1
|
Bhatt R, Wang A, Durrant JD. Teaching old docks new tricks with machine learning enhanced ensemble docking. Sci Rep 2024; 14:20722. [PMID: 39237737 PMCID: PMC11377811 DOI: 10.1038/s41598-024-71699-3] [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: 05/25/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024] Open
Abstract
We here introduce Ensemble Optimizer (EnOpt), a machine-learning tool to improve the accuracy and interpretability of ensemble virtual screening (VS). Ensemble VS is an established method for predicting protein/small-molecule (ligand) binding. Unlike traditional VS, which focuses on a single protein conformation, ensemble VS better accounts for protein flexibility by predicting binding to multiple protein conformations. Each compound is thus associated with a spectrum of scores (one score per protein conformation) rather than a single score. To effectively rank and prioritize the molecules for further evaluation (including experimental testing), researchers must select which protein conformations to consider and how best to map each compound's spectrum of scores to a single value, decisions that are system-specific. EnOpt uses machine learning to address these challenges. We perform benchmark VS to show that for many systems, EnOpt ranking distinguishes active compounds from inactive or decoy molecules more effectively than traditional ensemble VS methods. To encourage broad adoption, we release EnOpt free of charge under the terms of the MIT license.
Collapse
Affiliation(s)
- Roshni Bhatt
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Ann Wang
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Jacob D Durrant
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
2
|
McNutt AT, Koes DR. Open-ComBind: harnessing unlabeled data for improved binding pose prediction. J Comput Aided Mol Des 2023; 38:3. [PMID: 38062207 PMCID: PMC10703974 DOI: 10.1007/s10822-023-00544-y] [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: 09/06/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023]
Abstract
Determination of the bound pose of a ligand is a critical first step in many in silico drug discovery tasks. Molecular docking is the main tool for the prediction of non-covalent binding of a protein and ligand system. Molecular docking pipelines often only utilize the information of one ligand binding to the protein despite the commonly held hypothesis that different ligands share binding interactions when bound to the same receptor. Here we describe Open-ComBind, an easy-to-use, open-source version of the ComBind molecular docking pipeline that leverages information from multiple ligands without known bound structures to enhance pose selection. We first create distributions of feature similarities between ligand pose pairs, comparing near-native poses with all sampled docked poses. These distributions capture the likelihood of observing similar features, such as hydrogen bonds or hydrophobic contacts, in different pose configurations. These similarity distributions are then combined with a per-ligand docking score to enhance overall pose selection by 5% and 4.5% for high-affinity and congeneric series helper ligands, respectively. Open-ComBind reduces the average RMSD of ligands in our benchmark dataset by 9.0%. We provide Open-ComBind as an easy-to-use command line and Python API to increase pose prediction performance at www.github.com/drewnutt/open_combind .
Collapse
Affiliation(s)
- Andrew T McNutt
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - David Ryan Koes
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Rehman AU, Khurshid B, Ali Y, Rasheed S, Wadood A, Ng HL, Chen HF, Wei Z, Luo R, Zhang J. Computational approaches for the design of modulators targeting protein-protein interactions. Expert Opin Drug Discov 2023; 18:315-333. [PMID: 36715303 PMCID: PMC10149343 DOI: 10.1080/17460441.2023.2171396] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 01/18/2023] [Indexed: 01/31/2023]
Abstract
BACKGROUND Protein-protein interactions (PPIs) are intriguing targets for designing novel small-molecule inhibitors. The role of PPIs in various infectious and neurodegenerative disorders makes them potential therapeutic targets . Despite being portrayed as undruggable targets, due to their flat surfaces, disorderedness, and lack of grooves. Recent progresses in computational biology have led researchers to reconsider PPIs in drug discovery. AREAS COVERED In this review, we introduce in-silico methods used to identify PPI interfaces and present an in-depth overview of various computational methodologies that are successfully applied to annotate the PPIs. We also discuss several successful case studies that use computational tools to understand PPIs modulation and their key roles in various physiological processes. EXPERT OPINION Computational methods face challenges due to the inherent flexibility of proteins, which makes them expensive, and result in the use of rigid models. This problem becomes more significant in PPIs due to their flexible and flat interfaces. Computational methods like molecular dynamics (MD) simulation and machine learning can integrate the chemical structure data into biochemical and can be used for target identification and modulation. These computational methodologies have been crucial in understanding the structure of PPIs, designing PPI modulators, discovering new drug targets, and predicting treatment outcomes.
