1
|
Koby SB, Gutkin E, Patel S, Kurnikova MG. Automated On-the-Fly Optimization of Resource Allocation for Efficient Free Energy Simulations. J Chem Inf Model 2025; 65:4932-4951. [PMID: 40328725 DOI: 10.1021/acs.jcim.4c02107] [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: 05/08/2025]
Abstract
Computing the free energy of protein-ligand binding by employing molecular dynamics (MD) simulations is becoming a valuable tool in the early stages of drug discovery. However, the cost and complexity of such simulations are often prohibitive for high-throughput studies. We present an automated workflow for the thermodynamic integration scheme with the "on-the-fly" optimization of computational resource allocation for each λ-window of both relative and absolute binding free energy simulations. This iterative workflow utilizes automatic equilibration detection and convergence testing via the Jensen-Shannon distance to determine optimal simulation stopping points in an entirely data-driven manner. It is broadly applicable to multiple free energy calculations, such as ligand binding, amino acid mutations, and others, while utilizing different estimators, e.g., free energy perturbation, BAR, MBAR, etc. We benchmark our workflow on the well-characterized systems, namely, cyclin-dependent kinase 2 and T4 lysozyme L99A/M102Q mutant, and the more flexible SARS-CoV-2 papain-like protease. We demonstrate that this proposed protocol can achieve more than 85% reduction in computational expense while maintaining similar levels of accuracy compared to other benchmarking protocols. We examine the performance of this protocol on both small and large molecular transformations. The cost-accuracy tradeoff of repeated runs is also investigated.
Collapse
Affiliation(s)
- S Benjamin Koby
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Evgeny Gutkin
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Shree Patel
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Maria G Kurnikova
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
2
|
Lindahl E, Friedman R. Estimation of Absolute Binding Free Energies for Drugs That Bind Multiple Proteins. J Chem Inf Model 2025; 65:3431-3438. [PMID: 40163897 PMCID: PMC12004531 DOI: 10.1021/acs.jcim.4c01555] [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: 08/28/2024] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
The Gibbs energy of binding (absolute binding free energy, ABFE) of a drug to proteins in the body determines the drug's affinity to its molecular target and its selectivity. ABFE is challenging to measure, and experimental values are not available for many proteins together with potential drugs and other molecules that bind them. Accurate means of calculating such values are, therefore, highly in demand. Realizing that toxicity and side effects are closely related to off-target binding, here we calculate the ABFE of two drugs, each to multiple proteins, in order to examine whether it is possible to carry out such calculations and achieve the required accuracy. The methods that were used were free energy perturbation with replica exchange molecular dynamics (FEP/REMD) and density functional theory (DFT) with a cluster approach and a simplified model. DFT calculations were supplemented with energy decomposition analysis (EDA). The accuracy of each method is discussed, and suggestions are made for the approach toward better ABFE calculations.
Collapse
Affiliation(s)
- Erik Lindahl
- Department of Chemistry and
Biomedical Sciences, Linnaeus University, SE-39182 Kalmar, Sweden
| | - Ran Friedman
- Department of Chemistry and
Biomedical Sciences, Linnaeus University, SE-39182 Kalmar, Sweden
| |
Collapse
|
3
|
Bhati A, Wan S, Coveney PV. Equilibrium and Nonequilibrium Ensemble Methods for Accurate, Precise and Reproducible Absolute Binding Free Energy Calculations. J Chem Theory Comput 2025; 21:440-462. [PMID: 39680850 PMCID: PMC11736689 DOI: 10.1021/acs.jctc.4c01389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024]
Abstract
Free energy calculations for protein-ligand complexes have become widespread in recent years owing to several conceptual, methodological and technological advances. Central among these is the use of ensemble methods which permits accurate, precise and reproducible predictions and is necessary for uncertainty quantification. Absolute binding free energies (ABFEs) are challenging to predict using alchemical methods and their routine application in drug discovery has remained out of reach until now. Here, we apply ensemble alchemical ABFE methods to a large data set comprising 219 ligand-protein complexes and obtain statistically robust results with high accuracy (<1 kcal/mol). We compare equilibrium and nonequilibrium methods for ABFE predictions at large scale and provide a systematic critical assessment of each method. The equilibrium method is more accurate, precise, faster, computationally more cost-effective and requires a much simpler protocol, making it preferable for large scale and blind applications. We find that the calculated free energy distributions are non-normal and discuss the consequences. We recommend a definitive protocol to perform ABFE calculations optimally. Using this protocol, it is possible to perform thousands of ABFE calculations within a few hours on modern exascale machines.
Collapse
Affiliation(s)
- Agastya
P. Bhati
- Centre
for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, United Kingdom
| | - Shunzhou Wan
- Centre
for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, United Kingdom
| | - Peter V. Coveney
- Centre
for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, United Kingdom
- Computational
Science Laboratory, Institute for Informatics, Faculty of Science, University of Amsterdam, Amsterdam 1012, The Netherlands
- Advanced
Research Computing Centre, University College
London, London WC1H 9BT, United Kingdom
| |
Collapse
|
4
|
Zhang BW, Fajer M, Chen W, Moraca F, Wang L. Leveraging the Thermodynamics of Protein Conformations in Drug Discovery. J Chem Inf Model 2025; 65:252-264. [PMID: 39681511 DOI: 10.1021/acs.jcim.4c01612] [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: 12/18/2024]
Abstract
As the name implies, structure-based drug design requires confidence in the holo complex structure. The ability to clarify which protein conformation to use when ambiguity arises would be incredibly useful. We present a large scale validation of the computational method Protein Reorganization Free Energy Perturbation (PReorg-FEP) and demonstrate its quantitative accuracy in selecting the correct protein conformation among candidate models in apo or ligand induced states for 14 different systems. These candidate conformations are pulled from various drug discovery related campaigns: cryptic conformations induced by novel hits in lead identification, binding site rearrangement during lead optimization, and conflicting structural biology models. We also show an example of a pH-dependent conformational change, relevant to protein design.
Collapse
Affiliation(s)
- Bin W Zhang
- Schrödinger Inc., 1540 Broadway, 24th Floor, New York, New York 10036-4041, United States
| | - Mikolai Fajer
- Schrödinger Inc., 1540 Broadway, 24th Floor, New York, New York 10036-4041, United States
| | - Wei Chen
- Schrödinger Inc., 1540 Broadway, 24th Floor, New York, New York 10036-4041, United States
| | - Francesca Moraca
- Schrödinger Inc., 1540 Broadway, 24th Floor, New York, New York 10036-4041, United States
| | - Lingle Wang
- Schrödinger Inc., 1540 Broadway, 24th Floor, New York, New York 10036-4041, United States
| |
Collapse
|
5
|
Huang W, Liu R, Yao Y, Lai Y, Luo HB, Li Z. RED-E-Function-Based Equilibrium Parameter Finder: Finding the Best Restraint Parameters in Absolute Binding Free Energy Calculations. J Phys Chem Lett 2025; 16:253-260. [PMID: 39718976 DOI: 10.1021/acs.jpclett.4c02656] [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: 12/26/2024]
Abstract
Free energy perturbation (FEP)-based absolute binding free energy (ABFE) calculations have emerged as a powerful tool for the accurate prediction of ligand-protein binding affinities in drug discovery. The restraint addition is crucial in FEP-ABFE calculations; however, due to the non-orthogonal couplings between the restrained degrees of freedom, it typically requires numerous λ windows to ensure the phase-space overlap during restraint addition. This study introduces the RED-E-function-based equilibrium parameter finder (REPF), a novel method that relies on harmonic restraints to optimize the equilibrium values in restraints, enhancing phase-space overlap and improving the convergence of the restraint addition. REPF was applied to 44 protein-ligand complexes across 5 targets and compared to restraint schemes reported in the literature. We found that REPF-optimized restraints achieve an accuracy comparable to that of the 12λ approach while using only 2λ simulations, resulting in a significant reduction in computational costs. Extensive tests confirmed the improved convergence behavior and reduced energy fluctuations of REPF-optimized restraints.
Collapse
Affiliation(s)
- Wanyi Huang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Runduo Liu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yufen Yao
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yijun Lai
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hai-Bin Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- Song Li' Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Yazhou Bay, Sanya 572000, China
| | - Zhe Li
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
6
|
Baumann HM, Mobley DL. Impact of protein conformations on binding free energy calculations in the beta-secretase 1 system. J Comput Chem 2024; 45:2024-2033. [PMID: 38725239 PMCID: PMC11236511 DOI: 10.1002/jcc.27365] [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: 09/12/2023] [Revised: 01/13/2024] [Accepted: 03/24/2024] [Indexed: 07/11/2024]
Abstract
In binding free energy calculations, simulations must sample all relevant conformations of the system in order to obtain unbiased results. For instance, different ligands can bind to different metastable states of a protein, and if these protein conformational changes are not sampled in relative binding free energy calculations, the contribution of these states to binding is not accounted for and thus calculated binding free energies are inaccurate. In this work, we investigate the impact of different beta-sectretase 1 (BACE1) protein conformations obtained from x-ray crystallography on the binding of BACE1 inhibitors. We highlight how these conformational changes are not adequately sampled in typical molecular dynamics simulations. Furthermore, we show that insufficient sampling of relevant conformations induces substantial error in relative binding free energy calculations, as judged by a variation in calculated relative binding free energies up to 2 kcal/mol depending on the starting protein conformation. These results emphasize the importance of protein conformational sampling and pose this BACE1 system as a challenge case for further method development in the area of enhanced protein conformational sampling, either in combination with binding calculations or as an endpoint correction.
Collapse
Affiliation(s)
- Hannah M Baumann
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, California, USA
| | - David L Mobley
- Department of Chemistry, University of California, Irvine, California, USA
| |
Collapse
|
7
|
Hsu WT, Shirts MR. Replica Exchange of Expanded Ensembles: A Generalized Ensemble Approach with Enhanced Flexibility and Parallelizability. J Chem Theory Comput 2024; 20:6062-6081. [PMID: 39007702 PMCID: PMC11740319 DOI: 10.1021/acs.jctc.4c00484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Generalized ensemble methods such as Hamiltonian replica exchange (HREX) and expanded ensemble (EE) have been shown effective in free energy calculations for various contexts, given their ability to circumvent free energy barriers via nonphysical pathways defined by states with different modified Hamiltonians. However, both HREX and EE methods come with drawbacks, such as limited flexibility in parameter specification or the lack of parallelizability for more complicated applications. To address this challenge, we present the method of replica exchange of expanded ensembles (REXEE), which integrates the principles of HREX and EE methods by periodically exchanging coordinates of EE replicas sampling different yet overlapping sets of alchemical states. With the solvation free energy calculation of anthracene and binding free energy calculation of the CB7-10 binding complex, we show that the REXEE method achieves the same level of accuracy in free energy calculations as the HREX and EE methods, while offering enhanced flexibility and parallelizability. Additionally, we examined REXEE simulations with various setups to understand how different exchange frequencies and replica configurations influence the sampling efficiency in the fixed-weight phase and the weight convergence in the weight-updating phase. The REXEE approach can be further extended to support asynchronous parallelization schemes, allowing looser communications between larger numbers of loosely coupled processors such as cloud computing and therefore promising much more scalable and adaptive executions of alchemical free energy calculations. All algorithms for the REXEE method are available in the Python package ensemble_md, which offers an interface for REXEE simulation management without modifying the source code in GROMACS.
Collapse
Affiliation(s)
- Wei-Tse Hsu
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80305, United States
| | - Michael R Shirts
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80305, United States
| |
Collapse
|
8
|
Hahn DF, Gapsys V, de Groot BL, Mobley DL, Tresadern G. Current State of Open Source Force Fields in Protein-Ligand Binding Affinity Predictions. J Chem Inf Model 2024; 64:5063-5076. [PMID: 38895959 PMCID: PMC11234369 DOI: 10.1021/acs.jcim.4c00417] [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: 03/10/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 06/21/2024]
Abstract
In drug discovery, the in silico prediction of binding affinity is one of the major means to prioritize compounds for synthesis. Alchemical relative binding free energy (RBFE) calculations based on molecular dynamics (MD) simulations are nowadays a popular approach for the accurate affinity ranking of compounds. MD simulations rely on empirical force field parameters, which strongly influence the accuracy of the predicted affinities. Here, we evaluate the ability of six different small-molecule force fields to predict experimental protein-ligand binding affinities in RBFE calculations on a set of 598 ligands and 22 protein targets. The public force fields OpenFF Parsley and Sage, GAFF, and CGenFF show comparable accuracy, while OPLS3e is significantly more accurate. However, a consensus approach using Sage, GAFF, and CGenFF leads to accuracy comparable to OPLS3e. While Parsley and Sage are performing comparably based on aggregated statistics across the whole dataset, there are differences in terms of outliers. Analysis of the force field reveals that improved parameters lead to significant improvement in the accuracy of affinity predictions on subsets of the dataset involving those parameters. Lower accuracy can not only be attributed to the force field parameters but is also dependent on input preparation and sampling convergence of the calculations. Especially large perturbations and nonconverged simulations lead to less accurate predictions. The input structures, Gromacs force field files, as well as the analysis Python notebooks are available on GitHub.
Collapse
Affiliation(s)
- David F. Hahn
- Computational
Chemistry, Janssen Research & Development, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Vytautas Gapsys
- Computational
Chemistry, Janssen Research & Development, Turnhoutseweg 30, Beerse 2340, Belgium
- Computational
Biomolecular Dynamics Group, Max Planck
Institute for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
| | - Bert L. de Groot
- Computational
Biomolecular Dynamics Group, Max Planck
Institute for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
| | - David L. Mobley
- Department
of Chemistry, University of California, Irvine, California 92697, United States
- Department
of Pharmaceutical Sciences, University of
California, Irvine, California 92697, United States
| | - Gary Tresadern
- Computational
Chemistry, Janssen Research & Development, Turnhoutseweg 30, Beerse 2340, Belgium
| |
Collapse
|
9
|
Zeledon EV, Baxt LA, Khan TA, Michino M, Miller M, Huggins DJ, Jiang CS, Vosshall LB, Duvall LB. Next-generation neuropeptide Y receptor small-molecule agonists inhibit mosquito-biting behavior. Parasit Vectors 2024; 17:276. [PMID: 38937807 PMCID: PMC11212260 DOI: 10.1186/s13071-024-06347-w] [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: 03/25/2024] [Accepted: 06/09/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Female Aedes aegypti mosquitoes can spread disease-causing pathogens when they bite humans to obtain blood nutrients required for egg production. Following a complete blood meal, host-seeking is suppressed until eggs are laid. Neuropeptide Y-like receptor 7 (NPYLR7) plays a role in endogenous host-seeking suppression and previous work identified small-molecule NPYLR7 agonists that inhibit host-seeking and blood-feeding when fed to mosquitoes at high micromolar doses. METHODS Using structure-activity relationship analysis and structure-guided design we synthesized 128 compounds with similarity to known NPYLR7 agonists. RESULTS Although in vitro potency (EC50) was not strictly predictive of in vivo effect, we identified three compounds that reduced blood-feeding from a live host when fed to mosquitoes at a dose of 1 μM-a 100-fold improvement over the original reference compound. CONCLUSIONS Exogenous activation of NPYLR7 represents an innovative vector control strategy to block mosquito biting behavior and prevent mosquito-human host interactions that lead to pathogen transmission.
Collapse
Affiliation(s)
- Emely V Zeledon
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY, 10065, USA
- Howard Hughes Medical Institute, New York, NY, 10065, USA
| | - Leigh A Baxt
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, NY, 10065, USA
| | - Tanweer A Khan
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, NY, 10065, USA
- Atai Life Sciences, New York, NY, 10012, USA
| | - Mayako Michino
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, NY, 10065, USA
| | - Michael Miller
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, NY, 10065, USA
| | - David J Huggins
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, NY, 10065, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Caroline S Jiang
- Center for Clinical and Translational Science, The Rockefeller University, New York, NY, 10065, USA
| | - Leslie B Vosshall
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY, 10065, USA
- Howard Hughes Medical Institute, New York, NY, 10065, USA
- Kavli Neural Systems Institute, New York, NY, 10065, USA
| | - Laura B Duvall
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
10
|
Zhang X, Gao H, Wang H, Chen Z, Zhang Z, Chen X, Li Y, Qi Y, Wang R. PLANET: A Multi-objective Graph Neural Network Model for Protein-Ligand Binding Affinity Prediction. J Chem Inf Model 2024; 64:2205-2220. [PMID: 37319418 DOI: 10.1021/acs.jcim.3c00253] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Predicting protein-ligand binding affinity is a central issue in drug design. Various deep learning models have been published in recent years, where many of them rely on 3D protein-ligand complex structures as input and tend to focus on the single task of reproducing binding affinity. In this study, we have developed a graph neural network model called PLANET (Protein-Ligand Affinity prediction NETwork). This model takes the graph-represented 3D structure of the binding pocket on the target protein and the 2D chemical structure of the ligand molecule as input. It was trained through a multi-objective process with three related tasks, including deriving the protein-ligand binding affinity, protein-ligand contact map, and ligand distance matrix. Besides the protein-ligand complexes with known binding affinity data retrieved from the PDBbind database, a large number of non-binder decoys were also added to the training data for deriving the final model of PLANET. When tested on the CASF-2016 benchmark, PLANET exhibited a scoring power comparable to the best result yielded by other deep learning models as well as a reasonable ranking power and docking power. In virtual screening trials conducted on the DUD-E benchmark, PLANET's performance was notably better than several deep learning and machine learning models. As on the LIT-PCBA benchmark, PLANET achieved comparable accuracy as the conventional docking program Glide, but it only spent less than 1% of Glide's computation time to finish the same job because PLANET did not need exhaustive conformational sampling. Considering the decent accuracy and efficiency of PLANET in binding affinity prediction, it may become a useful tool for conducting large-scale virtual screening.
Collapse
Affiliation(s)
- Xiangying Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Haotian Gao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Haojie Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Zhihang Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Zhe Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Xinchong Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Yan Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Yifei Qi
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Renxiao Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| |
Collapse
|
11
|
Awoonor-Williams E, Abu-Saleh AAAA. Molecular Insights into the Impact of Mutations on the Binding Affinity of Targeted Covalent Inhibitors of BTK. J Phys Chem B 2024; 128:2874-2884. [PMID: 38502552 DOI: 10.1021/acs.jpcb.4c00310] [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: 03/21/2024]
Abstract
Targeted covalent inhibitors (TCIs) have witnessed a significant resurgence in recent years, particularly in the kinase drug discovery field for treating diverse clinical indications. The inhibition of Bruton's tyrosine kinase (BTK) for treating B-cell cancers is a classic example where TCIs such as ibrutinib have had breakthroughs in targeted therapy. However, selectivity remains challenging, and the emergence of resistance mutations is a critical concern for clinical efficacy. Computational methods that can accurately predict the impact of mutations on inhibitor binding affinity could prove helpful in informing targeted approaches─providing insights into drug resistance mechanisms. In addition, such systems could help guide the systematic evaluation and impact of mutations in disease models for optimal experimental design. Here, we have employed in silico physics-based methods to understand the effects of mutations on the binding affinity and conformational dynamics of select TCIs of BTK. The TCIs studied include ibrutinib, acalabrutinib, and zanubrutinib─all of which are FDA-approved drugs for treating multiple forms of leukemia and lymphoma. Our results offer useful molecular insights into the structural determinants, thermodynamics, and conformational energies that impact ligand binding for this biological target of clinical relevance.
Collapse
Affiliation(s)
- Ernest Awoonor-Williams
- Department of Chemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X7, Canada
| | - Abd Al-Aziz A Abu-Saleh
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9B 3P4, Canada
| |
Collapse
|
12
|
Zeledon EV, Baxt LA, Khan TA, Michino M, Miller M, Huggins DJ, Jiang CS, Vosshall LB, Duvall LB. Next Generation Neuropeptide Y Receptor Small Molecule Agonists Inhibit Mosquito Biting Behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582529. [PMID: 38464241 PMCID: PMC10925335 DOI: 10.1101/2024.02.28.582529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Female Aedes aegypti mosquitoes can spread disease-causing pathogens when they bite humans to obtain blood nutrients required for egg production. Following a complete blood meal, host-seeking is suppressed until eggs are laid. Neuropeptide Y-like Receptor 7 (NPYLR7) plays a role in endogenous host-seeking suppression and previous work identified small molecule NPYLR7 agonists that suppress host-seeking and blood feeding when fed to mosquitoes at high micromolar doses. Using structure activity relationship analysis and structure-guided design we synthesized 128 compounds with similarity to known NPYLR7 agonists. Although in vitro potency (EC50) was not strictly predictive of in vivo effect, we identified 3 compounds that suppressed blood feeding from a live host when fed to mosquitoes at a 1 μM dose, a 100-fold improvement over the original reference compound. Exogenous activation of NPYLR7 represents an innovative vector control strategy to block mosquito biting behavior and prevent mosquito/human host interactions that lead to pathogen transmission.
Collapse
Affiliation(s)
- Emely V. Zeledon
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York NY 10065, USA
- Howard Hughes Medical Institute, New York NY 10065, USA
| | - Leigh A. Baxt
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, New York 10065, USA
| | - Tanweer A. Khan
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, New York 10065, USA
| | - Mayako Michino
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, New York 10065, USA
| | - Michael Miller
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, New York 10065, USA
| | - David J. Huggins
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, New York 10065, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York NY 10065, USA
| | - Caroline S. Jiang
- Center for Clinical and Translational Science, The Rockefeller University, New York, NY 10065, USA
| | - Leslie B. Vosshall
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York NY 10065, USA
- Howard Hughes Medical Institute, New York NY 10065, USA
- Kavli Neural Systems Institute, New York, NY 10065, USA
| | - Laura B. Duvall
- Department of Biological Sciences, Columbia University, New York NY 10027, USA
| |
Collapse
|
13
|
Karrenbrock M, Rizzi V, Procacci P, Gervasio FL. Addressing Suboptimal Poses in Nonequilibrium Alchemical Calculations. J Phys Chem B 2024; 128:1595-1605. [PMID: 38323915 DOI: 10.1021/acs.jpcb.3c06516] [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: 02/08/2024]
Abstract
Alchemical transformations can be used to quantitatively estimate absolute binding free energies at a reasonable computational cost. However, most of the approaches currently in use require knowledge of the correct (crystallographic) pose. In this paper, we present a combined Hamiltonian replica exchange nonequilibrium alchemical method that allows us to reliably calculate absolute binding free energies, even when starting from suboptimal initial binding poses. Performing a preliminary Hamiltonian replica exchange enhances the sampling of slow degrees of freedom of the ligand and the target, allowing the system to populate the correct binding pose when starting from an approximate docking pose. We apply the method on 6 ligands of the first bromodomain of the BRD4 bromodomain-containing protein. For each ligand, we start nonequilibrium alchemical transformations from both the crystallographic pose and the top-scoring docked pose that are often significantly different. We show that the method produces statistically equivalent binding free energies, making it a useful tool for computational drug discovery pipelines.
Collapse
Affiliation(s)
- Maurice Karrenbrock
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, CH-1206 Geneva, Switzerland
| | - Valerio Rizzi
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, CH-1206 Geneva, Switzerland
| | - Piero Procacci
- Chemistry Department, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Francesco Luigi Gervasio
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, CH-1206 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, Switzerland
- Chemistry Department, University College London (UCL), WC1E 6BT London, U.K
- Swiss Bioinformatics Institute, University of Geneva, CH-1206 Geneva, Switzerland
| |
Collapse
|
14
|
Gartan P, Khorsand F, Mizar P, Vahokovski JI, Cervantes LF, Haug BE, Brenk R, Brooks CL, Reuter N. Investigating Polypharmacology through Targeting Known Human Neutrophil Elastase Inhibitors to Proteinase 3. J Chem Inf Model 2024; 64:621-626. [PMID: 38276895 PMCID: PMC10865350 DOI: 10.1021/acs.jcim.3c01949] [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: 12/07/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024]
Abstract
Using a combination of multisite λ-dynamics (MSλD) together with in vitro IC50 assays, we evaluated the polypharmacological potential of a scaffold currently in clinical trials for inhibition of human neutrophil elastase (HNE), targeting cardiopulmonary disease, for efficacious inhibition of Proteinase 3 (PR3), a related neutrophil serine proteinase. The affinities we observe suggest that the dihydropyrimidinone scaffold can serve as a suitable starting point for the establishment of polypharmacologically targeting both enzymes and enhancing the potential for treatments addressing diseases like chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Parveen Gartan
- Department
of Chemistry, University of Bergen, Bergen 5020, Norway
- Computational
Biology Unit, University of Bergen, Bergen 5020, Norway
| | - Fahimeh Khorsand
- Department
of Biomedicine, University of Bergen, Bergen 5020, Norway
| | - Pushpak Mizar
- Department
of Chemistry, University of Bergen, Bergen 5020, Norway
| | - Juha Ilmari Vahokovski
- Core
Facility for Biophysics, Structural Biology, and Screening, Department
of Biomedicine, University of Bergen, Bergen 5020, Norway
| | - Luis F. Cervantes
- Department
of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bengt Erik Haug
- Department
of Chemistry, University of Bergen, Bergen 5020, Norway
- Centre for
Pharmacy, University of Bergen, Bergen 5020, Norway
| | - Ruth Brenk
- Department
of Biomedicine, University of Bergen, Bergen 5020, Norway
| | - Charles L. Brooks
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Biophysics
Program, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Nathalie Reuter
- Department
of Chemistry, University of Bergen, Bergen 5020, Norway
- Computational
Biology Unit, University of Bergen, Bergen 5020, Norway
| |
Collapse
|
15
|
Wang S, Liu F, Li P, Wang JN, Mo Y, Lin B, Mei Y. Potent inhibitors targeting cyclin-dependent kinase 9 discovered via virtual high-throughput screening and absolute binding free energy calculations. Phys Chem Chem Phys 2024; 26:5377-5386. [PMID: 38269624 DOI: 10.1039/d3cp05582e] [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: 01/26/2024]
Abstract
Due to the crucial regulatory mechanism of cyclin-dependent kinase 9 (CDK9) in mRNA transcription, the development of kinase inhibitors targeting CDK9 holds promise as a potential treatment strategy for cancer. A structure-based virtual screening approach has been employed for the discovery of potential novel CDK9 inhibitors. First, compounds with kinase inhibitor characteristics were identified from the ZINC15 database via virtual high-throughput screening. Next, the predicted binding modes were optimized by molecular dynamics simulations, followed by precise estimation of binding affinities using absolute binding free energy calculations based on the free energy perturbation scheme. The binding mode of molecule 006 underwent an inward-to-outward flipping, and the new binding mode exhibited binding affinity comparable to the small molecule T6Q in the crystal structure (PDB ID: 4BCF), highlighting the essential role of molecular dynamics simulation in capturing a plausible binding pose bridging docking and absolute binding free energy calculations. Finally, structural modifications based on these findings further enhanced the binding affinity with CDK9. The results revealed that enhancing the molecule's rigidity through ring formation, while maintaining the major interactions, reduced the entropy loss during the binding process and, thus, enhanced binding affinities.
Collapse
Affiliation(s)
- Shipeng Wang
- School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Fengjiao Liu
- School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Pengfei Li
- Single Particle, LLC, 10531 4S Commons Dr 166-629, San Diego, CA 92127, USA
| | - Jia-Ning Wang
- State Key Laboratory of Precision Spectroscopy, School of Physics and Electronic Science, East China Normal University, Shanghai 200241, China
| | - Yan Mo
- State Key Laboratory of Precision Spectroscopy, School of Physics and Electronic Science, East China Normal University, Shanghai 200241, China
- NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, China
- Collaborative Innovation Center of Extreme Optics, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Bin Lin
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Ye Mei
- State Key Laboratory of Precision Spectroscopy, School of Physics and Electronic Science, East China Normal University, Shanghai 200241, China
- NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, China
- Collaborative Innovation Center of Extreme Optics, Shanxi University, Taiyuan, Shanxi 030006, China
| |
Collapse
|
16
|
Yang K, Liu H. Uncovering New Conformational States of the Substrate Binding Pocket of LSD1 Potential for Inhibitor Design via Funnel Metadynamics. J Phys Chem B 2024; 128:137-149. [PMID: 38151469 DOI: 10.1021/acs.jpcb.3c06900] [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: 12/29/2023]
Abstract
Lysine-specific demethylase 1 (LSD1) is a promising therapeutic target for cancer therapy. So far, over 80 crystal structures of LSD1 in different complex states have been deposited in the Protein Data Bank, which are valuable resources for performing structure-based drug design. However, among all of the crystal structures of LSD1, the substrate binding pocket, which is the most efficient druggable site for designing LSD1 inhibitors at present, is very similar no matter whether LSD1 is in the apo or any holo forms, which is inconsistent with its versatile demethylase functions. To investigate whether the substrate binding pocket is rigid or exhibits other representative conformations different from the crystal conformations that are feasible for designing new LSD1 inhibitors, we performed funnel metadynamics simulations to study the conformation dynamics of LSD1 in the binding process of two effective LSD1 inhibitors (CC-90011 and 6X0, CC-90011 undergoing clinical trials). Our results showed that the entrance of the substrate binding pocket is very flexible. Two representative entrance conformations of LSD1 counting against binding with the substrate of histone H3 were detected, which may be used for structure-based LSD1 inhibitor design. Besides, alternative optimal binding modes and prebinding modes for both inhibitors were also detected, which depicted that the key interactions changed along with the binding process. Our results should provide great help for LSD1 inhibitor design.
Collapse
Affiliation(s)
- Kecheng Yang
- National Supercomputing Center in Zhengzhou, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Hongmin Liu
- Key Lab of Advanced Drug Preparation Technologies, Ministry of Education of China, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| |
Collapse
|
17
|
Liu R, Li W, Yao Y, Wu Y, Luo HB, Li Z. Accelerating and Automating the Free Energy Perturbation Absolute Binding Free Energy Calculation with the RED-E Function. J Chem Inf Model 2023; 63:7755-7767. [PMID: 38048439 DOI: 10.1021/acs.jcim.3c01670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
The accurate prediction of the binding affinities between small molecules and biological macromolecules plays a fundamental role in structure-based drug design, which is still challenging. The free energy perturbation-based absolute binding free energy (FEP-ABFE) approach has shown potential in its reliability. To correctly calculate the energy related to the ligand being restrained by the receptor, additional restraints between the ligand and the receptor are needed. However, determining the restraint parameters for individual ligands empirically is too trivial to be automated, and usually gives rise to numerical instabilities, which set back the applications of FEP-ABFE. To address these issues, we derived the analytical expression for the probability distribution of energy differences, P(ΔU), during the process of restraint addition, which is called the RED-E (restraint energy distribution at equilibrium position) function. Simulations indicated that the RED-E function can accurately describe P(ΔU) when restraints are added at the equilibrium position. Based on the RED-E function, an automatic restraint selection method was proposed to select the best restraint. With this method, there is a high phase-space overlap between the free and restrained states, such that using a 2-λ perturbation can accurately calculate the free energy of the restraint addition, which is a nearly 6 times acceleration compared with current widely used 12-λ perturbation method. The RED-E function gives insight into the non-Gaussian behavior of the sampled P(ΔU) in certain FEP processes in an analytical way. The highly automated and accelerated restraint selection also makes it possible for the large-scale application of FEP-ABFE in real drug discovery practices.
Collapse
Affiliation(s)
- Runduo Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wenchao Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yufen Yao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yinuo Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hai-Bin Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, Hainan 570228, China
- Song Li' Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Yazhou Bay, Sanya 572000, China
| | - Zhe Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
18
|
Michino M, Beautrait A, Boyles NA, Nadupalli A, Dementiev A, Sun S, Ginn J, Baxt L, Suto R, Bryk R, Jerome SV, Huggins DJ, Vendome J. Shape-Based Virtual Screening of a Billion-Compound Library Identifies Mycobacterial Lipoamide Dehydrogenase Inhibitors. ACS BIO & MED CHEM AU 2023; 3:507-515. [PMID: 38144256 PMCID: PMC10739260 DOI: 10.1021/acsbiomedchemau.3c00046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 12/26/2023]
Abstract
Lpd (lipoamide dehydrogenase) in Mycobacterium tuberculosis (Mtb) is required for virulence and is a genetically validated tuberculosis (TB) target. Numerous screens have been performed over the last decade, yet only two inhibitor series have been identified. Recent advances in large-scale virtual screening methods combined with make-on-demand compound libraries have shown the potential for finding novel hits. In this study, the Enamine REAL library consisting of ∼1.12 billion compounds was efficiently screened using the GPU Shape screen method against Mtb Lpd to find additional chemical matter that would expand on the known sulfonamide inhibitor series. We identified six new inhibitors with IC50 in the range of 5-100 μM. While these compounds remained chemically close to the already known sulfonamide series inhibitors, some diversity was found in the cores of the hits. The two most potent hits were further validated by one-step potency optimization to submicromolar levels. The co-crystal structure of optimized analogue TDI-13537 provided new insights into the potency determinants of the series.
Collapse
Affiliation(s)
- Mayako Michino
- Sanders
Tri-Institutional Therapeutics Discovery Institute, 1230 York Avenue, Box 122, New York, New York 10065, United States
| | - Alexandre Beautrait
- Schrödinger,
Inc., 1540 Broadway, 24th Floor, New York, New York 10036, United States
| | - Nicholas A. Boyles
- Schrödinger,
Inc., 1540 Broadway, 24th Floor, New York, New York 10036, United States
| | - Aparna Nadupalli
- Schrödinger,
Inc., 12 Michigan Dr., Natick, Massachusetts 01760, United States
| | - Alexey Dementiev
- Schrödinger,
Inc., 12 Michigan Dr., Natick, Massachusetts 01760, United States
| | - Shan Sun
- Sanders
Tri-Institutional Therapeutics Discovery Institute, 1230 York Avenue, Box 122, New York, New York 10065, United States
| | - John Ginn
- Sanders
Tri-Institutional Therapeutics Discovery Institute, 1230 York Avenue, Box 122, New York, New York 10065, United States
| | - Leigh Baxt
- Sanders
Tri-Institutional Therapeutics Discovery Institute, 1230 York Avenue, Box 122, New York, New York 10065, United States
| | - Robert Suto
- Schrödinger,
Inc., 12 Michigan Dr., Natick, Massachusetts 01760, United States
| | - Ruslana Bryk
- Department
of Microbiology and Immunology, Weill Cornell
Medicine, New York, New York 10065, United States
| | - Steven V. Jerome
- Schrödinger,
Inc., 1540 Broadway, 24th Floor, New York, New York 10036, United States
| | - David J. Huggins
- Sanders
Tri-Institutional Therapeutics Discovery Institute, 1230 York Avenue, Box 122, New York, New York 10065, United States
- Department
of Physiology and Biophysics, Weill Cornell
Medicine, New York, New York 10021, United States
| | - Jeremie Vendome
- Schrödinger,
Inc., 1540 Broadway, 24th Floor, New York, New York 10036, United States
| |
Collapse
|
19
|
Fu H, Chipot C, Shao X, Cai W. Standard Binding Free-Energy Calculations: How Far Are We from Automation? J Phys Chem B 2023; 127:10459-10468. [PMID: 37824848 DOI: 10.1021/acs.jpcb.3c04370] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Recent success stories suggest that in silico protein-ligand binding free-energy calculations are approaching chemical accuracy. However, their widespread application remains limited by the extensive human intervention required, posing challenges for the neophyte. As such, it is critical to develop automated workflows for estimating protein-ligand binding affinities with minimum personal involvement. Key human efforts include setting up and tuning enhanced-sampling or alchemical-transformation algorithms as a preamble to computational binding free-energy estimations. Additionally, preparing input files, bookkeeping, and postprocessing represent nontrivial tasks. In this Perspective, we discuss recent progress in automating standard binding free-energy calculations, featuring the development of adaptive or parameter-free algorithms, standardization of binding free-energy calculation workflows, and the implementation of user-friendly software. We also assess the current state of automated standard binding free-energy calculations and evaluate the limitations of existing methods. Last, we outline the requirements for future algorithms and workflows to facilitate automated free-energy calculations for diverse protein-ligand complexes.
Collapse
Affiliation(s)
- Haohao Fu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Christophe Chipot
- Laboratoire International Associé CNRS and University of Illinois at Urbana-Champaign, UMR no. 7019, Université de Lorraine, BP 70239, F-54506 Vandoeuvre-lès-Nancy, France
- Department of Physics, University of Illinois at Urbana-Champaign, 1110 West Green Street, Urbana, Illinois 61801, United States
- Department of Chemistry, The University of Chicago, 5735 South Ellis Avenue, Chicago, Illinois 60637, United States
- Department of Chemistry, The University of Hawai'i at Ma̅noa, 2545 McCarthy Mall, Honolulu, Hawaii 96822, United States
| | - Xueguang Shao
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Wensheng Cai
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| |
Collapse
|
20
|
Wan S, Bhati AP, Coveney PV. Comparison of Equilibrium and Nonequilibrium Approaches for Relative Binding Free Energy Predictions. J Chem Theory Comput 2023; 19:7846-7860. [PMID: 37862058 PMCID: PMC10653111 DOI: 10.1021/acs.jctc.3c00842] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Indexed: 10/21/2023]
Abstract
Alchemical relative binding free energy calculations have recently found important applications in drug optimization. A series of congeneric compounds are generated from a preidentified lead compound, and their relative binding affinities to a protein are assessed in order to optimize candidate drugs. While methods based on equilibrium thermodynamics have been extensively studied, an approach based on nonequilibrium methods has recently been reported together with claims of its superiority. However, these claims pay insufficient attention to the basis and reliability of both methods. Here we report a comparative study of the two approaches across a large data set, comprising more than 500 ligand transformations spanning in excess of 300 ligands binding to a set of 14 diverse protein targets. Ensemble methods are essential to quantify the uncertainty in these calculations, not only for the reasons already established in the equilibrium approach but also to ensure that the nonequilibrium calculations reside within their domain of validity. If and only if ensemble methods are applied, we find that the nonequilibrium method can achieve accuracy and precision comparable to those of the equilibrium approach. Compared to the equilibrium method, the nonequilibrium approach can reduce computational costs but introduces higher computational complexity and longer wall clock times. There are, however, cases where the standard length of a nonequilibrium transition is not sufficient, necessitating a complete rerun of the entire set of transitions. This significantly increases the computational cost and proves to be highly inconvenient during large-scale applications. Our findings provide a key set of recommendations that should be adopted for the reliable implementation of nonequilibrium approaches to relative binding free energy calculations in ligand-protein systems.
Collapse
Affiliation(s)
- Shunzhou Wan
- Centre
for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, U.K.
| | - Agastya P. Bhati
- Centre
for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, U.K.
| | - Peter V. Coveney
- Centre
for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, U.K.
- Advanced
Research Computing Centre, University College
London, London WC1H 0AJ, U.K.
- Computational
Science Laboratory, Institute for Informatics, Faculty of Science, University of Amsterdam, Amsterdam 1012 WP, Netherlands
| |
Collapse
|
21
|
Papadourakis M, Sinenka H, Matricon P, Hénin J, Brannigan G, Pérez-Benito L, Pande V, van Vlijmen H, de Graaf C, Deflorian F, Tresadern G, Cecchini M, Cournia Z. Alchemical Free Energy Calculations on Membrane-Associated Proteins. J Chem Theory Comput 2023; 19:7437-7458. [PMID: 37902715 PMCID: PMC11017255 DOI: 10.1021/acs.jctc.3c00365] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Indexed: 10/31/2023]
Abstract
Membrane proteins have diverse functions within cells and are well-established drug targets. The advances in membrane protein structural biology have revealed drug and lipid binding sites on membrane proteins, while computational methods such as molecular simulations can resolve the thermodynamic basis of these interactions. Particularly, alchemical free energy calculations have shown promise in the calculation of reliable and reproducible binding free energies of protein-ligand and protein-lipid complexes in membrane-associated systems. In this review, we present an overview of representative alchemical free energy studies on G-protein-coupled receptors, ion channels, transporters as well as protein-lipid interactions, with emphasis on best practices and critical aspects of running these simulations. Additionally, we analyze challenges and successes when running alchemical free energy calculations on membrane-associated proteins. Finally, we highlight the value of alchemical free energy calculations calculations in drug discovery and their applicability in the pharmaceutical industry.
Collapse
Affiliation(s)
- Michail Papadourakis
- Biomedical
Research Foundation, Academy of Athens, 4 Soranou Ephessiou, 11527 Athens, Greece
| | - Hryhory Sinenka
- Institut
de Chimie de Strasbourg, UMR7177, CNRS, Université de Strasbourg, F-67083 Strasbourg Cedex, France
| | - Pierre Matricon
- Sosei
Heptares, Steinmetz Building,
Granta Park, Great Abington, Cambridge CB21 6DG, United
Kingdom
| | - Jérôme Hénin
- Laboratoire
de Biochimie Théorique UPR 9080, CNRS and Université Paris Cité, 75005 Paris, France
| | - Grace Brannigan
- Center
for Computational and Integrative Biology, Rutgers University−Camden, Camden, New Jersey 08103, United States of America
- Department
of Physics, Rutgers University−Camden, Camden, New Jersey 08102, United States
of America
| | - Laura Pérez-Benito
- CADD,
In Silico Discovery, Janssen Research &
Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Vineet Pande
- CADD,
In Silico Discovery, Janssen Research &
Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Herman van Vlijmen
- CADD,
In Silico Discovery, Janssen Research &
Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Chris de Graaf
- Sosei
Heptares, Steinmetz Building,
Granta Park, Great Abington, Cambridge CB21 6DG, United
Kingdom
| | - Francesca Deflorian
- Sosei
Heptares, Steinmetz Building,
Granta Park, Great Abington, Cambridge CB21 6DG, United
Kingdom
| | - Gary Tresadern
- CADD,
In Silico Discovery, Janssen Research &
Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Marco Cecchini
- Institut
de Chimie de Strasbourg, UMR7177, CNRS, Université de Strasbourg, F-67083 Strasbourg Cedex, France
| | - Zoe Cournia
- Biomedical
Research Foundation, Academy of Athens, 4 Soranou Ephessiou, 11527 Athens, Greece
| |
Collapse
|
22
|
Anichina K, Mavrova A, Vuchev D, Popova-Daskalova G, Bassi G, Rossi A, Montesi M, Panseri S, Fratev F, Naydenova E. Benzimidazoles Containing Piperazine Skeleton at C-2 Position as Promising Tubulin Modulators with Anthelmintic and Antineoplastic Activity. Pharmaceuticals (Basel) 2023; 16:1518. [PMID: 38004384 PMCID: PMC10675210 DOI: 10.3390/ph16111518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023] Open
Abstract
Benzimidazole anthelmintic drugs hold promise for repurposing as cancer treatments due to their interference with tubulin polymerization and depolymerization, manifesting anticancer properties. We explored the potential of benzimidazole compounds with a piperazine fragment at C-2 as tubulin-targeting agents. In particular, we assessed their anthelmintic activity against isolated Trichinella spiralis muscle larvae and their effects on glioblastoma (U-87 MG) and breast cancer (MDA-MB-231) cell lines. Compound 7c demonstrated exceptional anthelmintic efficacy, achieving a 92.7% reduction in parasite activity at 100 μg/mL after 48 hours. In vitro cytotoxicity analysis of MDA-MB 231 and U87 MG cell lines showed that derivatives 7b, 7d, and 7c displayed lower IC50 values compared to albendazole (ABZ), the control. These piperazine benzimidazoles effectively reduced cell migration in both cell lines, with compound 7c exhibiting the most significant reduction, making it a promising candidate for further study. The binding mode of the most promising compound 7c, was determined using the induced fit docking-molecular dynamics (IFD-MD) approach. Regular docking and IFD were also employed for comparison. The IFD-MD analysis revealed that 7c binds to tubulin in a unique binding cavity near that of ABZ, but the benzimidazole ring was fitted much deeper into the binding pocket. Finally, the absolute free energy of perturbation technique was applied to evaluate the 7c binding affinity, further confirming the observed binding mode.
Collapse
Affiliation(s)
- Kameliya Anichina
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria;
| | - Anelia Mavrova
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria;
| | - Dimitar Vuchev
- Department of Infectious Diseases, Parasitology and Tropical Medicine, Medical University, 15A Vasil Aprilov Blvd., 4002 Plovdiv, Bulgaria; (D.V.); (G.P.-D.)
| | - Galya Popova-Daskalova
- Department of Infectious Diseases, Parasitology and Tropical Medicine, Medical University, 15A Vasil Aprilov Blvd., 4002 Plovdiv, Bulgaria; (D.V.); (G.P.-D.)
| | - Giada Bassi
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy; (G.B.); (A.R.); (M.M.); (S.P.)
- Department of Neurosciences, Imaging and Clinical Sciences, University of G. D’Annunzio, Via Luigi Polacchi, 11, 66100 Chieti, Italy
| | - Arianna Rossi
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy; (G.B.); (A.R.); (M.M.); (S.P.)
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98166 Messina, Italy
| | - Monica Montesi
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy; (G.B.); (A.R.); (M.M.); (S.P.)
| | - Silvia Panseri
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy; (G.B.); (A.R.); (M.M.); (S.P.)
| | - Filip Fratev
- Micar Innovation (Micar 21) Ltd., 34B Persenk Str., 1407 Sofia, Bulgaria;
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas at El Paso, 1101 N Campbell St., El Paso, TX 79968, USA
| | - Emilia Naydenova
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria;
| |
Collapse
|
23
|
Shiota K, Akutsu T. Multi-shelled ECIF: improved extended connectivity interaction features for accurate binding affinity prediction. BIOINFORMATICS ADVANCES 2023; 3:vbad155. [PMID: 37928345 PMCID: PMC10625475 DOI: 10.1093/bioadv/vbad155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/20/2023] [Accepted: 10/19/2023] [Indexed: 11/07/2023]
Abstract
Motivation Extended connectivity interaction features (ECIF) is a method developed to predict protein-ligand binding affinity, allowing for detailed atomic representation. It performed very well in terms of Comparative Assessment of Scoring Functions 2016 (CASF-2016) scoring power. However, ECIF has the limitation of not being able to adequately account for interatomic distances. Results To investigate what kind of distance representation is effective for P-L binding affinity prediction, we have developed two algorithms that improved ECIF's feature extraction method to take distance into account. One is multi-shelled ECIF, which takes into account the distance between atoms by dividing the distance between atoms into multiple layers. The other is weighted ECIF, which weights the importance of interactions according to the distance between atoms. A comparison of these two methods shows that multi-shelled ECIF outperforms weighted ECIF and the original ECIF, achieving a CASF-2016 scoring power Pearson correlation coefficient of 0.877. Availability and implementation All the codes and data are available on GitHub (https://github.com/koji11235/MSECIFv2).
Collapse
Affiliation(s)
- Koji Shiota
- Department of Intelligence Science and Technology, Graduate School of Informatics, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Tatsuya Akutsu
- Department of Intelligence Science and Technology, Graduate School of Informatics, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| |
Collapse
|
24
|
Ross GA, Lu C, Scarabelli G, Albanese SK, Houang E, Abel R, Harder ED, Wang L. The maximal and current accuracy of rigorous protein-ligand binding free energy calculations. Commun Chem 2023; 6:222. [PMID: 37838760 PMCID: PMC10576784 DOI: 10.1038/s42004-023-01019-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 10/02/2023] [Indexed: 10/16/2023] Open
Abstract
Computational techniques can speed up the identification of hits and accelerate the development of candidate molecules for drug discovery. Among techniques for predicting relative binding affinities, the most consistently accurate is free energy perturbation (FEP), a class of rigorous physics-based methods. However, uncertainty remains about how accurate FEP is and can ever be. Here, we present what we believe to be the largest publicly available dataset of proteins and congeneric series of small molecules, and assess the accuracy of the leading FEP workflow. To ascertain the limit of achievable accuracy, we also survey the reproducibility of experimental relative affinity measurements. We find a wide variability in experimental accuracy and a correspondence between binding and functional assays. When careful preparation of protein and ligand structures is undertaken, FEP can achieve accuracy comparable to experimental reproducibility. Throughout, we highlight reliable protocols that can help maximize the accuracy of FEP in prospective studies.
Collapse
Affiliation(s)
- Gregory A Ross
- Schrödinger Inc, New York, NY, USA.
- Isomorphic Labs, London, UK.
| | - Chao Lu
- Schrödinger Inc, New York, NY, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Khan H, Waqas M, Khurshid B, Ullah N, Khalid A, Abdalla AN, Alamri MA, Wadood A. Investigating the role of Sterol C24-Methyl transferase mutation on drug resistance in leishmaniasis and identifying potential inhibitors. J Biomol Struct Dyn 2023; 42:10374-10387. [PMID: 37723868 DOI: 10.1080/07391102.2023.2256879] [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: 12/19/2022] [Accepted: 09/02/2023] [Indexed: 09/20/2023]
Abstract
Leishmaniasis is a fatal disease caused by the leishmania parasite. For the survival of the leishmania parasite, Sterol C24-Methyl Transferase (SMT) is essential which is an enzyme of the ergosterol pathway. SMT protein mutation is responsible for Amphotericin-B drug resistance in Leishmania, which is the main treatment for visceral leishmaniasis. Amphotericin-B resistance is caused by three mutated residues V131I, V321I and F72C. The underlying mechanisms and structural changes in SMT enzymes responsible for resistance due to mutation are still not well understood. In the current study, the potential mechanism of resistance due to these mutations and the structure variation of wild and mutant SMT proteins were investigated through molecular dynamics simulations and molecular docking analysis. The results showed that AmB established strong bonding interaction with wild SMT as compare to mutants SMT. The binding energy calculation showed that binding energy of AmB with mutants SMT increases as compare to the wild SMT. Further structural based virtual screening was carried out to design potential inhibitors for the mutant SMT. On the basis of structural-based virtual screening four inhibitors (SANC01057, SANC00882, SANC00414, SANC01047) were computationally identified as potential mutant SMT (F72C) inhibitors. This work provides valuable information for improved management of drug resistant Leishmaniasis.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Huma Khan
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Muhammad Waqas
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al-Mouz Nizwa, Oman
| | - Beenish Khurshid
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Nazif Ullah
- Department of Biotechnology, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan, Saudi Arabia
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mubarak A Alamri
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| |
Collapse
|
26
|
Ghahremanpour MM, Saar A, Tirado-Rives J, Jorgensen WL. Computation of Absolute Binding Free Energies for Noncovalent Inhibitors with SARS-CoV-2 Main Protease. J Chem Inf Model 2023; 63:5309-5318. [PMID: 37561001 DOI: 10.1021/acs.jcim.3c00874] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Accurate, routine calculation of absolute binding free energies (ABFEs) for protein-ligand complexes remains a key goal of computer-aided drug design since it can enable screening and optimization of drug candidates. For development and testing of related methods, it is important to have high-quality datasets. To this end, from our own experimental studies, we have selected a set of 16 inhibitors of the SARS-CoV-2 main protease (Mpro) with structural diversity and well-distributed BFEs covering a 5 kcal/mol range. There is also minimal structural uncertainty since X-ray crystal structures have been deposited for 12 of the compounds. For methods testing, we report ABFE results from 2 μs molecular dynamics (MD) simulations using free energy perturbation (FEP) theory. The correlation of experimental and computed results is encouraging, with a Pearson's r2 of 0.58 and a Kendall τ of 0.24. The results indicate that current FEP-based ABFE calculations can be used for identification of active compounds (hits). While their accuracy for lead optimization is not yet sufficient, this activity remains addressable in separate lead series by relative BFE calculations.
Collapse
Affiliation(s)
| | - Anastasia Saar
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Julian Tirado-Rives
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - William L Jorgensen
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| |
Collapse
|
27
|
Baumann H, Dybeck E, McClendon CL, Pickard FC, Gapsys V, Pérez-Benito L, Hahn DF, Tresadern G, Mathiowetz AM, Mobley DL. Broadening the Scope of Binding Free Energy Calculations Using a Separated Topologies Approach. J Chem Theory Comput 2023; 19:5058-5076. [PMID: 37487138 PMCID: PMC10413862 DOI: 10.1021/acs.jctc.3c00282] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Indexed: 07/26/2023]
Abstract
Binding free energy calculations predict the potency of compounds to protein binding sites in a physically rigorous manner and see broad application in prioritizing the synthesis of novel drug candidates. Relative binding free energy (RBFE) calculations have emerged as an industry-standard approach to achieve highly accurate rank-order predictions of the potency of related compounds; however, this approach requires that the ligands share a common scaffold and a common binding mode, restricting the methods' domain of applicability. This is a critical limitation since complex modifications to the ligands, especially core hopping, are very common in drug design. Absolute binding free energy (ABFE) calculations are an alternate method that can be used for ligands that are not congeneric. However, ABFE suffers from a known problem of long convergence times due to the need to sample additional degrees of freedom within each system, such as sampling rearrangements necessary to open and close the binding site. Here, we report on an alternative method for RBFE, called Separated Topologies (SepTop), which overcomes the issues in both of the aforementioned methods by enabling large scaffold changes between ligands with a convergence time comparable to traditional RBFE. Instead of only mutating atoms that vary between two ligands, this approach performs two absolute free energy calculations at the same time in opposite directions, one for each ligand. Defining the two ligands independently allows the comparison of the binding of diverse ligands without the artificial constraints of identical poses or a suitable atom-atom mapping. This approach also avoids the need to sample the unbound state of the protein, making it more efficient than absolute binding free energy calculations. Here, we introduce an implementation of SepTop. We developed a general and efficient protocol for running SepTop, and we demonstrated the method on four diverse, pharmaceutically relevant systems. We report the performance of the method, as well as our practical insights into the strengths, weaknesses, and challenges of applying this method in an industrial drug design setting. We find that the accuracy of the approach is sufficiently high to rank order ligands with an accuracy comparable to traditional RBFE calculations while maintaining the additional flexibility of SepTop.
Collapse
Affiliation(s)
- Hannah
M. Baumann
- Department
of Pharmaceutical Sciences, University of
California, Irvine, Irvine, California 92697, United States
| | - Eric Dybeck
- Pfizer
Worldwide Research, Development, and Medical, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Christopher L. McClendon
- Pfizer
Worldwide Research, Development, and Medical, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Frank C. Pickard
- Pfizer
Worldwide Research, Development, and Medical, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Vytautas Gapsys
- Computational
Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Laura Pérez-Benito
- Computational
Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - David F. Hahn
- Computational
Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Gary Tresadern
- Computational
Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Alan M. Mathiowetz
- Pfizer
Worldwide Research, Development, and Medical, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - David L. Mobley
- Department
of Pharmaceutical Sciences, University of
California, Irvine, Irvine, California 92697, United States
- Department
of Chemistry, University of California,
Irvine, Irvine, California 92697, United States
| |
Collapse
|
28
|
Clark F, Robb G, Cole DJ, Michel J. Comparison of Receptor-Ligand Restraint Schemes for Alchemical Absolute Binding Free Energy Calculations. J Chem Theory Comput 2023; 19:3686-3704. [PMID: 37285579 PMCID: PMC10308817 DOI: 10.1021/acs.jctc.3c00139] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Indexed: 06/09/2023]
Abstract
Alchemical absolute binding free energy calculations are of increasing interest in drug discovery. These calculations require restraints between the receptor and ligand to restrict their relative positions and, optionally, orientations. Boresch restraints are commonly used, but they must be carefully selected in order to sufficiently restrain the ligand and to avoid inherent instabilities. Applying multiple distance restraints between anchor points in the receptor and ligand provides an alternative framework without inherent instabilities which may provide convergence benefits by more strongly restricting the relative movements of the receptor and ligand. However, there is no simple method to calculate the free energy of releasing these restraints due to the coupling of the internal and external degrees of freedom of the receptor and ligand. Here, a method to rigorously calculate free energies of binding with multiple distance restraints by imposing intramolecular restraints on the anchor points is proposed. Absolute binding free energies for the human macrophage migration inhibitory factor/MIF180, system obtained using a variety of Boresch restraints and rigorous and nonrigorous implementations of multiple distance restraints are compared. It is shown that several multiple distance restraint schemes produce estimates in good agreement with Boresch restraints. In contrast, calculations without orientational restraints produce erroneously favorable free energies of binding by up to approximately 4 kcal mol-1. These approaches offer new options for the deployment of alchemical absolute binding free energy calculations.
Collapse
Affiliation(s)
- Finlay Clark
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh EH9 3FJ, United Kingdom
| | - Graeme Robb
- Oncology
R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | - Daniel J. Cole
- School
of Natural and Environmental Sciences, Newcastle
University, Newcastle
upon Tyne NE1 7RU, United Kingdom
| | - Julien Michel
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh EH9 3FJ, United Kingdom
| |
Collapse
|
29
|
Akkus E, Tayfuroglu O, Yildiz M, Kocak A. Revisiting MMPBSA by Adoption of MC-Based Surface Area/Volume, ANI-ML Potentials, and Two-Valued Interior Dielectric Constant. J Phys Chem B 2023; 127:4415-4429. [PMID: 37171911 DOI: 10.1021/acs.jpcb.3c00834] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Here, we report the accuracy improvements of molecular mechanics Poisson-Boltzmann surface area (MMPBSA) calculations by adoption of ANI-ML potentials in replacement of MM terms, the use of solvent-accessible surface area (SASA) and volume (SAV) values from the Monte Carlo sampling of the probe, and introducing two different interior dielectric constants for electrostatic interactions of protein-ligand (P-L) and polar solvation term in the MMPBSA calculations. Our results show that the Pearson correlation coefficients of MMPBSA-calculated values with respect to experimental binding free energies can be drastically improved from 0.48 to 0.90 by adoption of ANI-ML potentials in replacement of MM energy terms in the equation, referred to as ANI-PBSA. Moreover, we show that the SASA/SAV-combined equation in the scaled particle theory (SPT) can be a better choice to model nonpolar solvation term, reaching nearly the same accuracy by ANI-PBSA calculations. Finally, we introduce two different values of interior dielectric constants, which could be an alternative strategy between the single and variable constant definitions.
Collapse
Affiliation(s)
- Ebru Akkus
- Department of Chemistry, Gebze Technical University, 41400 Kocaeli, Turkey
| | - Omer Tayfuroglu
- Department of Chemistry, Gebze Technical University, 41400 Kocaeli, Turkey
| | - Muslum Yildiz
- Department of Molecular Biology and Genetics, Gebze Technical University, 41400 Kocaeli, Turkey
| | - Abdulkadir Kocak
- Department of Chemistry, Gebze Technical University, 41400 Kocaeli, Turkey
| |
Collapse
|
30
|
Chen W, Cui D, Jerome SV, Michino M, Lenselink EB, Huggins DJ, Beautrait A, Vendome J, Abel R, Friesner RA, Wang L. Enhancing Hit Discovery in Virtual Screening through Absolute Protein-Ligand Binding Free-Energy Calculations. J Chem Inf Model 2023; 63:3171-3185. [PMID: 37167486 DOI: 10.1021/acs.jcim.3c00013] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
In the hit identification stage of drug discovery, a diverse chemical space needs to be explored to identify initial hits. Contrary to empirical scoring functions, absolute protein-ligand binding free-energy perturbation (ABFEP) provides a theoretically more rigorous and accurate description of protein-ligand binding thermodynamics and could, in principle, greatly improve the hit rates in virtual screening. In this work, we describe an implementation of an accurate and reliable ABFEP method in FEP+. We validated the ABFEP method on eight congeneric compound series binding to eight protein receptors including both neutral and charged ligands. For ligands with net charges, the alchemical ion approach is adopted to avoid artifacts in electrostatic potential energy calculations. The calculated binding free energies correlate with experimental results with a weighted average of R2 = 0.55 for the entire dataset. We also observe an overall root-mean-square error (RMSE) of 1.1 kcal/mol after shifting the zero-point of the simulation data to match the average experimental values. Through ABFEP calculations using apo versus holo protein structures, we demonstrated that the protein conformational and protonation state changes between the apo and holo proteins are the main physical factors contributing to the protein reorganization free energy manifested by the overestimation of raw ABFEP calculated binding free energies using the holo structures of the proteins. Furthermore, we performed ABFEP calculations in three virtual screening applications for hit enrichment. ABFEP greatly improves the hit rates as compared to docking scores or other methods like metadynamics. The good performance of ABFEP in rank ordering compounds demonstrated in this work confirms it as a useful tool to improve the hit rates in virtual screening, thus facilitating hit discovery.
Collapse
Affiliation(s)
- Wei Chen
- Schrödinger, Inc., 1540 Broadway, 24th Floor, New York, New York 10036, United States
| | - Di Cui
- Schrödinger, Inc., 1540 Broadway, 24th Floor, New York, New York 10036, United States
| | - Steven V Jerome
- Schrödinger, Inc., 10201 Wateridge Circle, Suite 220, San Diego, California 92121, United States
| | - Mayako Michino
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th Street, New York, New York 10065, United States
| | | | - David J Huggins
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th Street, New York, New York 10065, United States
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, New York 10065, United States
| | - Alexandre Beautrait
- Schrödinger, Inc., 1540 Broadway, 24th Floor, New York, New York 10036, United States
| | - Jeremie Vendome
- Schrödinger, Inc., 1540 Broadway, 24th Floor, New York, New York 10036, United States
| | - Robert Abel
- Schrödinger, Inc., 1540 Broadway, 24th Floor, New York, New York 10036, United States
| | - Richard A Friesner
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Lingle Wang
- Schrödinger, Inc., 1540 Broadway, 24th Floor, New York, New York 10036, United States
| |
Collapse
|
31
|
Moore JH, Margreitter C, Janet JP, Engkvist O, de Groot BL, Gapsys V. Automated relative binding free energy calculations from SMILES to ΔΔG. Commun Chem 2023; 6:82. [PMID: 37106032 PMCID: PMC10140266 DOI: 10.1038/s42004-023-00859-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 03/22/2023] [Indexed: 04/29/2023] Open
Abstract
In drug discovery, computational methods are a key part of making informed design decisions and prioritising experiments. In particular, optimizing compound affinity is a central concern during the early stages of development. In the last 10 years, alchemical free energy (FE) calculations have transformed our ability to incorporate accurate in silico potency predictions in design decisions, and represent the 'gold standard' for augmenting experiment-driven drug discovery. However, relative FE calculations are complex to set up, require significant expert intervention to prepare the calculation and analyse the results or are provided only as closed-source software, not allowing for fine-grained control over the underlying settings. In this work, we introduce an end-to-end relative FE workflow based on the non-equilibrium switching approach that facilitates calculation of binding free energies starting from SMILES strings. The workflow is implemented using fully modular steps, allowing various components to be exchanged depending on licence availability. We further investigate the dependence of the calculated free energy accuracy on the initial ligand pose generated by various docking algorithms. We show that both commercial and open-source docking engines can be used to generate poses that lead to good correlation of free energies with experimental reference data.
Collapse
Affiliation(s)
- J Harry Moore
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Jon Paul Janet
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Ola Engkvist
- Molecular AI, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| | - Bert L de Groot
- Computational Biomolecular Dynamics Group, Department of Theoretical and Computational Biophysics, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, D-37077, Göttingen, Germany.
| | - Vytautas Gapsys
- Computational Biomolecular Dynamics Group, Department of Theoretical and Computational Biophysics, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, D-37077, Göttingen, Germany.
| |
Collapse
|
32
|
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: 366] [Impact Index Per Article: 183.0] [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
|
33
|
Potlitz F, Link A, Schulig L. Advances in the discovery of new chemotypes through ultra-large library docking. Expert Opin Drug Discov 2023; 18:303-313. [PMID: 36714919 DOI: 10.1080/17460441.2023.2171984] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION The size and complexity of virtual screening libraries in drug discovery have skyrocketed in recent years, reaching up to multiple billions of accessible compounds. However, virtual screening of such ultra-large libraries poses several challenges associated with preparing the libraries, sampling, and pre-selection of suitable compounds. The utilization of artificial intelligence (AI)-assisted screening approaches, such as deep learning, poses a promising countermeasure to deal with this rapidly expanding chemical space. For example, various AI-driven methods were recently successfully used to identify novel small molecule inhibitors of the SARS-CoV-2 main protease (Mpro). AREAS COVERED This review focuses on presenting various kinds of virtual screening methods suitable for dealing with ultra-large libraries. Challenges associated with these computational methodologies are discussed, and recent advances are highlighted in the example of the discovery of novel Mpro inhibitors targeting the SARS-CoV-2 virus. EXPERT OPINION With the rapid expansion of the virtual chemical space, the methodologies for docking and screening such quantities of molecules need to keep pace. Employment of AI-driven screening compounds has already been shown to be effective in a range from a few thousand to multiple billion compounds, furthered by de novo generation of drug-like molecules without human interference.
Collapse
Affiliation(s)
- Felix Potlitz
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Germany
| | - Andreas Link
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Germany
| | - Lukas Schulig
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Germany
| |
Collapse
|
34
|
Losev TV, Gerasimov IS, