraskevi Gkeka
- Integrated Drug Discovery, Molecular Design Sciences, Sanofi, Vitry-sur-Seine 91380, France
| | - Fredrik Svensson
- Cancer Research Horizons, Jonas Webb Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | | | | | - Steven V Jerome
- Schrödinger, Inc., 1540 Broadway, 24th Floor, New York, New York 10036, United States
| |
Collapse
|
5
|
Medel-Lacruz B, Herrero A, Martín F, Herrero E, Luque FJ, Vázquez J. Synthon-Based Strategies Exploiting Molecular Similarity and Protein-Ligand Interactions for Efficient Screening of Ultra-Large Chemical Libraries. J Chem Inf Model 2025. [PMID: 40294889 DOI: 10.1021/acs.jcim.5c00222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The rapid expansion of ultralarge chemical libraries has revolutionized drug discovery, providing access to billions of compounds. However, this growth poses relevant challenges for traditional virtual screening (VS) methods. To address these limitations, synthon-based approaches have emerged as scalable alternatives, exploiting combinatorial chemistry principles to prioritize building blocks over enumerated molecules. In this work, we present exaScreen and exaDock, two novel synthon-based methodologies designed for ligand-based and structure-based VS, respectively. In the former case, synthon selection is guided by the 3D hydrophobic/philic distribution pattern in conjunction with a specific synthon alignment protocol based on a quadrupolar expansion over the atoms that participate in the linking bonds between fragments. On the other hand, accommodation to the binding site under a geometrically restrained docking of synthon-based hybrid compounds is used in the selection of the optimal synthon combinations. These strategies exhibit comparable performance to the search performed using fully enumerated libraries in identifying active compounds with significantly lower computational cost, offering computationally efficient strategies for VS in ultralarge chemical spaces.
Collapse
Affiliation(s)
- Brian Medel-Lacruz
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain and Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), 08003 Barcelona, Spain
- Pharmacelera, Parc Científic de Barcelona (PCB), 08028 Barcelona, Spain
| | - Albert Herrero
- Pharmacelera, Parc Científic de Barcelona (PCB), 08028 Barcelona, Spain
| | - Fernando Martín
- Pharmacelera, Parc Científic de Barcelona (PCB), 08028 Barcelona, Spain
| | - Enric Herrero
- Pharmacelera, Parc Científic de Barcelona (PCB), 08028 Barcelona, Spain
| | - F Javier Luque
- Departament de Nutrició, Cieǹcies de l'Alimentació i Gastronomia, Facultat de Farmàcia i Cieǹcies de l'Alimentació, Institut de Biomedicina (IBUB) and Institut de Química Teòrica i Computacional (IQTCUB), Santa Coloma de Gramenet, 08921 Barcelona, Spain
| | - Javier Vázquez
- Pharmacelera, Parc Científic de Barcelona (PCB), 08028 Barcelona, Spain
- Departament de Nutrició, Cieǹcies de l'Alimentació i Gastronomia, Facultat de Farmac̀ia i Cieǹcies de l'Alimentació, Institut de Biomedicina (IBUB), Santa Coloma de Gramenet, 08921 Barcelona, Spain
| |
Collapse
|
6
|
Pérez KL, Jung V, Chen L, Huddleston K, Miranda-Quintana RA. BitBIRCH: efficient clustering of large molecular libraries. DIGITAL DISCOVERY 2025; 4:1042-1051. [PMID: 40109497 PMCID: PMC11912344 DOI: 10.1039/d5dd00030k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
The widespread use of Machine Learning (ML) techniques in chemical applications has come with the pressing need to analyze extremely large molecular libraries. In particular, clustering remains one of the most common tools to dissect the chemical space. Unfortunately, most current approaches present unfavorable time and memory scaling, which makes them unsuitable to handle million- and billion-sized sets. Here, we propose to bypass these problems with a time- and memory-efficient clustering algorithm, BitBIRCH. This method uses a tree structure similar to the one found in the Balanced Iterative Reducing and Clustering using Hierarchies (BIRCH) algorithm to ensure O(N) time scaling. BitBIRCH leverages the instant similarity (iSIM) formalism to process binary fingerprints, allowing the use of Tanimoto similarity, and reducing memory requirements. Our tests show that BitBIRCH is already >1000 times faster than standard implementations of the Taylor-Butina clustering for libraries with 1 500 000 molecules. BitBIRCH increases efficiency without compromising the quality of the resulting clusters. We explore strategies to handle large sets, which we applied in the clustering of one billion molecules under 5 hours using a parallel/iterative BitBIRCH approximation.
Collapse
Affiliation(s)
- Kenneth López Pérez
- Department of Chemistry & Quantum Theory Project, University of Florida Gainesville Florida 32611 USA
| | - Vicky Jung
- Department of Chemistry & Quantum Theory Project, University of Florida Gainesville Florida 32611 USA
| | - Lexin Chen
- Department of Chemistry & Quantum Theory Project, University of Florida Gainesville Florida 32611 USA
| | - Kate Huddleston
- Department of Chemistry & Quantum Theory Project, University of Florida Gainesville Florida 32611 USA
| | | |
Collapse
|
7
|
Bienstock RJ. AI/ML methodologies and the future-will they be successful in designing the next generation of new chemical entities? J Cheminform 2025; 17:46. [PMID: 40189582 PMCID: PMC11974048 DOI: 10.1186/s13321-025-00995-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 03/22/2025] [Indexed: 04/09/2025] Open
Abstract
Cheminformatics and chemical databases are essential to drug discovery. However, machine learning (ML) and artificial intelligence (AI) methodologies are changing the way in which chemical data is used. How will the use of chemical data change in drug discovery moving forward? How do the new ML methods in molecular property prediction, hit and lead and target identification and structure prediction differ and compare with previous computational methods? Will new ML methodologies improve chemical diversity in ligand design, and offer computational enhancements. There are still many advantages to physics based methods and they offer something lacking in ML/ AI based methods. Additionally, ML training methods often give the best results when experimental assay measurements are fed back into the model. Often modeling and experimental methods are not diametrically opposed but offer the greatest advantage when used complementary.
Collapse
|
8
|
Luttens A, Cabeza de Vaca I, Sparring L, Brea J, Martínez AL, Kahlous NA, Radchenko DS, Moroz YS, Loza MI, Norinder U, Carlsson J. Rapid traversal of vast chemical space using machine learning-guided docking screens. NATURE COMPUTATIONAL SCIENCE 2025; 5:301-312. [PMID: 40082701 PMCID: PMC12021657 DOI: 10.1038/s43588-025-00777-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 02/04/2025] [Indexed: 03/16/2025]
Abstract
The accelerating growth of make-on-demand chemical libraries provides unprecedented opportunities to identify starting points for drug discovery with virtual screening. However, these multi-billion-scale libraries are challenging to screen, even for the fastest structure-based docking methods. Here we explore a strategy that combines machine learning and molecular docking to enable rapid virtual screening of databases containing billions of compounds. In our workflow, a classification algorithm is trained to identify top-scoring compounds based on molecular docking of 1 million compounds to the target protein. The conformal prediction framework is then used to make selections from the multi-billion-scale library, reducing the number of compounds to be scored by docking. The CatBoost classifier showed an optimal balance between speed and accuracy and was used to adapt the workflow for screens of ultralarge libraries. Application to a library of 3.5 billion compounds demonstrated that our protocol can reduce the computational cost of structure-based virtual screening by more than 1,000-fold. Experimental testing of predictions identified ligands of G protein-coupled receptors and demonstrated that our approach enables discovery of compounds with multi-target activity tailored for therapeutic effect.
Collapse
Affiliation(s)
- Andreas Luttens
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden.
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Institute for Medical Engineering and Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Israel Cabeza de Vaca
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden
| | - Leonard Sparring
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden
| | - José Brea
- Innopharma Drug Screening and Pharmacogenomics Platform, BioFarma research group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Antón Leandro Martínez
- Innopharma Drug Screening and Pharmacogenomics Platform, BioFarma research group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Nour Aldin Kahlous
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden
| | | | - Yurii S Moroz
- Enamine Ltd, Kyiv, Ukraine
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
- Chemspace LLC, Kyiv, Ukraine
| | - María Isabel Loza
- Innopharma Drug Screening and Pharmacogenomics Platform, BioFarma research group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain.
- Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Ulf Norinder
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| | - Jens Carlsson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden.
| |
Collapse
|
9
|
Pérez KL, Huddleston K, Jung V, Miranda-Quintana RA. BitBIRCH Clustering Refinement Strategies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644337. [PMID: 40196520 PMCID: PMC11974763 DOI: 10.1101/2025.03.20.644337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Chemical libraries are becoming not only increasingly bigger, but they are doing so at an accelerated pace. Keeping up with this explosion in chemical data demands more than just hardware upgrades, we need dramatically more efficient algorithms as well. We have been working in this direction, with the introduction of the iSIM framework, which uses n-ary similarity to speed up the processing of very large sets. Recently, we showed how to use this technique to cluster billions of molecules with unprecedented efficiency through the BitBIRCH algorithm. In this Application Note we present a package fully-dedicated to expanding on the BitBIRCH method, including multiple options that give the user appreciable control over the tree structure, while dramatically improving the quality of the final partitions. Remarkably, this is achieved without compromising the efficiency of the original method. We also present new post-processing tools that help dissect the clustering information, as well as ample examples showcasing the new functionalities. BitBIRCH is publicly available at: https://github.com/mqcomplab/bitbirch.
Collapse
Affiliation(s)
- Kenneth López Pérez
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, FL 32603, USA
| | - Kate Huddleston
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, FL 32603, USA
| | - Vicky Jung
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, FL 32603, USA
| | | |
Collapse
|
10
|
Kramer C, Chodera J, Damm-Ganamet KL, Gilson MK, Günther J, Lessel U, Lewis RA, Mobley D, Nittinger E, Pecina A, Schapira M, Walters WP. The Need for Continuing Blinded Pose- and Activity Prediction Benchmarks. J Chem Inf Model 2025; 65:2180-2190. [PMID: 39951479 DOI: 10.1021/acs.jcim.4c02296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2025]
Abstract
Computational tools for structure-based drug design (SBDD) are widely used in drug discovery and can provide valuable insights to advance projects in an efficient and cost-effective manner. However, despite the importance of SBDD to the field, the underlying methodologies and techniques have many limitations. In particular, binding pose and activity predictions (P-AP) are still not consistently reliable. We strongly believe that a limiting factor is the lack of a widely accepted and established community benchmarking process that independently assesses the performance and drives the development of methods, similar to the CASP benchmarking challenge for protein structure prediction. Here, we provide an overview of P-AP, unblinded benchmarking data sets, and blinded benchmarking initiatives (concluded and ongoing) and offer a perspective on learnings and the future of the field. To accelerate a breakthrough on the development of novel P-AP methods, it is necessary for the community to establish and support a long-term benchmark challenge that provides nonbiased training/test/validation sets, a systematic independent validation, and a forum for scientific discussions.
Collapse
Affiliation(s)
- Christian Kramer
- F. Hoffmann-La Roche Ltd. Pharma Research and Early Development, Basel 4070, Switzerland
| | - John Chodera
- Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Kelly L Damm-Ganamet
- In Silico Discovery, Therapeutics Discovery, Johnson & Johnson Innovative Medicine, San Diego, California 92121, United States
| | - Michael K Gilson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0736, United States
| | - Judith Günther
- Bayer AG, Drug Discovery Sciences, 13353 Berlin, Germany
| | - Uta Lessel
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss 88397, Germany
| | - Richard A Lewis
- Global Discovery Chemistry, Novartis Pharma AG, Basel 4002, Switzerland
| | - David Mobley
- Departments of Pharmaceutical Sciences and Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - Eva Nittinger
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, 43183 Gothenburg, Sweden
| | - Adam Pecina
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 16000, Czech Republic
| | - Matthieu Schapira
- Structural Genomics Consortium and Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - W Patrick Walters
- Computation, Relay Therapeutics, Cambridge, Massachusetts 02141, United States
| |
Collapse
|
11
|
Bui TTM, Ko H, Um S, Jeong H, Kang SW, Kim H, Song DG, Jung SH, Moon K. Discovery of Dual ROCK1/2 Inhibitors from Nocardiopsis sp. under Metal Stress. ACS Chem Biol 2025; 20:432-441. [PMID: 39893657 DOI: 10.1021/acschembio.4c00736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Rho-associated protein kinase (ROCK) inhibitors are promising therapeutic agents for reducing elevated intraocular pressure in patients with glaucoma. We explored new ROCK inhibitors derived from bioactive metabolites produced by microbes, specifically cryptic metabolites from Nocardiopsis sp. MCY7, using a liquid chromatography-mass spectrometry-based chemical analysis approach integrated with metal stress-driven isolation. This strategy led to the identification of two previously undescribed linear peptides, nocarnickelamides A and B (1 and 2), and an unreported cittilin derivative, cittilin C (3). The planar structures of 1-3 were elucidated using UV spectroscopy, high-resolution mass spectrometry, and nuclear magnetic resonance. The absolute configurations of 1 and 2 were assigned using the advanced Marfey's method. Biological assays demonstrated that nocarnickelamides (1 and 2) exhibited dual inhibitory activity against ROCK1 (IC50 29.8 and 14.9 μM, respectively) and ROCK2 (IC50 27.0 and 21.9 μM, respectively), with molecular simulations suggesting binding to the ATP-binding site. In human trabecular meshwork cells, 2 significantly inhibited the activation of ROCK-regulated cytoskeletal contraction markers such as the myosin light chain. Nocarnickelamide B (2) is a novel dual ROCK1/2 inhibitor and a potential pharmacophore for designing new therapeutic agents to reduce intraocular pressure in glaucoma.
Collapse
Affiliation(s)
- Thinh T M Bui
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyejin Ko
- Natural Product Drug Development Division, Korea Institute of Science and Technology (KIST) Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea
| | - Soohyun Um
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, South Korea
| | - Hyeongju Jeong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Suk Woo Kang
- Natural Product Drug Development Division, Korea Institute of Science and Technology (KIST) Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea
| | - Hasun Kim
- Natural Product Drug Development Division, Korea Institute of Science and Technology (KIST) Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea
| | - Dae-Geun Song
- Natural Product Drug Development Division, Korea Institute of Science and Technology (KIST) Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea
- Natural Product Applied Science, KIST School, University of Science and Technology, Gangneung 25451, Republic of Korea
| | - Sang Hoon Jung
- Natural Product Drug Development Division, Korea Institute of Science and Technology (KIST) Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea
- Natural Product Applied Science, KIST School, University of Science and Technology, Gangneung 25451, Republic of Korea
| | - Kyuho Moon
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
12
|
Sun Y, Wu J, Shen B, Yang H, Cui H, Han W, Luo R, Zhang S, Li H, Qian B, Fan L, Zhang J, Wang T, Xia X, Yan F, Gao Y. Discovery of TRPV4-Targeting Small Molecules with Anti-Influenza Effects Through Machine Learning and Experimental Validation. Int J Mol Sci 2025; 26:1381. [PMID: 39941149 PMCID: PMC11818416 DOI: 10.3390/ijms26031381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a calcium-permeable cation channel critical for maintaining intracellular Ca2+ homeostasis and is essential in regulating immune responses, metabolic processes, and signal transduction. Recent studies have shown that TRPV4 activation enhances influenza A virus infection, promoting viral replication and transmission. However, there has been limited exploration of antiviral drugs targeting the TRPV4 channel. In this study, we developed the first machine learning model specifically designed to predict TRPV4 inhibitory small molecules, providing a novel approach for rapidly identifying repurposed drugs with potential antiviral effects. Our approach integrated machine learning, virtual screening, data analysis, and experimental validation to efficiently screen and evaluate candidate molecules. For high-throughput virtual screening, we employed computational methods to screen open-source molecular databases targeting the TRPV4 receptor protein. The virtual screening results were ranked based on predicted scores from our optimized model and binding energy, allowing us to prioritize potential inhibitors. Fifteen small-molecule drugs were selected for further in vitro and in vivo antiviral testing against influenza. Notably, glecaprevir and everolimus demonstrated significant inhibitory effects on the influenza virus, markedly improving survival rates in influenza-infected mice (protection rates of 80% and 100%, respectively). We also validated the mechanisms by which these drugs interact with the TRPV4 channel. In summary, our study presents the first predictive model for identifying TRPV4 inhibitors, underscoring TRPV4 inhibition as a promising strategy for antiviral drug development against influenza. This pioneering approach lays the groundwork for future clinical research targeting the TRPV4 channel in antiviral therapies.
Collapse
Affiliation(s)
- Yan Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China;
- State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (J.W.); (B.S.); (R.L.); (S.Z.); (H.L.); (B.Q.); (L.F.); (J.Z.); (T.W.); (X.X.)
| | - Jiajing Wu
- State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (J.W.); (B.S.); (R.L.); (S.Z.); (H.L.); (B.Q.); (L.F.); (J.Z.); (T.W.); (X.X.)
| | - Beilei Shen
- State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (J.W.); (B.S.); (R.L.); (S.Z.); (H.L.); (B.Q.); (L.F.); (J.Z.); (T.W.); (X.X.)
| | - Hengzheng Yang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China; (H.Y.); (H.C.); (W.H.)
| | - Huizi Cui
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China; (H.Y.); (H.C.); (W.H.)
| | - Weiwei Han
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China; (H.Y.); (H.C.); (W.H.)
| | - Rongbo Luo
- State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (J.W.); (B.S.); (R.L.); (S.Z.); (H.L.); (B.Q.); (L.F.); (J.Z.); (T.W.); (X.X.)
| | - Shijun Zhang
- State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (J.W.); (B.S.); (R.L.); (S.Z.); (H.L.); (B.Q.); (L.F.); (J.Z.); (T.W.); (X.X.)
| | - He Li
- State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (J.W.); (B.S.); (R.L.); (S.Z.); (H.L.); (B.Q.); (L.F.); (J.Z.); (T.W.); (X.X.)
| | - Bingshuo Qian
- State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (J.W.); (B.S.); (R.L.); (S.Z.); (H.L.); (B.Q.); (L.F.); (J.Z.); (T.W.); (X.X.)
| | - Lingjun Fan
- State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (J.W.); (B.S.); (R.L.); (S.Z.); (H.L.); (B.Q.); (L.F.); (J.Z.); (T.W.); (X.X.)
| | - Junkui Zhang
- State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (J.W.); (B.S.); (R.L.); (S.Z.); (H.L.); (B.Q.); (L.F.); (J.Z.); (T.W.); (X.X.)
| | - Tiecheng Wang
- State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (J.W.); (B.S.); (R.L.); (S.Z.); (H.L.); (B.Q.); (L.F.); (J.Z.); (T.W.); (X.X.)
| | - Xianzhu Xia
- State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (J.W.); (B.S.); (R.L.); (S.Z.); (H.L.); (B.Q.); (L.F.); (J.Z.); (T.W.); (X.X.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Fang Yan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China;
| | - Yuwei Gao
- State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (J.W.); (B.S.); (R.L.); (S.Z.); (H.L.); (B.Q.); (L.F.); (J.Z.); (T.W.); (X.X.)
| |
Collapse
|
13
|
Kozyrev V, Sindt F, Rognan D. Active Learning to Select the Most Suitable Reagents and One-Step Organic Chemistry Reactions for Prioritizing Target-Specific Hits from Ultralarge Chemical Spaces. J Chem Inf Model 2025; 65:693-704. [PMID: 39815802 DOI: 10.1021/acs.jcim.4c02097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Designing chemically novel and synthesizable ligands from the largest possible chemical space is a major issue in modern drug discovery to identify early hits that are easily amenable to medicinal chemistry optimization. Starting from the sole three-dimensional structure of a protein binding site, we herewith describe a fully automated active learning protocol to propose the commercial chemical reagents and one-step organic chemistry reactions necessary to enumerate target-specific primary hits from ultralarge chemical spaces. When applied in different scenarios (single transform and multiple transforms) addressing chemical spaces of various sizes (from 670 million to 4.5 billion compounds), the method was able to recover up to 98% of virtual hits discovered by an exhaustive docking-based approach while scanning only 5% of the full chemical space. It is therefore applicable to the structure-based screening of trillion-sized chemical spaces at a very high throughput with minimal computational resources.
Collapse
Affiliation(s)
- Vladimir Kozyrev
- Laboratoire d'Innovation Thérapeutique, UMR7200 CNRS-Université de Strasbourg, F-67400 Illkirch, France
| | - François Sindt
- Laboratoire d'Innovation Thérapeutique, UMR7200 CNRS-Université de Strasbourg, F-67400 Illkirch, France
| | - Didier Rognan
- Laboratoire d'Innovation Thérapeutique, UMR7200 CNRS-Université de Strasbourg, F-67400 Illkirch, France
| |
Collapse
|
14
|
Corrêa Veríssimo G, Salgado Ferreira R, Gonçalves Maltarollo V. Ultra-Large Virtual Screening: Definition, Recent Advances, and Challenges in Drug Design. Mol Inform 2025; 44:e202400305. [PMID: 39635776 DOI: 10.1002/minf.202400305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
Virtual screening (VS) in drug design employs computational methodologies to systematically rank molecules from a virtual compound library based on predicted features related to their biological activities or chemical properties. The recent expansion in commercially accessible compound libraries and the advancements in artificial intelligence (AI) and computational power - including enhanced central processing units (CPUs), graphics processing units (GPUs), high-performance computing (HPC), and cloud computing - have significantly expanded our capacity to screen libraries containing over 109 molecules. Herein, we review the concept of ultra-large virtual screening (ULVS), focusing on the various algorithms and methodologies employed for virtual screening at this scale. In this context, we present the software utilized, applications, and results of different approaches, such as brute force docking, reaction-based docking approaches, machine learning (ML) strategies applied to docking or other VS methods, and similarity/pharmacophore search-based techniques. These examples represent a paradigm shift in the drug discovery process, demonstrating not only the feasibility of billion-scale compound screening but also their potential to identify hit candidates and increase the structural diversity of novel compounds with biological activities.
Collapse
Affiliation(s)
- Gabriel Corrêa Veríssimo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
- Programa de Pós-Graduação em Bioinformática, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
| | - Rafaela Salgado Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
| | - Vinícius Gonçalves Maltarollo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
| |
Collapse
|
15
|
Moesgaard L, Kongsted J. Introducing SpaceGA: A Search Tool to Accelerate Large Virtual Screenings of Combinatorial Libraries. J Chem Inf Model 2024; 64:8123-8130. [PMID: 39475501 DOI: 10.1021/acs.jcim.4c01308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2024]
Abstract
The growth of make-on-demand libraries in recent years has provided completely new possibilities for virtual screening for discovering new hit compounds with specific and favorable properties. However, since these libraries now contain billions of compounds, screening them using traditional methods such as molecular docking has become challenging and requires substantial computational resources. Thus, to take real advantage of the new possibilities introduced by the make-on-demand libraries, different methods have been proposed to accelerate the screening process and prioritize molecules for evaluation. Here, we introduce SpaceGA, a genetic algorithm that leverages the rapid similarity search tool SpaceLight (Bellmann, L.; Penner, P.; Rarey, M. Topological similarity search in large combinatorial fragment spaces. J. Chem. Inf. Model. 2021, 61, 238-251). to constrain the optimization process to accessible compounds within desired combinatorial libraries. As shown herein, SpaceGA is able to efficiently identify molecules with desired properties from trillions of synthesizable compounds by enumerating and evaluating only a small fraction of them. On this basis, SpaceGA represents a promising new tool for accelerating and simplifying virtual screens of ultralarge combinatorial databases.
Collapse
Affiliation(s)
- Laust Moesgaard
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, Odense DK-5230, Denmark
| | - Jacob Kongsted
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, Odense DK-5230, Denmark
| |
Collapse
|
16
|
Nakata S, Mori Y, Tanaka S. Navigating Ultralarge Virtual Chemical Spaces with Product-of-Experts Chemical Language Models. J Chem Inf Model 2024; 64:7873-7884. [PMID: 39413401 DOI: 10.1021/acs.jcim.4c01214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Ultralarge virtual chemical spaces have emerged as a valuable resource for drug discovery, providing access to billions of make-on-demand compounds with high synthetic success rates. Chemical language models can potentially accelerate the exploration of these vast spaces through direct compound generation. However, existing models are not designed to navigate specific virtual chemical spaces and often overlook synthetic accessibility. To address this gap, we introduce product-of-experts (PoE) chemical language models, a modular and scalable approach to navigating ultralarge virtual chemical spaces. This method allows for controlled compound generation within a desired chemical space by combining a prior model pretrained on the target space with expert and anti-expert models fine-tuned using external property-specific data sets. We demonstrate that the PoE chemical language model can generate compounds with desirable properties, such as those that favorably dock to dopamine receptor D2 (DRD2) and are predicted to cross the blood-brain barrier (BBB), while ensuring that the majority of generated compounds are present within the target chemical space. Our results highlight the potential of chemical language models for navigating ultralarge virtual chemical spaces, and we anticipate that this study will motivate further research in this direction. The source code and data are freely available at https://github.com/shuyana/poeclm.
Collapse
Affiliation(s)
- Shuya Nakata
- Graduate School of System Informatics, Kobe University, Kobe 657-8501, Japan
| | - Yoshiharu Mori
- Graduate School of System Informatics, Kobe University, Kobe 657-8501, Japan
| | - Shigenori Tanaka
- Graduate School of System Informatics, Kobe University, Kobe 657-8501, Japan
| |
Collapse
|
17
|
Wang K, Huang Y, Wang Y, You Q, Wang L. Recent advances from computer-aided drug design to artificial intelligence drug design. RSC Med Chem 2024; 15:d4md00522h. [PMID: 39493228 PMCID: PMC11523840 DOI: 10.1039/d4md00522h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024] Open
Abstract
Computer-aided drug design (CADD), a cornerstone of modern drug discovery, can predict how a molecular structure relates to its activity and interacts with its target using structure-based and ligand-based methods. Fueled by ever-increasing data availability and continuous model optimization, artificial intelligence drug design (AIDD), as an enhanced iteration of CADD, has thrived in the past decade. AIDD demonstrates unprecedented opportunities in protein folding, property prediction, and molecular generation. It can also facilitate target identification, high-throughput screening (HTS), and synthetic route prediction. With AIDD involved, the process of drug discovery is greatly accelerated. Notably, AIDD offers the potential to explore uncharted territories of chemical space beyond current knowledge. In this perspective, we began by briefly outlining the main workflows and components of CADD. Then through showcasing exemplary cases driven by AIDD in recent years, we describe the evolving role of artificial intelligence (AI) in drug discovery from three distinct stages, that is, chemical library screening, linker generation, and de novo molecular generation. In this process, we attempted to draw comparisons between the features of CADD and AIDD.
Collapse
Affiliation(s)
- Keran Wang
- State Key Laboratory of Natural Medicines and, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University Nanjing 210009 China +86 025 83271351 +86 15261483858
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University Nanjing 210009 China
| | - Yanwen Huang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University Beijing 100191 China
| | - Yan Wang
- Department of Urology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine Shanghai 201203 China +86 13122152007
| | - Qidong You
- State Key Laboratory of Natural Medicines and, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University Nanjing 210009 China +86 025 83271351 +86 15261483858
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University Nanjing 210009 China
| | - Lei Wang
- State Key Laboratory of Natural Medicines and, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University Nanjing 210009 China +86 025 83271351 +86 15261483858
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University Nanjing 210009 China
| |
Collapse
|
18
|
Wu Y, Liu F, Glenn I, Fonseca-Valencia K, Paris L, Xiong Y, Jerome SV, Brooks CL, Shoichet BK. Identifying Artifacts from Large Library Docking. J Med Chem 2024; 67:16796-16806. [PMID: 39255340 PMCID: PMC11890070 DOI: 10.1021/acs.jmedchem.4c01632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
While large library docking has discovered potent ligands for multiple targets, as the libraries have grown the hit lists can become dominated by rare artifacts that cheat our scoring functions. Here, we investigate rescoring top-ranked docked molecules with orthogonal methods to identify these artifacts, exploring implicit solvent models and absolute binding free energy perturbation as cross-filters. In retrospective studies, this approach deprioritized high-ranking nonbinders for nine targets while leaving true ligands relatively unaffected. We tested the method prospectively against hits from docking against AmpC β-lactamase. We prioritized 128 high-ranking molecules for synthesis and testing, a mixture of 39 molecules flagged as likely cheaters and 89 that were plausible inhibitors. None of the predicted cheating compounds inhibited AmpC detectably, while 57% of the 89 plausible compounds did so. As our libraries continue to grow, deprioritizing docking artifacts by rescoring with orthogonal methods may find wide use.
Collapse
Affiliation(s)
- Yujin Wu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, United States
| | - Fangyu Liu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, United States
| | - Isabella Glenn
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, United States
| | - Karla Fonseca-Valencia
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, United States
| | - Lu Paris
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, United States
| | - Yuyue Xiong
- Schrödinger, Inc., 9868 Scranton Road, San Diego, California 92121, United States
| | - Steven V Jerome
- Schrödinger, Inc., 1540 Broadway, New York, New York 10036, United States
| | - Charles L Brooks
- Biophysics Program, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, United States
| |
Collapse
|
19
|
Wang N, Zang ZH, Sun BB, Li B, Tian JL. Recent advances in computational prediction of molecular properties in food chemistry. Food Res Int 2024; 192:114776. [PMID: 39147479 DOI: 10.1016/j.foodres.2024.114776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/10/2024] [Accepted: 07/14/2024] [Indexed: 08/17/2024]
Abstract
The combination of food chemistry and computational simulation has brought many impacts to food research, moving from experimental chemistry to computer chemistry. This paper will systematically review in detail the important role played by computational simulations in the development of the molecular structure of food, mainly from the atomic, molecular, and multicomponent dimension. It will also discuss how different computational chemistry models can be constructed and analyzed to obtain reliable conclusions. From the calculation principle to case analysis, this paper focuses on the selection and application of quantum mechanics, molecular mechanics and coarse-grained molecular dynamics in food chemistry research. Finally, experiments and computations of food chemistry are compared and summarized to obtain the best balance between them. The above review and outlook will provide an important reference for the intersection of food chemistry and computational chemistry, and is expected to provide innovative thinking for structural research in food chemistry.
Collapse
Affiliation(s)
- Nuo Wang
- College of Food Science, Shenyang Agricultural University, National R&D Professional Center for Berry Processing, National Engineering and Technology of Research Center for Small berry, Key Laborotary of Healthy Food Nutrition and Innovative Manufacturing, Liaoning Province, Shenyang, Liaoning 110866, China
| | - Zhi-Huan Zang
- College of Food Science, Shenyang Agricultural University, National R&D Professional Center for Berry Processing, National Engineering and Technology of Research Center for Small berry, Key Laborotary of Healthy Food Nutrition and Innovative Manufacturing, Liaoning Province, Shenyang, Liaoning 110866, China
| | - Bing-Bing Sun
- College of Food Science, Shenyang Agricultural University, National R&D Professional Center for Berry Processing, National Engineering and Technology of Research Center for Small berry, Key Laborotary of Healthy Food Nutrition and Innovative Manufacturing, Liaoning Province, Shenyang, Liaoning 110866, China
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, National R&D Professional Center for Berry Processing, National Engineering and Technology of Research Center for Small berry, Key Laborotary of Healthy Food Nutrition and Innovative Manufacturing, Liaoning Province, Shenyang, Liaoning 110866, China
| | - Jin-Long Tian
- College of Food Science, Shenyang Agricultural University, National R&D Professional Center for Berry Processing, National Engineering and Technology of Research Center for Small berry, Key Laborotary of Healthy Food Nutrition and Innovative Manufacturing, Liaoning Province, Shenyang, Liaoning 110866, China.
| |
Collapse
|
20
|
Pala D, Clark DE. Caught between a ROCK and a hard place: current challenges in structure-based drug design. Drug Discov Today 2024; 29:104106. [PMID: 39029868 DOI: 10.1016/j.drudis.2024.104106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/27/2024] [Accepted: 07/13/2024] [Indexed: 07/21/2024]
Abstract
The discipline of structure-based drug design (SBDD) is several decades old and it is tempting to think that the proliferation of experimental structures for many drug targets might make computer-aided drug design (CADD) straightforward. However, this is far from true. In this review, we illustrate some of the challenges that CADD scientists face every day in their work, even now. We use Rho-associated protein kinase (ROCK), and public domain structures and data, as an example to illustrate some of the challenges we have experienced during our project targeting this protein. We hope that this will help to prevent unrealistic expectations of what CADD can accomplish and to educate non-CADD scientists regarding the challenges still facing their CADD colleagues.