Collapse
Affiliation(s)
- Ashfaq Ur Rehman
- Departments of Molecular Biology and Biochemistry, Chemical and Biomolecular Engineering, Materials Science and Engineering, and Biomedical Engineering, Graduate Program in Chemical and Materials Physics, University of California Irvine, Irvine, California, USA
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai, Zhejiang, China
| | - Beenish Khurshid
- Department of Biochemistry, Abdul Wali Khan University Mardan, Pakistan
| | - Yasir Ali
- National Center for Bioinformatics, Quaid-e-Azam University, Islamabad, Pakistan
| | - Salman Rasheed
- National Center for Bioinformatics, Quaid-e-Azam University, Islamabad, Pakistan
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Pakistan
| | - Ho-Leung Ng
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas, USA
| | - Hai-Feng Chen
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, Zhejiang, China
| | - Zhiqiang Wei
- Medicinal Chemistry and Bioinformatics Center, Ocean University of China, Qingdao, Shandong, China
| | - Ray Luo
- Departments of Molecular Biology and Biochemistry, Chemical and Biomolecular Engineering, Materials Science and Engineering, and Biomedical Engineering, Graduate Program in Chemical and Materials Physics, University of California Irvine, Irvine, California, USA
| | - Jian Zhang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai, Zhejiang, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Moreira JS, Galvão DS, Xavier CFC, Cunha S, Pita SSDR, Reis JN, Freitas HFD. Phenotypic and in silico studies for a series of synthetic thiosemicarbazones as New Delhi metallo-beta-lactamase carbapenemase inhibitors. J Biomol Struct Dyn 2022; 40:14223-14235. [PMID: 34766882 DOI: 10.1080/07391102.2021.2001379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The past two decades have been marked by a global spread of bacterial resistance to β-lactam drugs and carbapenems derivatives are the ultimate treatment against multidrug-resistant bacteria. β-lactamase expression is related to resistance which demands the development of bacterial resistance blockers. Drug inhibitor combinations of serine-β-lactamase and β-lactam were successful employed in therapy despite their inactivity against New Delhi metallo-beta-lactamase (NDM). Until now, few compounds are active against NDM-producing bacteria and no specific inhibitors are available yet. The rational strategy for NDM inhibitors development starts with in vitro assays aiming to seek compounds that could act synergistically with β-lactam antibiotics. Thus, eight thiosemicarbazone derivatives were synthesized and investigated for their ability to reverse the resistant phenotype in NDM in Enterobacter cloacae. Phenotypic screening indicated that four isatin-beta-thiosemicarbazones showed Fractional Inhibitory Concentration (FIC) ≤ 250 µM in the presence of meropenem (4 µg/mL). The most promising compound (FIC= 31.25 µM) also presented synergistic effect (FICI = 0.34). Docking and molecular dynamics studies on NDM-thiosemicarbazone complex suggested that 2,3-dihydro-1H-indol-2-one subunit interacts with catalytic zinc and interacted through hydrogen bonds with Asp124 acting like a carboxylic acid bioisostere. Additionally, thiosemicarbazone tautomer with oxidized sulfur (thione) seems to act as a spacer rather than zinc chelator, and the aromatic moieties are stabilized by pi-pi and cation-pi interactions with His189 and Lys221 residues. Our results addressed some thiosemicarbazone structural changes to increase its biological activity against NDM and highlight its scaffold as promising alternatives to treat bacterial resistance.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Jonatham Souza Moreira
- Post-Graduation Program in Pharmacy, Pharmacy College, Federal University of Bahia, Salvador, Bahia, Brazil
| | | | | | - Silvio Cunha
- Chemistry Institute, Federal University of Bahia, Ondina, Salvador, Bahia, Brazil
| | - Samuel Silva da Rocha Pita
- Pharmacy College, Federal University of Bahia, Salvador, Bahia, Brazil.,Bioinformatics and Molecular Modeling Laboratory (LaBiMM), Federal University of Bahia, Salvador, Bahia, Brazil
| | - Joice Neves Reis
- Post-Graduation Program in Pharmacy, Pharmacy College, Federal University of Bahia, Salvador, Bahia, Brazil.,Pharmacy College, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Humberto Fonseca de Freitas
- Post-Graduation Program in Pharmacy, Pharmacy College, Federal University of Bahia, Salvador, Bahia, Brazil.,Pharmacy College, Federal University of Bahia, Salvador, Bahia, Brazil.,Bioinformatics and Molecular Modeling Laboratory (LaBiMM), Federal University of Bahia, Salvador, Bahia, Brazil
| |
Collapse
|
5
|
Miñarro-Lleonar M, Ruiz-Carmona S, Alvarez-Garcia D, Schmidtke P, Barril X. Development of an Automatic Pipeline for Participation in the CELPP Challenge. Int J Mol Sci 2022; 23:ijms23094756. [PMID: 35563148 PMCID: PMC9105952 DOI: 10.3390/ijms23094756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 12/01/2022] Open
Abstract
The prediction of how a ligand binds to its target is an essential step for Structure-Based Drug Design (SBDD) methods. Molecular docking is a standard tool to predict the binding mode of a ligand to its macromolecular receptor and to quantify their mutual complementarity, with multiple applications in drug design. However, docking programs do not always find correct solutions, either because they are not sampled or due to inaccuracies in the scoring functions. Quantifying the docking performance in real scenarios is essential to understanding their limitations, managing expectations and guiding future developments. Here, we present a fully automated pipeline for pose prediction validated by participating in the Continuous Evaluation of Ligand Pose Prediction (CELPP) Challenge. Acknowledging the intrinsic limitations of the docking method, we devised a strategy to automatically mine and exploit pre-existing data, defining—whenever possible—empirical restraints to guide the docking process. We prove that the pipeline is able to generate predictions for most of the proposed targets as well as obtain poses with low RMSD values when compared to the crystal structure. All things considered, our pipeline highlights some major challenges in the automatic prediction of protein–ligand complexes, which will be addressed in future versions of the pipeline.
Collapse
Affiliation(s)
- Marina Miñarro-Lleonar
- Pharmacy Faculty, University of Barcelona, Av. de Joan XXIII 27-31, 08028 Barcelona, Spain;
| | | | - Daniel Alvarez-Garcia
- GAIN Therapeutics, Parc Cientific de Barcelona, Baldiri i Reixac 10, 08029 Barcelona, Spain;
| | - Peter Schmidtke
- Discngine S.A.S., 79 Avenue Ledru Rollin, 75012 Paris, France;
| | - Xavier Barril
- Pharmacy Faculty, University of Barcelona, Av. de Joan XXIII 27-31, 08028 Barcelona, Spain;
- GAIN Therapeutics, Parc Cientific de Barcelona, Baldiri i Reixac 10, 08029 Barcelona, Spain;
- Catalan Institute for Research and Advanced Studies (ICREA), Passeig de Lluis Companys 23, 08010 Barcelona, Spain
- Correspondence:
| |
Collapse
|
6
|
A Structure Based Study of Selective Inhibition of Factor IXa over Factor Xa. Molecules 2021; 26:molecules26175372. [PMID: 34500804 PMCID: PMC8434132 DOI: 10.3390/molecules26175372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 11/25/2022] Open
Abstract
Blood coagulation is an essential physiological process for hemostasis; however, abnormal coagulation can lead to various potentially fatal disorders, generally known as thromboembolic disorders, which are a major cause of mortality in the modern world. Recently, the FDA has approved several anticoagulant drugs for Factor Xa (FXa) which work via the common pathway of the coagulation cascade. A main side effect of these drugs is the potential risk for bleeding in patients. Coagulation Factor IXa (FIXa) has recently emerged as the strategic target to ease these risks as it selectively regulates the intrinsic pathway. These aforementioned coagulation factors are highly similar in structure, functional architecture, and inhibitor binding mode. Therefore, it remains a challenge to design a selective inhibitor which may affect only FIXa. With the availability of a number of X-ray co-crystal structures of these two coagulation factors as protein–ligand complexes, structural alignment, molecular docking, and pharmacophore modeling were employed to derive the relevant criteria for selective inhibition of FIXa over FXa. In this study, six ligands (three potent, two selective, and one inactive) were selected for FIXa inhibition and six potent ligands (four FDA approved drugs) were considered for FXa. The pharmacophore hypotheses provide the distribution patterns for the principal interactions that take place in the binding site. None of the pharmacophoric patterns of the FXa inhibitors matched with any of the patterns of FIXa inhibitors. Based on pharmacophore analysis, a selectivity of a ligand for FIXa over FXa may be defined quantitatively as a docking score of lower than −8.0 kcal/mol in the FIXa-grids and higher than −7.5 kcal/mol in the FXa-grids.
Collapse
|
7
|
Lian J. An optimization model of cross-docking scheduling of cold chain logistics based on fuzzy time window. JOURNAL OF INTELLIGENT & FUZZY SYSTEMS 2021. [DOI: 10.3233/jifs-210611] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In order to improve the distribution efficiency of cold chain logistics and reduce the distribution cost, an optimization model of cross-docking scheduling of cold chain logistics based on fuzzy time window is constructed. According to the complexity of cold chain logistics network, a multi-objective optimization model of cross-docking scheduling of cold chain logistics vehicle routing with fuzzy time window is established. In order to ensure the lowest total cost of cold chain logistics distribution and improve the overall customer satisfaction with service time, the Drosophila optimization algorithm is used to solve the model to obtain the optimal vehicle routing of cross-docking scheduling optimization of cold chain logistics. The simulation test results show that: after the application of the model, the cold chain logistics distribution time is significantly shortened, the distribution cost is significantly reduced, the damage cost is reduced, the carbon emission of vehicles is reduced, and the economic and low-carbon benefits are significantly improved, which can be used as an effective tool to solve the cross-docking scheduling optimization problem of cold chain logistics.
Collapse
Affiliation(s)
- Jie Lian
- Fujian Business University, Fuzhou, Fujian, China
| |
Collapse
|
8
|
Fischer A, Sellner M, Mitusińska K, Bzówka M, Lill MA, Góra A, Smieško M. Computational Selectivity Assessment of Protease Inhibitors against SARS-CoV-2. Int J Mol Sci 2021; 22:2065. [PMID: 33669738 PMCID: PMC7922391 DOI: 10.3390/ijms22042065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 12/27/2022] Open
Abstract
The pandemic of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a serious global health threat. Since no specific therapeutics are available, researchers around the world screened compounds to inhibit various molecular targets of SARS-CoV-2 including its main protease (Mpro) essential for viral replication. Due to the high urgency of these discovery efforts, off-target binding, which is one of the major reasons for drug-induced toxicity and safety-related drug attrition, was neglected. Here, we used molecular docking, toxicity profiling, and multiple molecular dynamics (MD) protocols to assess the selectivity of 33 reported non-covalent inhibitors of SARS-CoV-2 Mpro against eight proteases and 16 anti-targets. The panel of proteases included SARS-CoV Mpro, cathepsin G, caspase-3, ubiquitin carboxy-terminal hydrolase L1 (UCHL1), thrombin, factor Xa, chymase, and prostasin. Several of the assessed compounds presented considerable off-target binding towards the panel of proteases, as well as the selected anti-targets. Our results further suggest a high risk of off-target binding to chymase and cathepsin G. Thus, in future discovery projects, experimental selectivity assessment should be directed toward these proteases. A systematic selectivity assessment of SARS-CoV-2 Mpro inhibitors, as we report it, was not previously conducted.
Collapse
Affiliation(s)
- André Fischer
- Computational Pharmacy, Departement of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (A.F.); (M.S.)
| | - Manuel Sellner
- Computational Pharmacy, Departement of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (A.F.); (M.S.)
| | - Karolina Mitusińska
- Tunneling Group, Biotechnology Centre, ul. Krzywoustego 8, Silesian University of Technology, 44-100 Gliwice, Poland; (K.M.); (M.B.)
| | - Maria Bzówka
- Tunneling Group, Biotechnology Centre, ul. Krzywoustego 8, Silesian University of Technology, 44-100 Gliwice, Poland; (K.M.); (M.B.)
| | - Markus A. Lill
- Computational Pharmacy, Departement of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (A.F.); (M.S.)
| | - Artur Góra
- Tunneling Group, Biotechnology Centre, ul. Krzywoustego 8, Silesian University of Technology, 44-100 Gliwice, Poland; (K.M.); (M.B.)
| | - Martin Smieško
- Computational Pharmacy, Departement of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (A.F.); (M.S.)
| |
Collapse
|
9
|
Vázquez J, López M, Gibert E, Herrero E, Luque FJ. Merging Ligand-Based and Structure-Based Methods in Drug Discovery: An Overview of Combined Virtual Screening Approaches. Molecules 2020; 25:E4723. [PMID: 33076254 PMCID: PMC7587536 DOI: 10.3390/molecules25204723] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/06/2020] [Accepted: 10/11/2020] [Indexed: 12/20/2022] Open
Abstract
Virtual screening (VS) is an outstanding cornerstone in the drug discovery pipeline. A variety of computational approaches, which are generally classified as ligand-based (LB) and structure-based (SB) techniques, exploit key structural and physicochemical properties of ligands and targets to enable the screening of virtual libraries in the search of active compounds. Though LB and SB methods have found widespread application in the discovery of novel drug-like candidates, their complementary natures have stimulated continued efforts toward the development of hybrid strategies that combine LB and SB techniques, integrating them in a holistic computational framework that exploits the available information of both ligand and target to enhance the success of drug discovery projects. In this review, we analyze the main strategies and concepts that have emerged in the last years for defining hybrid LB + SB computational schemes in VS studies. Particularly, attention is focused on the combination of molecular similarity and docking, illustrating them with selected applications taken from the literature.
Collapse
Affiliation(s)
- Javier Vázquez
- Pharmacelera, Plaça Pau Vila, 1, Sector C 2a, Edificio Palau de Mar, 08039 Barcelona, Spain;
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), and Institute of Theoretical and Computational Chemistry (IQTC-UB), University of Barcelona, Av. Prat de la Riba 171, E-08921 Santa Coloma de Gramanet, Spain
| | - Manel López
- AB Science, Parc Scientifique de Luminy, Zone Luminy Enterprise, Case 922, 163 Av. de Luminy, 13288 Marseille, France;
| | - Enric Gibert
- Pharmacelera, Plaça Pau Vila, 1, Sector C 2a, Edificio Palau de Mar, 08039 Barcelona, Spain;
| | - Enric Herrero
- Pharmacelera, Plaça Pau Vila, 1, Sector C 2a, Edificio Palau de Mar, 08039 Barcelona, Spain;
| | - F. Javier Luque
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), and Institute of Theoretical and Computational Chemistry (IQTC-UB), University of Barcelona, Av. Prat de la Riba 171, E-08921 Santa Coloma de Gramanet, Spain
| |
Collapse
|
10
|
Computational methods-guided design of modulators targeting protein-protein interactions (PPIs). Eur J Med Chem 2020; 207:112764. [PMID: 32871340 DOI: 10.1016/j.ejmech.2020.112764] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/09/2020] [Accepted: 08/16/2020] [Indexed: 12/15/2022]
Abstract
Protein-protein interactions (PPIs) play a pivotal role in extensive biological processes and are thus crucial to human health and the development of disease states. Due to their critical implications, PPIs have been spotlighted as promising drug targets of broad-spectrum therapeutic interests. However, owing to the general properties of PPIs, such as flat surfaces, featureless conformations, difficult topologies, and shallow pockets, previous attempts were faced with serious obstacles when targeting PPIs and almost portrayed them as "intractable" for decades. To date, rapid progress in computational chemistry and structural biology methods has promoted the exploitation of PPIs in drug discovery. These techniques boost their cost-effective and high-throughput traits, and enable the study of dynamic PPI interfaces. Thus, computational methods represent an alternative strategy to target "undruggable" PPI interfaces and have attracted intense pharmaceutical interest in recent years, as exemplified by the accumulating number of successful cases. In this review, we first introduce a diverse set of computational methods used to design PPI modulators. Herein, we focus on the recent progress in computational strategies and provide a comprehensive overview covering various methodologies. Importantly, a list of recently-reported successful examples is highlighted to verify the feasibility of these computational approaches. Finally, we conclude the general role of computational methods in targeting PPIs, and also discuss future perspectives on the development of such aids.
Collapse
|
11
|
Parks CD, Gaieb Z, Chiu M, Yang H, Shao C, Walters WP, Jansen JM, McGaughey G, Lewis RA, Bembenek SD, Ameriks MK, Mirzadegan T, Burley SK, Amaro RE, Gilson MK. D3R grand challenge 4: blind prediction of protein-ligand poses, affinity rankings, and relative binding free energies. J Comput Aided Mol Des 2020; 34:99-119. [PMID: 31974851 PMCID: PMC7261493 DOI: 10.1007/s10822-020-00289-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022]
Abstract
The Drug Design Data Resource (D3R) aims to identify best practice methods for computer aided drug design through blinded ligand pose prediction and affinity challenges. Herein, we report on the results of Grand Challenge 4 (GC4). GC4 focused on proteins beta secretase 1 and Cathepsin S, and was run in an analogous manner to prior challenges. In Stage 1, participant ability to predict the pose and affinity of BACE1 ligands were assessed. Following the completion of Stage 1, all BACE1 co-crystal structures were released, and Stage 2 tested affinity rankings with co-crystal structures. We provide an analysis of the results and discuss insights into determined best practice methods.
Collapse
Affiliation(s)
- Conor D Parks
- Drug Design Data Resource, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Zied Gaieb
- Drug Design Data Resource, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Michael Chiu
- Drug Design Data Resource, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Huanwang Yang
- RCSB Protein Data Bank, Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08903, USA
- San Diego Supercomputer Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Chenghua Shao
- RCSB Protein Data Bank, Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08903, USA
- San Diego Supercomputer Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | | | - Johanna M Jansen
- Novartis Institutes for BioMedical Research, Emeryville, CA, 94608, USA
| | | | - Richard A Lewis
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002, Basel, Switzerland
| | | | | | | | - Stephen K Burley
- RCSB Protein Data Bank, Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08903, USA
- San Diego Supercomputer Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Rommie E Amaro
- Drug Design Data Resource, University of California, San Diego, La Jolla, CA, 92093, USA.
- Department of Chemistry and Biochemistry, UC San Diego, La Jolla, CA, 92093-0340, USA.
| | - Michael K Gilson
- Drug Design Data Resource, University of California, San Diego, La Jolla, CA, 92093, USA.
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, 9500 Gilman Drive, MC0751, La Jolla, CA, 92093, USA.
| |
Collapse
|
12
|
Molecular Docking: Shifting Paradigms in Drug Discovery. Int J Mol Sci 2019; 20:ijms20184331. [PMID: 31487867 PMCID: PMC6769923 DOI: 10.3390/ijms20184331] [Citation(s) in RCA: 1068] [Impact Index Per Article: 178.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/02/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022] Open
Abstract
Molecular docking is an established in silico structure-based method widely used in drug discovery. Docking enables the identification of novel compounds of therapeutic interest, predicting ligand-target interactions at a molecular level, or delineating structure-activity relationships (SAR), without knowing a priori the chemical structure of other target modulators. Although it was originally developed to help understanding the mechanisms of molecular recognition between small and large molecules, uses and applications of docking in drug discovery have heavily changed over the last years. In this review, we describe how molecular docking was firstly applied to assist in drug discovery tasks. Then, we illustrate newer and emergent uses and applications of docking, including prediction of adverse effects, polypharmacology, drug repurposing, and target fishing and profiling, discussing also future applications and further potential of this technique when combined with emergent techniques, such as artificial intelligence.
Collapse
|
13
|
Wagner JR, Churas CP, Liu S, Swift RV, Chiu M, Shao C, Feher VA, Burley SK, Gilson MK, Amaro RE. Continuous Evaluation of Ligand Protein Predictions: A Weekly Community Challenge for Drug Docking. Structure 2019; 27:1326-1335.e4. [PMID: 31257108 DOI: 10.1016/j.str.2019.05.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/14/2019] [Accepted: 05/30/2019] [Indexed: 12/19/2022]
Abstract
Docking calculations can accelerate drug discovery by predicting the bound poses of ligands for a targeted protein. However, it is not clear which docking methods work best. Furthermore, predicting poses requires steps outside the docking algorithm itself, such as preparation of the protein and ligand, and it is not known which components are most in need of improvement. The Continuous Evaluation of Ligand Protein Predictions (CELPP) is a blinded prediction challenge designed to address these issues. Participants create a workflow to predict protein-ligand binding poses, which is then tasked with predicting 10-100 new protein-ligand crystal structures each week. CELPP evaluates the accuracy of each workflow's predictions and posts the scores online. The results can be used to identify the strengths and weaknesses of current approaches, help map docking problems to the algorithms most likely to overcome them, and illuminate areas of unmet need in structure-guided drug design.
Collapse
Affiliation(s)
- Jeffrey R Wagner
- Drug Design Data Resource, University of California San Diego, La Jolla, CA 92093, USA
| | - Christopher P Churas
- Drug Design Data Resource, University of California San Diego, La Jolla, CA 92093, USA
| | - Shuai Liu
- Drug Design Data Resource, University of California San Diego, La Jolla, CA 92093, USA
| | - Robert V Swift
- Drug Design Data Resource, University of California San Diego, La Jolla, CA 92093, USA
| | - Michael Chiu
- Drug Design Data Resource, University of California San Diego, La Jolla, CA 92093, USA
| | - Chenghua Shao
- RCSB Protein Data Bank, Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Victoria A Feher
- Drug Design Data Resource, University of California San Diego, La Jolla, CA 92093, USA
| | - Stephen K Burley
- RCSB Protein Data Bank, Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Michael K Gilson
- Drug Design Data Resource, University of California San Diego, La Jolla, CA 92093, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA.
| | - Rommie E Amaro
- Drug Design Data Resource, University of California San Diego, La Jolla, CA 92093, USA; Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
14
|
Raman EP. Template-Based Method for Conformation Generation and Scoring for Congeneric Series of Ligands. J Chem Inf Model 2019; 59:2690-2701. [PMID: 31045363 DOI: 10.1021/acs.jcim.9b00032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Physics-based prediction of protein-ligand binding affinities for a congeneric series of ligands in lead optimization requires their geometries as a first step. In this paper, we report a method that uses the 3D conformation of a lead compound in complex with a protein as a template to generate conformations of a series of related analog compounds. The method uses the Maximal Common Substructure (MCS) computed between lead and analog ligands to assign coordinates for the atoms shared between the ligands. For the differing atoms, a conformation generation procedure is implemented that results in a diversity of conformations. The generated conformations are sorted using a score based on the Molecular Mechanics and Generalized Born with Solvent Accessible Surface Area contribution (MM-GBSA) method. The accuracy of the generated conformations is tested retrospectively using a cross-validation approach applied to four data sets obtained from the Drug Design Data Resource (D3R) by measuring the RMSD of the top scored conformation with respect to the crystallographic pose. The scoring ability of the method is independently assessed using data for the same protein targets to test the rank ordering ability and separating active and inactive ligands. We tested the effect of protein flexibility during structural optimization and scoring approaches with and without strain energies. Retrospective validation on data sets comprising 4 targets shows that the method outperforms random selection for all targets and outperforms a molecular weight-based null model in 3 out of 4 targets in separating active and inactive compounds. Therefore, the presented method is expected to be of utility in lead optimization for rapidly screening analog ligands and generating initial conformations for use in more detailed physics-based binding affinity prediction methods.
Collapse
Affiliation(s)
- E Prabhu Raman
- BIOVIA, Dassault Systemes, 5005 Wateridge Vista Drive , San Diego , California 92121 , United States
| |
Collapse
|
15
|
Mavrogeni ME, Pronios F, Zareifi D, Vasilakaki S, Lozach O, Alexopoulos L, Meijer L, Myrianthopoulos V, Mikros E. A facile consensus ranking approach enhances virtual screening robustness and identifies a cell-active DYRK1α inhibitor. Future Med Chem 2018; 10:2411-2430. [PMID: 30325204 PMCID: PMC6479281 DOI: 10.4155/fmc-2018-0198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 08/16/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Virtual screening is vital for contemporary drug discovery but striking performance fluctuations are commonly encountered, thus hampering error-free use. Results and Methodology: A conceptual framework is suggested for combining screening algorithms characterized by orthogonality (docking-scoring calculations, 3D shape similarity, 2D fingerprint similarity) into a simple, efficient and expansible python-based consensus ranking scheme. An original experimental dataset is created for comparing individual screening methods versus the novel approach. Its utilization leads to identification and phosphoproteomic evaluation of a cell-active DYRK1α inhibitor. CONCLUSION Consensus ranking considerably stabilizes screening performance at reasonable computational cost, whereas individual screens are heavily dependent on calculation settings. Results indicate that the novel approach, currently available as a free online tool, is highly suitable for prospective screening by nonexperts.
Collapse
Affiliation(s)
- Maria E Mavrogeni
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Athens, Panepistimiopolis Zografou, 157 71 Athens, Greece
| | - Filippos Pronios
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Athens, Panepistimiopolis Zografou, 157 71 Athens, Greece
| | - Danae Zareifi
- ProtATonce Ltd, Dimokritos Science Park, Agia Paraskevi, 153 43 Athens, Greece
| | - Sofia Vasilakaki
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Athens, Panepistimiopolis Zografou, 157 71 Athens, Greece
| | - Olivier Lozach
- Laboratoire Chimie Electrochimie Moléculaires et Chimie Analytique, University of Brest, 29238 Brest, France
| | - Leonidas Alexopoulos
- School of Mechanical Engineering, National Technical University of Athens, 157 80 Athens, Greece
| | - Laurent Meijer
- ManRos Therapeutics, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Vassilios Myrianthopoulos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Athens, Panepistimiopolis Zografou, 157 71 Athens, Greece
- ‘Athena’ Research & Innovation Center, 151 25 Athens, Greece
| | - Emmanuel Mikros
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Athens, Panepistimiopolis Zografou, 157 71 Athens, Greece
- ‘Athena’ Research & Innovation Center, 151 25 Athens, Greece
| |
Collapse
|
16
|
Kumar A, Zhang KYJ. Shape similarity guided pose prediction: lessons from D3R Grand Challenge 3. J Comput Aided Mol Des 2018; 33:47-59. [PMID: 30084081 DOI: 10.1007/s10822-018-0142-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 08/01/2018] [Indexed: 12/15/2022]
Abstract
To extend the utility of ligand 3D shape similarity into pose prediction and virtual screening, we have previously developed CDVS and PoPSS methods. Both of them utilize ligand 3D shape similarity with the crystallographic ligands to improve pose prediction. While CDVS utilizes shape similarity to select suitable receptor structures for molecular docking, PoPSS places a ligand conformation of the highest shape similarity with crystal ligands into the target protein binding pocket which is then refined by side-chain repacking and Monte Carlo energy minimization. Analyses of PoPSS revealed some drawbacks in ligand conformation generation and the scoring scheme used. Moreover, as PoPSS does not sample the ligand conformation after placing it in the binding pocket, it relies solely on conformation generation methods to produce native like conformations. To address these limitations of PoPSS method, we report here a modified approach named as PoPSS-Lite, where side-chain repacking was replaced by a simple grid-based energy minimization. This modification also allowed the sampling of terminal functional groups while keeping the core scaffold fixed. Furthermore, shape similarity calculations were improved by increasing the number of ligand conformations and using a different similarity metric. The performance of PoPSS-Lite was prospectively evaluated in D3R GC3. Comparison of PoPSS-Lite demonstrated superior performance over PoPSS and CDVS with lower mean and median RMSDs. Furthermore, comparison with other D3R GC3 pose prediction submissions revealed top performance for PoPSS-Lite. Our D3R GC3 result extends our perspective that ligand 3D shape similarity with known crystallographic information can be successfully used to predict the binding pose of ligands with unknown binding modes. Our D3R GC3 results further highlight the necessity for improvement in conformer generation methods in order to improve shape similarity guided pose prediction.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan
| | - Kam Y J Zhang
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.
| |
Collapse
|
17
|
Kumar A, Zhang KYJ. Advances in the Development of Shape Similarity Methods and Their Application in Drug Discovery. Front Chem 2018; 6:315. [PMID: 30090808 PMCID: PMC6068280 DOI: 10.3389/fchem.2018.00315] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/09/2018] [Indexed: 12/21/2022] Open
Abstract
Molecular similarity is a key concept in drug discovery. It is based on the assumption that structurally similar molecules frequently have similar properties. Assessment of similarity between small molecules has been highly effective in the discovery and development of various drugs. Especially, two-dimensional (2D) similarity approaches have been quite popular due to their simplicity, accuracy and efficiency. Recently, the focus has been shifted toward the development of methods involving the representation and comparison of three-dimensional (3D) conformation of small molecules. Among the 3D similarity methods, evaluation of shape similarity is now gaining attention for its application not only in virtual screening but also in molecular target prediction, drug repurposing and scaffold hopping. A wide range of methods have been developed to describe molecular shape and to determine the shape similarity between small molecules. The most widely used methods include atom distance-based methods, surface-based approaches such as spherical harmonics and 3D Zernike descriptors, atom-centered Gaussian overlay based representations. Several of these methods demonstrated excellent virtual screening performance not only retrospectively but also prospectively. In addition to methods assessing the similarity between small molecules, shape similarity approaches have been developed to compare shapes of protein structures and binding pockets. Additionally, shape comparisons between atomic models and 3D density maps allowed the fitting of atomic models into cryo-electron microscopy maps. This review aims to summarize the methodological advances in shape similarity assessment highlighting advantages, disadvantages and their application in drug discovery.
Collapse
Affiliation(s)
| | - Kam Y. J. Zhang
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, Yokohama, Japan
| |
Collapse
|
18
|
Berishvili VP, Voronkov AE, Radchenko EV, Palyulin VA. Machine Learning Classification Models to Improve the Docking-based Screening: A Case of PI3K-Tankyrase Inhibitors. Mol Inform 2018; 37:e1800030. [DOI: 10.1002/minf.201800030] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/28/2018] [Indexed: 01/20/2023]
Affiliation(s)
- Vladimir P. Berishvili
- Department of Chemistry; Lomonosov Moscow State University; Leninskie gory 1/3 Moscow 119991 Russia
| | - Andrew E. Voronkov
- Department of Chemistry; Lomonosov Moscow State University; Leninskie gory 1/3 Moscow 119991 Russia
- Digital BioPharm Ltd.; Hovseterveien 42 A, H0301 Oslo 0768 Norway
| | - Eugene V. Radchenko
- Department of Chemistry; Lomonosov Moscow State University; Leninskie gory 1/3 Moscow 119991 Russia
| | - Vladimir A. Palyulin
- Department of Chemistry; Lomonosov Moscow State University; Leninskie gory 1/3 Moscow 119991 Russia
| |
Collapse
|