Collapse
Affiliation(s)
- Daniele Pala
- Medicinal Chemistry and Drug Design Technologies Department, Chiesi Farmaceutici S.p.A, Research Center, Largo Belloli 11/a, 43122 Parma, Italy
| | - David E Clark
- Charles River, 6-9 Spire Green Centre, Flex Meadow, Harlow CM19 5TR, UK.
| |
Collapse
|
21
|
Weller J, Rohs R. Structure-Based Drug Design with a Deep Hierarchical Generative Model. J Chem Inf Model 2024; 64:6450-6463. [PMID: 39058534 PMCID: PMC11350878 DOI: 10.1021/acs.jcim.4c01193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024]
Abstract
Recently, the remarkable growth of available crystal structure data and libraries of commercially available or readily synthesizable molecules have unlocked previously inaccessible regions of chemical space for drug development. Paired with improvements in virtual ligand screening methods, these expanded libraries are having a notable impact on early drug design efforts. Yet screening-based methods still face scalability limits, due to computational constraints and the sheer scale of drug-like space. Machine learning approaches are overcoming these limitations by learning the fundamental intra- and intermolecular relationships in drug-target systems from existing data. Here, we introduce DrugHIVE, a deep hierarchical variational autoencoder that outperforms state-of-the-art autoregressive and diffusion-based methods in both speed and performance on common generative benchmarks. DrugHIVE's hierarchical design enables improved control over molecular generation. Its capabilities include dramatically increasing virtual screening efficiency and accelerating a wide range of common drug design tasks, including de novo generation, molecular optimization, scaffold hopping, linker design, and high-throughput pattern replacement. Our highly scalable method can even be applied to receptors with high-confidence AlphaFold-predicted structures, extending the ability to generate high-quality drug-like molecules to a majority of the unsolved human proteome.
Collapse
Affiliation(s)
- Jesse
A. Weller
- Department
of Quantitative and Computational Biology, University of Southern California, Los Angeles, California 90089, United States
- Department
of Physics and Astronomy, University of
Southern California, Los Angeles, California 90089, United States
| | - Remo Rohs
- Department
of Quantitative and Computational Biology, University of Southern California, Los Angeles, California 90089, United States
- Department
of Physics and Astronomy, University of
Southern California, Los Angeles, California 90089, United States
- Department
of Chemistry, University of Southern California, Los Angeles, California 90089, United States
- Thomas
Lord Department of Computer Science, University
of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
22
|
Correy GJ, Rachman M, Togo T, Gahbauer S, Doruk YU, Stevens M, Jaishankar P, Kelley B, Goldman B, Schmidt M, Kramer T, Ashworth A, Riley P, Shoichet BK, Renslo AR, Walters WP, Fraser JS. Extensive exploration of structure activity relationships for the SARS-CoV-2 macrodomain from shape-based fragment merging and active learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.25.609621. [PMID: 39253507 PMCID: PMC11383323 DOI: 10.1101/2024.08.25.609621] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
The macrodomain contained in the SARS-CoV-2 non-structural protein 3 (NSP3) is required for viral pathogenesis and lethality. Inhibitors that block the macrodomain could be a new therapeutic strategy for viral suppression. We previously performed a large-scale X-ray crystallography-based fragment screen and discovered a sub-micromolar inhibitor by fragment linking. However, this carboxylic acid-containing lead had poor membrane permeability and other liabilities that made optimization difficult. Here, we developed a shape-based virtual screening pipeline - FrankenROCS - to identify new macrodomain inhibitors using fragment X-ray crystal structures. We used FrankenROCS to exhaustively screen the Enamine high-throughput screening (HTS) collection of 2.1 million compounds and selected 39 compounds for testing, with the most potent compound having an IC50 value equal to 130 μM. We then paired FrankenROCS with an active learning algorithm (Thompson sampling) to efficiently search the Enamine REAL database of 22 billion molecules, testing 32 compounds with the most potent having an IC50 equal to 220 μM. Further optimization led to analogs with IC50 values better than 10 μM, with X-ray crystal structures revealing diverse binding modes despite conserved chemical features. These analogs represent a new lead series with improved membrane permeability that is poised for optimization. In addition, the collection of 137 X-ray crystal structures with associated binding data will serve as a resource for the development of structure-based drug discovery methods. FrankenROCS may be a scalable method for fragment linking to exploit ever-growing synthesis-on-demand libraries.
Collapse
Affiliation(s)
- Galen J. Correy
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158
| | - Moira Rachman
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158
| | - Takaya Togo
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158
| | - Stefan Gahbauer
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158
| | - Yagmur U. Doruk
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158
| | - Maisie Stevens
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158
| | - Priyadarshini Jaishankar
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158
| | | | | | | | | | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158
| | | | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158
| | - Adam R. Renslo
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158
| | | | - James S. Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158
| |
Collapse
|
23
|
Pérez KL, Jung V, Chen L, Huddleston K, Miranda-Quintana RA. Efficient clustering of large molecular libraries. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.10.607459. [PMID: 39149242 PMCID: PMC11326248 DOI: 10.1101/2024.08.10.607459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The widespread use of Machine Learning (ML) techniques in chemical applications has come with the pressing need to analyze extremely large molecular libraries. In particular, clustering remains one of the most common tools to dissect the chemical space. Unfortunately, most current approaches present unfavorable time and memory scaling, which makes them unsuitable to handle million- and billion-sized sets. Here, we propose to bypass these problems with a time- and memory-efficient clustering algorithm, BitBIRCH. This method uses a tree structure similar to the one found in the Balanced Iterative Reducing and Clustering using Hierarchies (BIRCH) algorithm to ensure O N time scaling. BitBIRCH leverages the instant similarity (iSIM) formalism to process binary fingerprints, allowing the use of Tanimoto similarity, and reducing memory requirements. Our tests show that BitBIRCH is already > 1,000 times faster than standard implementations of the Taylor-Butina clustering for libraries with 1,500,000 molecules. BitBIRCH increases efficiency without compromising the quality of the resulting clusters. We explore strategies to handle large sets, which we applied in the clustering of one billion molecules under 5 hours using a parallel/iterative BitBIRCH approximation.
Collapse
Affiliation(s)
| | | | - Lexin Chen
- Department of Chemistry & Quantum Theory Project, University of Florida, Gainesville, Florida 32611
| | - Kate Huddleston
- Department of Chemistry & Quantum Theory Project, University of Florida, Gainesville, Florida 32611
| | | |
Collapse
|
24
|
Carlsson J, Luttens A. Structure-based virtual screening of vast chemical space as a starting point for drug discovery. Curr Opin Struct Biol 2024; 87:102829. [PMID: 38848655 DOI: 10.1016/j.sbi.2024.102829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/16/2024] [Accepted: 04/21/2024] [Indexed: 06/09/2024]
Abstract
Structure-based virtual screening aims to find molecules forming favorable interactions with a biological macromolecule using computational models of complexes. The recent surge of commercially available chemical space provides the opportunity to search for ligands of therapeutic targets among billions of compounds. This review offers a compact overview of structure-based virtual screens of vast chemical spaces, highlighting successful applications in early drug discovery for therapeutically important targets such as G protein-coupled receptors and viral enzymes. Emphasis is placed on strategies to explore ultra-large chemical libraries and synergies with emerging machine learning techniques. The current opportunities and future challenges of virtual screening are discussed, indicating that this approach will play an important role in the next-generation drug discovery pipeline.
Collapse
Affiliation(s)
- Jens Carlsson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24 Uppsala, Sweden.
| | - Andreas Luttens
- Institute for Medical Engineering & Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
25
|
Wu Y, Liu F, Glenn I, Fonseca-Valencia K, Paris L, Xiong Y, Jerome SV, Brooks CL, Shoichet BK. Identifying Artifacts from Large Library Docking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.603966. [PMID: 39071262 PMCID: PMC11275789 DOI: 10.1101/2024.07.17.603966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
While large library docking has discovered potent ligands for multiple targets, as the libraries have grown, the very top of the hit-lists can become populated with artifacts that cheat our scoring functions. Though these cheating molecules are rare, they become ever-more dominant with library growth. Here, we investigate rescoring top-ranked molecules from docking screens with orthogonal methods to identify these artifacts, exploring implicit solvent models and absolute binding free energy perturbation (AB-FEP) as cross-filters. In retrospective studies, this approach deprioritized high-ranking non-binders for nine targets while leaving true ligands relatively unaffected. We tested the method prospectively against results from large library docking AmpC β-lactamase. From the very top of the docking hit lists, we prioritized 128 molecules for synthesis and experimental testing, a mixture of 39 molecules that rescoring flagged as likely cheaters and another 89 that were plausible true actives. None of the 39 predicted cheating compounds inhibited AmpC up to 200 μ M in enzyme assays, while 57% of the 89 plausible true actives did do so, with 19 of them inhibiting the enzyme with apparentK i values better than 50 μ M . As our libraries continue to grow, a strategy of catching docking artifacts by rescoring with orthogonal methods may find wide use in the field.
Collapse
Affiliation(s)
- Yujin Wu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158, United States
| | - Fangyu Liu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158, United States
| | - Isabella Glenn
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158, United States
| | - Karla Fonseca-Valencia
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158, United States
| | - Lu Paris
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158, United States
| | - Yuyue Xiong
- Schrödinger, Inc., 9868 Scranton Road, San Diego, California 92121, United States
| | - Steven V. Jerome
- Schrödinger, Inc., 1540 Broadway, New York, New York, 10036, United States
| | - Charles L. Brooks
- Biophysics Program, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158, United States
| |
Collapse
|
26
|
Bedart C, Simoben CV, Schapira M. Emerging structure-based computational methods to screen the exploding accessible chemical space. Curr Opin Struct Biol 2024; 86:102812. [PMID: 38603987 DOI: 10.1016/j.sbi.2024.102812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 04/13/2024]
Abstract
Structure-based virtual screening can be a valuable approach to computationally select hit candidates based on their predicted interaction with a protein of interest. The recent explosion in the size of chemical libraries increases the chances of hitting high-quality compounds during virtual screening exercises but also poses new challenges as the number of chemically accessible molecules grows faster than the computing power necessary to screen them. We review here two novel approaches rapidly gaining in popularity to address this problem: machine learning-accelerated and synthon-based library screening. We summarize the results from seminal proof-of-concept studies, highlight the latest developments, and discuss limitations and future directions.
Collapse
Affiliation(s)
- Corentin Bedart
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | - Conrad Veranso Simoben
- Structural Genomics Consortium, University of Toronto, 101 College Street, MaRS South Tower, Suite 700, Toronto, Ontario M5G 1L7, Canada
| | - Matthieu Schapira
- Structural Genomics Consortium, University of Toronto, 101 College Street, MaRS South Tower, Suite 700, Toronto, Ontario M5G 1L7, Canada; Department of Pharmacology and Toxicology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
27
|
Song RX, Nicklaus MC, Tarasova NI. Correlation of protein binding pocket properties with hits' chemistries used in generation of ultra-large virtual libraries. J Comput Aided Mol Des 2024; 38:22. [PMID: 38753096 PMCID: PMC11098933 DOI: 10.1007/s10822-024-00562-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/22/2024] [Indexed: 05/19/2024]
Abstract
Although the size of virtual libraries of synthesizable compounds is growing rapidly, we are still enumerating only tiny fractions of the drug-like chemical universe. Our capability to mine these newly generated libraries also lags their growth. That is why fragment-based approaches that utilize on-demand virtual combinatorial libraries are gaining popularity in drug discovery. These à la carte libraries utilize synthetic blocks found to be effective binders in parts of target protein pockets and a variety of reliable chemistries to connect them. There is, however, no data on the potential impact of the chemistries used for making on-demand libraries on the hit rates during virtual screening. There are also no rules to guide in the selection of these synthetic methods for production of custom libraries. We have used the SAVI (Synthetically Accessible Virtual Inventory) library, constructed using 53 reliable reaction types (transforms), to evaluate the impact of these chemistries on docking hit rates for 40 well-characterized protein pockets. The data shows that the virtual hit rates differ significantly for different chemistries with cross coupling reactions such as Sonogashira, Suzuki-Miyaura, Hiyama and Liebeskind-Srogl coupling producing the highest hit rates. Virtual hit rates appear to depend not only on the property of the formed chemical bond but also on the diversity of available building blocks and the scope of the reaction. The data identifies reactions that deserve wider use through increasing the number of corresponding building blocks and suggests the reactions that are more effective for pockets with certain physical and hydrogen bond-forming properties.
Collapse
Affiliation(s)
- Robert X Song
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Marc C Nicklaus
- Computer-Aided Drug Design Group, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Frederick, MD, 21702, USA
| | - Nadya I Tarasova
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA.
| |
Collapse
|
28
|
Lim VJY, Gerber HD, Schihada H, Trinh VT, Hilger D, Vázquez O, Kolb P. A virtual library of small molecules mimicking dipeptides. Arch Pharm (Weinheim) 2024; 357:e2300636. [PMID: 38332463 DOI: 10.1002/ardp.202300636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/13/2023] [Accepted: 01/08/2024] [Indexed: 02/10/2024]
Abstract
Virtual combinatorial libraries are prevalent in drug discovery due to improvements in the prediction of synthetic reactions that can be performed. This has gone hand in hand with the development of virtual screening capabilities to effectively screen the large chemical spaces spanned by exhaustive enumeration of reaction products. In this study, we generated a small-molecule dipeptide mimic library to target proteins binding small peptides. The library was created based on the general idea of peptide synthesis, that is, amino acid mimics were reacted in silico to form the dipeptide mimics, yielding 2,036,819 unique compounds. After docking calculations, two compounds from the library were synthesized and tested against WD repeat-containing protein 5 (WDR5) and histamine receptors H1-H4 to evaluate whether these molecules are viable in assays. The compounds showed the highest potency at the histamine H3 receptor, with Ki values in the two-digit micromolar range.
Collapse
Affiliation(s)
- Victor Jun Yu Lim
- Pharmaceutical Chemistry, Department of Pharmacy, University of Marburg, Marburg, Germany
| | - Hans-Dieter Gerber
- Pharmaceutical Chemistry, Department of Pharmacy, University of Marburg, Marburg, Germany
| | - Hannes Schihada
- Pharmaceutical Chemistry, Department of Pharmacy, University of Marburg, Marburg, Germany
| | - Van Tuan Trinh
- Chemical Biology, Department of Chemistry, University of Marburg, Marburg, Germany
| | - Daniel Hilger
- Pharmaceutical Chemistry, Department of Pharmacy, University of Marburg, Marburg, Germany
| | - Olalla Vázquez
- Chemical Biology, Department of Chemistry, University of Marburg, Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), University of Marburg, Marburg, Germany
| | - Peter Kolb
- Pharmaceutical Chemistry, Department of Pharmacy, University of Marburg, Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), University of Marburg, Marburg, Germany
| |
Collapse
|
29
|
Marin E, Kovaleva M, Kadukova M, Mustafin K, Khorn P, Rogachev A, Mishin A, Guskov A, Borshchevskiy V. Regression-Based Active Learning for Accessible Acceleration of Ultra-Large Library Docking. J Chem Inf Model 2024; 64:2612-2623. [PMID: 38157481 PMCID: PMC11005039 DOI: 10.1021/acs.jcim.3c01661] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024]
Abstract
Structure-based drug discovery is a process for both hit finding and optimization that relies on a validated three-dimensional model of a target biomolecule, used to rationalize the structure-function relationship for this particular target. An ultralarge virtual screening approach has emerged recently for rapid discovery of high-affinity hit compounds, but it requires substantial computational resources. This study shows that active learning with simple linear regression models can accelerate virtual screening, retrieving up to 90% of the top-1% of the docking hit list after docking just 10% of the ligands. The results demonstrate that it is unnecessary to use complex models, such as deep learning approaches, to predict the imprecise results of ligand docking with a low sampling depth. Furthermore, we explore active learning meta-parameters and find that constant batch size models with a simple ensembling method provide the best ligand retrieval rate. Finally, our approach is validated on the ultralarge size virtual screening data set, retrieving 70% of the top-0.05% of ligands after screening only 2% of the library. Altogether, this work provides a computationally accessible approach for accelerated virtual screening that can serve as a blueprint for the future design of low-compute agents for exploration of the chemical space via large-scale accelerated docking. With recent breakthroughs in protein structure prediction, this method can significantly increase accessibility for the academic community and aid in the rapid discovery of high-affinity hit compounds for various targets.
Collapse
Affiliation(s)
- Egor Marin
- Research
Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Margarita Kovaleva
- Research
Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Maria Kadukova
- Research
Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- University
Grenoble Alpes, Inria, CNRS, Grenoble INP, LJK, 38000 Grenoble, France
| | - Khalid Mustafin
- Research
Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Polina Khorn
- Research
Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Andrey Rogachev
- Research
Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Joint
Institute for Nuclear Research, Dubna 141980, Russian
Federation
| | - Alexey Mishin
- Research
Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Albert Guskov
- Groningen
Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Valentin Borshchevskiy
- Research
Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Joint
Institute for Nuclear Research, Dubna 141980, Russian
Federation
| |
Collapse
|
30
|
Schuck B, Brenk R. On the hunt for metalloenzyme inhibitors: Investigating the presence of metal-coordinating compounds in screening libraries and chemical spaces. Arch Pharm (Weinheim) 2024; 357:e2300648. [PMID: 38279543 DOI: 10.1002/ardp.202300648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/28/2024]
Abstract
Metalloenzymes play vital roles in various biological processes, requiring the search for inhibitors to develop treatment options for diverse diseases. While compound library screening is a conventional approach, the exploration of virtual chemical spaces housing trillions of compounds has emerged as an alternative strategy. In this study, we investigated the suitability of selected screening libraries and chemical spaces for discovering inhibitors of metalloenzymes featuring common ions (Mg2+, Mn2+, and Zn2+). First, metal-coordinating groups from ligands interacting with ions in the Protein Data Bank were extracted. Subsequently, the prevalence of these groups in two focused screening libraries (Life Chemicals' chelator library, comprising 6,428 compounds, and Otava's chelator fragment library, with 1,784 fragments) as well as two chemical spaces (GalaXi and REAL space, containing billions of virtual products) was investigated. In total, 1,223 metal-coordinating groups were identified, with about a quarter of these groups found within the examined libraries and spaces. Our results indicate that these can serve as valuable starting points for drug discovery targeting metalloenzymes. In addition, this study suggests ways to improve libraries and spaces for better success in finding potential inhibitors for metalloenzymes.
Collapse
Affiliation(s)
- Bruna Schuck
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ruth Brenk
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Computational Biology Unit, University of Bergen, Bergen, Norway
| |
Collapse
|
31
|
Sindt F, Seyller A, Eguida M, Rognan D. Protein Structure-Based Organic Chemistry-Driven Ligand Design from Ultralarge Chemical Spaces. ACS CENTRAL SCIENCE 2024; 10:615-627. [PMID: 38559302 PMCID: PMC10979501 DOI: 10.1021/acscentsci.3c01521] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 04/04/2024]
Abstract
Ultralarge chemical spaces describing several billion compounds are revolutionizing hit identification in early drug discovery. Because of their size, such chemical spaces cannot be fully enumerated and require ad-hoc computational tools to navigate them and pick potentially interesting hits. We here propose a structure-based approach to ultralarge chemical space screening in which commercial chemical reagents are first docked to the target of interest and then directly connected according to organic chemistry and topological rules, to enumerate drug-like compounds under three-dimensional constraints of the target. When applied to bespoke chemical spaces of different sizes and chemical complexity targeting two receptors of pharmaceutical interest (estrogen β receptor, dopamine D3 receptor), the computational method was able to quickly enumerate hits that were either known ligands (or very close analogs) of targeted receptors as well as chemically novel candidates that could be experimentally confirmed by in vitro binding assays. The proposed approach is generic, can be applied to any docking algorithm, and requires few computational resources to prioritize easily synthesizable hits from billion-sized chemical spaces.
Collapse
Affiliation(s)
- François Sindt
- Laboratoire d’innovation
thérapeutique, UMR7200 CNRS-Université de Strasbourg, Illkirch 67400, France
| | - Anthony Seyller
- Laboratoire d’innovation
thérapeutique, UMR7200 CNRS-Université de Strasbourg, Illkirch 67400, France
| | | | - Didier Rognan
- Laboratoire d’innovation
thérapeutique, UMR7200 CNRS-Université de Strasbourg, Illkirch 67400, France
| |
Collapse
|
32
|
Weller JA, Rohs R. DrugHIVE: Target-specific spatial drug design and optimization with a hierarchical generative model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.22.573155. [PMID: 38187658 PMCID: PMC10769420 DOI: 10.1101/2023.12.22.573155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Rapid advancement in the computational methods of structure-based drug design has led to their widespread adoption as key tools in the early drug development process. Recently, the remarkable growth of available crystal structure data and libraries of commercially available or readily synthesizable molecules have unlocked previously inaccessible regions of chemical space for drug development. Paired with improvements in virtual ligand screening methods, these expanded libraries are having a significant impact on the success of early drug design efforts. However, screening-based methods are limited in their scalability due to computational limits and the sheer scale of drug-like space. An approach within the quickly evolving field of artificial intelligence (AI), deep generative modeling, is extending the reach of molecular design beyond classical methods by learning the fundamental intra- and inter-molecular relationships in drug-target systems from existing data. In this work we introduce DrugHIVE, a deep hierarchical structure-based generative model that enables fine-grained control over molecular generation. Our model outperforms state of the art autoregressive and diffusion-based methods on common benchmarks and in speed of generation. Here, we demonstrate DrugHIVEs capacity to accelerate a wide range of common drug design tasks such as de novo generation, molecular optimization, scaffold hopping, linker design, and high throughput pattern replacement. Our method is highly scalable and can be applied to high confidence AlphaFold predicted receptors, extending our ability to generate high quality drug-like molecules to a majority of the unsolved human proteome.
Collapse
|
33
|
Hönig SMN, Flachsenberg F, Ehrt C, Neumann A, Schmidt R, Lemmen C, Rarey M. SpaceGrow: efficient shape-based virtual screening of billion-sized combinatorial fragment spaces. J Comput Aided Mol Des 2024; 38:13. [PMID: 38493240 PMCID: PMC10944417 DOI: 10.1007/s10822-024-00551-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/13/2024] [Indexed: 03/18/2024]
Abstract
The growing size of make-on-demand chemical libraries is posing new challenges to cheminformatics. These ultra-large chemical libraries became too large for exhaustive enumeration. Using a combinatorial approach instead, the resource requirement scales approximately with the number of synthons instead of the number of molecules. This gives access to billions or trillions of compounds as so-called chemical spaces with moderate hardware and in a reasonable time frame. While extremely performant ligand-based 2D methods exist in this context, 3D methods still largely rely on exhaustive enumeration and therefore fail to apply. Here, we present SpaceGrow: a novel shape-based 3D approach for ligand-based virtual screening of billions of compounds within hours on a single CPU. Compared to a conventional superposition tool, SpaceGrow shows comparable pose reproduction capacity based on RMSD and superior ranking performance while being orders of magnitude faster. Result assessment of two differently sized subsets of the eXplore space reveals a higher probability of finding superior results in larger spaces highlighting the potential of searching in ultra-large spaces. Furthermore, the application of SpaceGrow in a drug discovery workflow was investigated in four examples involving G protein-coupled receptors (GPCRs) with the aim to identify compounds with similar binding capabilities and molecular novelty.
Collapse
Affiliation(s)
- Sophia M N Hönig
- BioSolveIT, An der Ziegelei 79, 53757, Sankt Augustin, Germany
- Universität Hamburg, ZBH - Center for Bioinformatics, Albert-Einstein-Ring 8-10, 22761, Hamburg, Germany
| | | | - Christiane Ehrt
- Universität Hamburg, ZBH - Center for Bioinformatics, Albert-Einstein-Ring 8-10, 22761, Hamburg, Germany
| | | | - Robert Schmidt
- BioSolveIT, An der Ziegelei 79, 53757, Sankt Augustin, Germany
| | | | - Matthias Rarey
- Universität Hamburg, ZBH - Center for Bioinformatics, Albert-Einstein-Ring 8-10, 22761, Hamburg, Germany.
| |
Collapse
|
34
|
Cheng C, Beroza P. Shape-Aware Synthon Search (SASS) for Virtual Screening of Synthon-Based Chemical Spaces. J Chem Inf Model 2024; 64:1251-1260. [PMID: 38335044 DOI: 10.1021/acs.jcim.3c01865] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Virtual screening of large-scale chemical libraries has become increasingly useful for identifying high-quality candidates for drug discovery. While it is possible to exhaustively screen chemical spaces that number on the order of billions, indirect combinatorial approaches are needed to efficiently navigate larger, synthon-based virtual spaces. We describe Shape-Aware Synthon Search (SASS), a synthon-based virtual screening method that carries out shape similarity searches in the synthon space instead of the enumerated product space. SASS can replicate results from exhaustive searches in ultralarge, combinatorial spaces with high recall on a variety of query molecules while only scoring a small subspace of possible enumerated products, thereby significantly accelerating large-scale, shape-based virtual screening.
Collapse
Affiliation(s)
- Chen Cheng
- Discovery Chemistry, Genentech, South San Francisco, California 94080, United States
| | - Paul Beroza
- Discovery Chemistry, Genentech, South San Francisco, California 94080, United States
| |
Collapse
|
35
|
Klarich K, Goldman B, Kramer T, Riley P, Walters WP. Thompson Sampling─An Efficient Method for Searching Ultralarge Synthesis on Demand Databases. J Chem Inf Model 2024; 64:1158-1171. [PMID: 38316125 PMCID: PMC10900287 DOI: 10.1021/acs.jcim.3c01790] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/07/2024]
Abstract
Over the last five years, virtual screening of ultralarge synthesis on-demand libraries has emerged as a powerful tool for hit identification in drug discovery programs. As these libraries have grown to tens of billions of molecules, we have reached a point where it is no longer cost-effective to screen every molecule virtually. To address these challenges, several groups have developed heuristic search methods to rapidly identify the best molecules on a virtual screen. This article describes the application of Thompson sampling (TS), an active learning approach that streamlines the virtual screening of large combinatorial libraries by performing a probabilistic search in the reagent space, thereby never requiring the full enumeration of the library. TS is a general technique that can be applied to various virtual screening modalities, including 2D and 3D similarity search, docking, and application of machine-learning models. In an illustrative example, we show that TS can identify more than half of the top 100 molecules from a docking-based virtual screen of 335 million molecules by evaluating 1% of the data set.
Collapse
Affiliation(s)
- Kathryn Klarich
- ReNAgade
Therapeutics, 640 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Brian Goldman
- Relay
Therapeutics, 399 Binney Street, Cambridge, Massachusetts 02141, United States
| | - Trevor Kramer
- Relay
Therapeutics, 399 Binney Street, Cambridge, Massachusetts 02141, United States
| | - Patrick Riley
- Relay
Therapeutics, 399 Binney Street, Cambridge, Massachusetts 02141, United States
| | - W. Patrick Walters
- Relay
Therapeutics, 399 Binney Street, Cambridge, Massachusetts 02141, United States
| |
Collapse
|
36
|
Chakrabarti M, Tan YS, Balius TE. Considerations Around Structure-Based Drug Discovery for KRAS Using DOCK. Methods Mol Biol 2024; 2797:67-90. [PMID: 38570453 DOI: 10.1007/978-1-0716-3822-4_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Molecular docking is a popular computational tool in drug discovery. Leveraging structural information, docking software predicts binding poses of small molecules to cavities on the surfaces of proteins. Virtual screening for ligand discovery is a useful application of docking software. In this chapter, using the enigmatic KRAS protein as an example system, we endeavor to teach the reader about best practices for performing molecular docking with UCSF DOCK. We discuss methods for virtual screening and docking molecules on KRAS. We present the following six points to optimize our docking setup for prosecuting a virtual screen: protein structure choice, pocket selection, optimization of the scoring function, modification of sampling spheres and sampling procedures, choosing an appropriate portion of chemical space to dock, and the choice of which top scoring molecules to pick for purchase.
Collapse
Affiliation(s)
- Mayukh Chakrabarti
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Y Stanley Tan
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Trent E Balius
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| |
Collapse
|
37
|
Menchon G, Maveyraud L, Czaplicki G. Molecular Dynamics as a Tool for Virtual Ligand Screening. Methods Mol Biol 2024; 2714:33-83. [PMID: 37676592 DOI: 10.1007/978-1-0716-3441-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Rational drug design is essential for new drugs to emerge, especially when the structure of a target protein or nucleic acid is known. To that purpose, high-throughput virtual ligand screening campaigns aim at discovering computationally new binding molecules or fragments to modulate particular biomolecular interactions or biological activities, related to a disease process. The structure-based virtual ligand screening process primarily relies on docking methods which allow predicting the binding of a molecule to a biological target structure with a correct conformation and the best possible affinity. The docking method itself is not sufficient as it suffers from several and crucial limitations (lack of full protein flexibility information, no solvation and ion effects, poor scoring functions, and unreliable molecular affinity estimation).At the interface of computer techniques and drug discovery, molecular dynamics (MD) allows introducing protein flexibility before or after a docking protocol, refining the structure of protein-drug complexes in the presence of water, ions, and even in membrane-like environments, describing more precisely the temporal evolution of the biological complex and ranking these complexes with more accurate binding energy calculations. In this chapter, we describe the up-to-date MD, which plays the role of supporting tools in the virtual ligand screening (VS) process.Without a doubt, using docking in combination with MD is an attractive approach in structure-based drug discovery protocols nowadays. It has proved its efficiency through many examples in the literature and is a powerful method to significantly reduce the amount of required wet experimentations (Tarcsay et al, J Chem Inf Model 53:2990-2999, 2013; Barakat et al, PLoS One 7:e51329, 2012; De Vivo et al, J Med Chem 59:4035-4061, 2016; Durrant, McCammon, BMC Biol 9:71-79, 2011; Galeazzi, Curr Comput Aided Drug Des 5:225-240, 2009; Hospital et al, Adv Appl Bioinforma Chem 8:37-47, 2015; Jiang et al, Molecules 20:12769-12786, 2015; Kundu et al, J Mol Graph Model 61:160-174, 2015; Mirza et al, J Mol Graph Model 66:99-107, 2016; Moroy et al, Future Med Chem 7:2317-2331, 2015; Naresh et al, J Mol Graph Model 61:272-280, 2015; Nichols et al, J Chem Inf Model 51:1439-1446, 2011; Nichols et al, Methods Mol Biol 819:93-103, 2012; Okimoto et al, PLoS Comput Biol 5:e1000528, 2009; Rodriguez-Bussey et al, Biopolymers 105:35-42, 2016; Sliwoski et al, Pharmacol Rev 66:334-395, 2014).
Collapse
Affiliation(s)
- Grégory Menchon
- Inserm U1242, Oncogenesis, Stress and Signaling (OSS), Université de Rennes 1, Rennes, France
| | - Laurent Maveyraud
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Georges Czaplicki
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France.
| |
Collapse
|
38
|
Yu L, He X, Fang X, Liu L, Liu J. Deep Learning with Geometry-Enhanced Molecular Representation for Augmentation of Large-Scale Docking-Based Virtual Screening. J Chem Inf Model 2023; 63:6501-6514. [PMID: 37882338 DOI: 10.1021/acs.jcim.3c01371] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
Structure-based virtual screening has been a crucial tool in drug discovery for decades. However, as the chemical space expands, the existing structure-based virtual screening techniques based on molecular docking and scoring struggle to handle billion-entry ultralarge libraries due to the high computational cost. To address this challenge, people have resorted to machine learning techniques to enhance structure-based virtual screening for efficiently exploring the vast chemical space. In those cases, compounds are usually treated as sequential strings or two-dimensional topology graphs, limiting their ability to incorporate three-dimensional structural information for downstream tasks. We herein propose a novel deep learning protocol, GEM-Screen, which utilizes the geometry-enhanced molecular representation of the compounds docking to a specific target and is trained on docking scores of a small fraction of a library through an active learning strategy to approximate the docking outcome for yet nontraining entries. This protocol is applied to virtual screening campaigns against the AmpC and D4 targets, demonstrating that GEM-Screen enriches more than 90% of the hit scaffolds for AmpC in the top 4% of model predictions and more than 80% of the hit scaffolds for D4 in the same top-ranking size of library. GEM-Screen can be used in conjunction with traditional docking programs for docking of only the top-ranked compounds to avoid the exhaustive docking of the whole library, thus allowing for discovering top-scoring compounds from billion-entry libraries in a rapid yet accurate fashion.
Collapse
Affiliation(s)
- Lan Yu
- School of Science, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao He
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- New York University-East China Normal University Center for Computational Chemistry, New York University Shanghai, Shanghai 200062, China
| | - Xiaomin Fang
- Baidu International Technology (Shenzhen) Co., Ltd., Shenzhen 518063, China
| | - Lihang Liu
- Baidu International Technology (Shenzhen) Co., Ltd., Shenzhen 518063, China
| | - Jinfeng Liu
- School of Science, China Pharmaceutical University, Nanjing 210009, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
39
|
Silva-Júnior EFD. "You've got the Body I've got the Brains" - Could the current AI-based tools replace the human ingenuity for designing new drug candidates? Bioorg Med Chem 2023; 94:117475. [PMID: 37741120 DOI: 10.1016/j.bmc.2023.117475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/12/2023] [Accepted: 09/12/2023] [Indexed: 09/25/2023]
Abstract
The emergence of artificial intelligence (AI) tools has transformed the landscape of drug discovery, providing unprecedented speed, efficiency, and cost-effectiveness in the search for new therapeutics. From target identification to drug formulation and delivery, AI-driven algorithms have revolutionized various aspects of medicinal chemistry, significantly accelerating the drug design process. Despite the transformative power of AI, this perspective article emphasizes the limitations of AI tools in drug discovery, requiring inventive skills of medicinal chemists. However, the article highlighted that there is a need for a harmonious integration of AI-based tools and human expertise in drug discovery. Such a synergistic approach promises to lead to groundbreaking therapies that address unmet medical needs and benefit humankind. As the world evolves technologically, the question remains: When will AI tools effectively design and develop drugs? The answer may lie in the seamless collaboration between AI and human researchers, unlocking transformative therapies that combat diseases effectively.
Collapse
Affiliation(s)
- Edeildo Ferreira da Silva-Júnior
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, 57072-970 Alagoas, Maceió, Brazil
| |
Collapse
|
40
|
Gehringer M, Pape F, Méndez M, Barbie P, Unzue Lopez A, Lefranc J, Klingler FM, Hessler G, Langer T, Diamanti E, Schiedel M. Back in Person: Frontiers in Medicinal Chemistry 2023. ChemMedChem 2023; 18:e202300344. [PMID: 37485831 DOI: 10.1002/cmdc.202300344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/12/2023] [Indexed: 07/25/2023]
Abstract
The Frontiers in Medicinal Chemistry (FiMC) is the largest international Medicinal Chemistry conference in the German speaking area and took place from April 3rd to 5th 2023 in Vienna (Austria). Fortunately, after being cancelled in 2020 and two years (2021-2022) of entirely virtual meetings, due to the COVID-19 pandemic, the FiMC could be held in a face-to-face format again. Organized by the Division of Medicinal Chemistry of the German Chemical Society (GDCh), the Division of Pharmaceutical and Medicinal Chemistry of the German Pharmaceutical Society (DPhG), together with the Division of Medicinal Chemistry of the Austrian Chemical Society (GÖCH), the Austrian Pharmaceutical Society (ÖPhG), and a local organization committee from the University of Vienna headed by Thierry Langer, the meeting brought together 260 participants from 21 countries. The program included 38 lectures by leading scientists from industry and academia as well as early career investigators. Moreover, 102 posters were presented in two highly interactive poster sessions.
Collapse
Affiliation(s)
- Matthias Gehringer
- Institute of Pharmaceutical Sciences, Pharmaceutical/Medicinal Chemistry Department, University of Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Felix Pape
- NUVISAN Innovation Campus Berlin, NUVISAN ICB GmbH, Muellerstraße 178, 13353, Berlin, Germany
| | - María Méndez
- Sanofi R&D, Integrated Drug Discovery, Industriepark Höchst, Bldg. G838, 65926, Frankfurt am Main, Germany
| | - Philipp Barbie
- Bayer AG, R&D, Pharmaceuticals, Laboratory IV, Bldg. S106, 231, 13342, Berlin, Germany
| | - Andrea Unzue Lopez
- Merck Healthcare KGaA, Frankfurter Straße 250, 64293, Darmstadt, Germany
| | - Julien Lefranc
- Merck Healthcare KGaA, Frankfurter Straße 250, 64293, Darmstadt, Germany
| | | | - Gerhard Hessler
- Sanofi R&D, Integrated Drug Discovery, Industriepark Höchst, Bldg. G877, 65926, Frankfurt am Main, Germany
| | - Thierry Langer
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090, Vienna, Austria
| | - Eleonora Diamanti
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Matthias Schiedel
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106, Braunschweig, Germany
| |
Collapse
|
41
|
Sivula T, Yetukuri L, Kalliokoski T, Käsnänen H, Poso A, Pöhner I. Machine Learning-Boosted Docking Enables the Efficient Structure-Based Virtual Screening of Giga-Scale Enumerated Chemical Libraries. J Chem Inf Model 2023; 63:5773-5783. [PMID: 37655823 PMCID: PMC10523430 DOI: 10.1021/acs.jcim.3c01239] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Indexed: 09/02/2023]
Abstract
The emergence of ultra-large screening libraries, filled to the brim with billions of readily available compounds, poses a growing challenge for docking-based virtual screening. Machine learning (ML)-boosted strategies like the tool HASTEN combine rapid ML prediction with the brute-force docking of small fractions of such libraries to increase screening throughput and take on giga-scale libraries. In our case study of an anti-bacterial chaperone and an anti-viral kinase, we first generated a brute-force docking baseline for 1.56 billion compounds in the Enamine REAL lead-like library with the fast Glide high-throughput virtual screening protocol. With HASTEN, we observed robust recall of 90% of the true 1000 top-scoring virtual hits in both targets when docking only 1% of the entire library. This reduction of the required docking experiments by 99% significantly shortens the screening time. In the kinase target, the employment of a hydrogen bonding constraint resulted in a major proportion of unsuccessful docking attempts and hampered ML predictions. We demonstrate the optimization potential in the treatment of failed compounds when performing ML-boosted screening and benchmark and showcase HASTEN as a fast and robust tool in a growing arsenal of approaches to unlock the chemical space covered by giga-scale screening libraries for everyday drug discovery campaigns.
Collapse
Affiliation(s)
- Toni Sivula
- School
of Pharmacy, University of Eastern Finland, Kuopio FI-70211, Finland
| | | | - Tuomo Kalliokoski
- Computational
Medicine Design, Orion Pharma, Orionintie 1A, Espoo FI-02101, Finland
| | - Heikki Käsnänen
- Computational
Medicine Design, Orion Pharma, Orionintie 1A, Espoo FI-02101, Finland
| | - Antti Poso
- School
of Pharmacy, University of Eastern Finland, Kuopio FI-70211, Finland
- Department
of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical
Sciences, Eberhard Karls University, Tübingen DE-72076, Germany
- Cluster
of Excellence iFIT (EXC 2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, University
of Tübingen, Tübingen DE-72076, Germany
- Tübingen
Center for Academic Drug Discovery & Development (TüCAD2), Tübingen DE-72076, Germany
| | - Ina Pöhner
- School
of Pharmacy, University of Eastern Finland, Kuopio FI-70211, Finland
| |
Collapse
|
42
|
Alnammi M, Liu S, Ericksen SS, Ananiev GE, Voter AF, Guo S, Keck JL, Hoffmann FM, Wildman SA, Gitter A. Evaluating Scalable Supervised Learning for Synthesize-on-Demand Chemical Libraries. J Chem Inf Model 2023; 63:5513-5528. [PMID: 37625010 PMCID: PMC10538940 DOI: 10.1021/acs.jcim.3c00912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Indexed: 08/27/2023]
Abstract
Traditional small-molecule drug discovery is a time-consuming and costly endeavor. High-throughput chemical screening can only assess a tiny fraction of drug-like chemical space. The strong predictive power of modern machine-learning methods for virtual chemical screening enables training models on known active and inactive compounds and extrapolating to much larger chemical libraries. However, there has been limited experimental validation of these methods in practical applications on large commercially available or synthesize-on-demand chemical libraries. Through a prospective evaluation with the bacterial protein-protein interaction PriA-SSB, we demonstrate that ligand-based virtual screening can identify many active compounds in large commercial libraries. We use cross-validation to compare different types of supervised learning models and select a random forest (RF) classifier as the best model for this target. When predicting the activity of more than 8 million compounds from Aldrich Market Select, the RF substantially outperforms a naïve baseline based on chemical structure similarity. 48% of the RF's 701 selected compounds are active. The RF model easily scales to score one billion compounds from the synthesize-on-demand Enamine REAL database. We tested 68 chemically diverse top predictions from Enamine REAL and observed 31 hits (46%), including one with an IC50 value of 1.3 μM.
Collapse
Affiliation(s)
- Moayad Alnammi
- Department
of Computer Sciences, University of Wisconsin−Madison, Madison, Wisconsin 53706, United States
- Morgridge
Institute for Research, Madison, Wisconsin 53715, United States
- Department
of Information and Computer Science, King
Fahd University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
| | - Shengchao Liu
- Department
of Computer Sciences, University of Wisconsin−Madison, Madison, Wisconsin 53706, United States
- Morgridge
Institute for Research, Madison, Wisconsin 53715, United States
| | - Spencer S. Ericksen
- Small
Molecule Screening Facility, University
of Wisconsin−Madison, Madison, Wisconsin 53792, United States
| | - Gene E. Ananiev
- Small
Molecule Screening Facility, University
of Wisconsin−Madison, Madison, Wisconsin 53792, United States
| | - Andrew F. Voter
- Department
of Biomolecular Chemistry, University of
Wisconsin−Madison, Madison, Wisconsin 53706, United States
| | - Song Guo
- Small
Molecule Screening Facility, University
of Wisconsin−Madison, Madison, Wisconsin 53792, United States
| | - James L. Keck
- Department
of Biomolecular Chemistry, University of
Wisconsin−Madison, Madison, Wisconsin 53706, United States
| | - F. Michael Hoffmann
- Small
Molecule Screening Facility, University
of Wisconsin−Madison, Madison, Wisconsin 53792, United States
- McArdle Laboratory
for Cancer Research, University of Wisconsin−Madison, Madison, Wisconsin 53705, United States
| | - Scott A. Wildman
- Small
Molecule Screening Facility, University
of Wisconsin−Madison, Madison, Wisconsin 53792, United States
| | - Anthony Gitter
- Department
of Computer Sciences, University of Wisconsin−Madison, Madison, Wisconsin 53706, United States
- Morgridge
Institute for Research, Madison, Wisconsin 53715, United States
- Department
of Biostatistics and Medical Informatics, University of Wisconsin−Madison, Madison, Wisconsin 53792, United States
| |
Collapse
|
43
|
Abstract
Drug development is a wide scientific field that faces many challenges these days. Among them are extremely high development costs, long development times, and a small number of new drugs that are approved each year. New and innovative technologies are needed to solve these problems that make the drug discovery process of small molecules more time and cost efficient, and that allow previously undruggable receptor classes to be targeted, such as protein-protein interactions. Structure-based virtual screenings (SBVSs) have become a leading contender in this context. In this review, we give an introduction to the foundations of SBVSs and survey their progress in the past few years with a focus on ultralarge virtual screenings (ULVSs). We outline key principles of SBVSs, recent success stories, new screening techniques, available deep learning-based docking methods, and promising future research directions. ULVSs have an enormous potential for the development of new small-molecule drugs and are already starting to transform early-stage drug discovery.
Collapse
Affiliation(s)
- Christoph Gorgulla
- Harvard Medical School and Physics Department, Harvard University, Boston, Massachusetts, USA;
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Current affiliation: Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
44
|
Rinotas V, Liepouri F, Ouzouni MD, Chalkidi N, Papaneophytou C, Lampropoulou M, Vidali VP, Kontopidis G, Couladouros E, Eliopoulos E, Papakyriakou A, Douni E. Structure-Based Discovery of Receptor Activator of Nuclear Factor-κB Ligand (RANKL)-Induced Osteoclastogenesis Inhibitors. Int J Mol Sci 2023; 24:11290. [PMID: 37511048 PMCID: PMC10379842 DOI: 10.3390/ijms241411290] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
Receptor activator of nuclear factor-κB ligand (RANKL) has been actively pursued as a therapeutic target for osteoporosis, given that RANKL is the master mediator of bone resorption as it promotes osteoclast differentiation, activity and survival. We employed a structure-based virtual screening approach comprising two stages of experimental evaluation and identified 11 commercially available compounds that displayed dose-dependent inhibition of osteoclastogenesis. Their inhibitory effects were quantified through TRAP activity at the low micromolar range (IC50 < 5 μΜ), but more importantly, 3 compounds displayed very low toxicity (LC50 > 100 μΜ). We also assessed the potential of an N-(1-aryl-1H-indol-5-yl)aryl-sulfonamide scaffold that was based on the structure of a hit compound, through synthesis of 30 derivatives. Their evaluation revealed 4 additional hits that inhibited osteoclastogenesis at low micromolar concentrations; however, cellular toxicity concerns preclude their further development. Taken together with the structure-activity relationships provided by the hit compounds, our study revealed potent inhibitors of RANKL-induced osteoclastogenesis of high therapeutic index, which bear diverse scaffolds that can be employed in hit-to-lead optimization for the development of therapeutics against osteolytic diseases.
Collapse
Affiliation(s)
- Vagelis Rinotas
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", 34 Fleming Street, 16672 Vari, Greece
| | | | - Maria-Dimitra Ouzouni
- Laboratory of General Chemistry, Department of Food Science and Human Nutrition, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece
| | - Niki Chalkidi
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", 34 Fleming Street, 16672 Vari, Greece
| | - Christos Papaneophytou
- Department of Biochemistry, Veterinary School, University of Thessaly, 224 Trikalon, 43131 Karditsa, Greece
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, 46 Makedonitissas Avenue, 2417 Nicosia, Cyprus
| | | | - Veroniki P Vidali
- Institute of Nanoscience and Nanotechnology, National Centre for Scientific Research "Demokritos", Patr. Gregoriou E & 27 Neapoleos Str, 15341 Athens, Greece
| | - George Kontopidis
- Department of Biochemistry, Veterinary School, University of Thessaly, 224 Trikalon, 43131 Karditsa, Greece
| | - Elias Couladouros
- proACTINA SA, 20 Delfon Street, 15125 Athens, Greece
- Laboratory of General Chemistry, Department of Food Science and Human Nutrition, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece
| | - Elias Eliopoulos
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece
| | - Athanasios Papakyriakou
- Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Patr. Gregoriou E & 27 Neapoleos Str, 15341 Athens, Greece
| | - Eleni Douni
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", 34 Fleming Street, 16672 Vari, Greece
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece
| |
Collapse
|
45
|
Neumann A, Marrison L, Klein R. Relevance of the Trillion-Sized Chemical Space "eXplore" as a Source for Drug Discovery. ACS Med Chem Lett 2023; 14:466-472. [PMID: 37077402 PMCID: PMC10108389 DOI: 10.1021/acsmedchemlett.3c00021] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
Within the past two decades, virtual combinatorial compound collections, so-called chemical spaces, became an important molecule source for pharmaceutical research all over the world. The emergence of compound vendor chemical spaces with rapidly growing numbers of molecules raises questions about their application suitability and the quality of the content. Here, we examine the composition of the recently published and, so far, biggest chemical space, "eXplore", which comprises approximately 2.8 trillion virtual product molecules. The utility of eXplore to retrieve interesting chemistry around approved drugs and common Bemis Murcko scaffolds has been assessed with several methods (FTrees, SpaceLight, SpaceMACS). Further, the overlap between several vendor chemical spaces and a physicochemical property distribution analysis has been performed. Despite the straightforward chemical reactions underlying its setup, eXplore is demonstrated to provide relevant and, most importantly, easily accessible molecules for drug discovery campaigns.
Collapse
Affiliation(s)
| | - Lester Marrison
- eMolecules, 3430 Carmel Mountain Road, Suite
250, San Diego, California 92121, United States
| | - Raphael Klein
- BioSolveIT
GmbH, An der Ziegelei 79, 53757 Sankt Augustin, Germany
| |
Collapse
|
46
|
Korn M, Ehrt C, Ruggiu F, Gastreich M, Rarey M. Navigating large chemical spaces in early-phase drug discovery. Curr Opin Struct Biol 2023; 80:102578. [PMID: 37019067 DOI: 10.1016/j.sbi.2023.102578] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/28/2023] [Accepted: 02/26/2023] [Indexed: 04/07/2023]
Abstract
The size of actionable chemical spaces is surging, owing to a variety of novel techniques, both computational and experimental. As a consequence, novel molecular matter is now at our fingertips that cannot and should not be neglected in early-phase drug discovery. Huge, combinatorial, make-on-demand chemical spaces with high probability of synthetic success rise exponentially in content, generative machine learning models go hand in hand with synthesis prediction, and DNA-encoded libraries offer new ways of hit structure discovery. These technologies enable to search for new chemical matter in a much broader and deeper manner with less effort and fewer financial resources. These transformational developments require new cheminformatics approaches to make huge chemical spaces searchable and analyzable with low resources, and with as little energy consumption as possible. Substantial progress has been made in the past years with respect to computation as well as organic synthesis. First examples of bioactive compounds resulting from the successful use of these novel technologies demonstrate their power to contribute to tomorrow's drug discovery programs. This article gives a compact overview of the state-of-the-art.
Collapse
Affiliation(s)
- Malte Korn
- Universität Hamburg, ZBH - Center for Bioinformatics, Bundesstr. 43, 20146 Hamburg, Germany
| | - Christiane Ehrt
- Universität Hamburg, ZBH - Center for Bioinformatics, Bundesstr. 43, 20146 Hamburg, Germany
| | - Fiorella Ruggiu
- insitro, 279 E Grand Ave., CA 94608, South San Francisco, USA
| | - Marcus Gastreich
- BioSolveIT GmbH, An der Ziegelei 79, 53757 Sankt Augustin, Germany
| | - Matthias Rarey
- Universität Hamburg, ZBH - Center for Bioinformatics, Bundesstr. 43, 20146 Hamburg, Germany.
| |
Collapse
|
47
|
Sadybekov AV, Katritch V. Computational approaches streamlining drug discovery. Nature 2023; 616:673-685. [PMID: 37100941 DOI: 10.1038/s41586-023-05905-z] [Citation(s) in RCA: 369] [Impact Index Per Article: 184.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 03/01/2023] [Indexed: 04/28/2023]
Abstract
Computer-aided drug discovery has been around for decades, although the past few years have seen a tectonic shift towards embracing computational technologies in both academia and pharma. This shift is largely defined by the flood of data on ligand properties and binding to therapeutic targets and their 3D structures, abundant computing capacities and the advent of on-demand virtual libraries of drug-like small molecules in their billions. Taking full advantage of these resources requires fast computational methods for effective ligand screening. This includes structure-based virtual screening of gigascale chemical spaces, further facilitated by fast iterative screening approaches. Highly synergistic are developments in deep learning predictions of ligand properties and target activities in lieu of receptor structure. Here we review recent advances in ligand discovery technologies, their potential for reshaping the whole process of drug discovery and development, as well as the challenges they encounter. We also discuss how the rapid identification of highly diverse, potent, target-selective and drug-like ligands to protein targets can democratize the drug discovery process, presenting new opportunities for the cost-effective development of safer and more effective small-molecule treatments.
Collapse
Affiliation(s)
- Anastasiia V Sadybekov
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
- Center for New Technologies in Drug Discovery and Development, Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
| | - Vsevolod Katritch
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA.
- Center for New Technologies in Drug Discovery and Development, Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA.
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
48
|
Zimmermann RA, Fischer TR, Schwickert M, Nidoieva Z, Schirmeister T, Kersten C. Chemical Space Virtual Screening against Hard-to-Drug RNA Methyltransferases DNMT2 and NSUN6. Int J Mol Sci 2023; 24:ijms24076109. [PMID: 37047081 PMCID: PMC10094593 DOI: 10.3390/ijms24076109] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/20/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Targeting RNA methyltransferases with small molecules as inhibitors or tool compounds is an emerging field of interest in epitranscriptomics and medicinal chemistry. For two challenging RNA methyltransferases that introduce the 5-methylcytosine (m5C) modification in different tRNAs, namely DNMT2 and NSUN6, an ultra-large commercially available chemical space was virtually screened by physicochemical property filtering, molecular docking, and clustering to identify new ligands for those enzymes. Novel chemotypes binding to DNMT2 and NSUN6 with affinities down to KD,app = 37 µM and KD,app = 12 µM, respectively, were identified using a microscale thermophoresis (MST) binding assay. These compounds represent the first molecules with a distinct structure from the cofactor SAM and have the potential to be developed into activity-based probes for these enzymes. Additionally, the challenges and strategies of chemical space docking screens with special emphasis on library focusing and diversification are discussed.
Collapse
Affiliation(s)
- Robert A Zimmermann
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University, Staudingerweg 5, 55128 Mainz, Germany
| | - Tim R Fischer
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University, Staudingerweg 5, 55128 Mainz, Germany
| | - Marvin Schwickert
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University, Staudingerweg 5, 55128 Mainz, Germany
| | - Zarina Nidoieva
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University, Staudingerweg 5, 55128 Mainz, Germany
| | - Tanja Schirmeister
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University, Staudingerweg 5, 55128 Mainz, Germany
| | - Christian Kersten
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University, Staudingerweg 5, 55128 Mainz, Germany
| |
Collapse
|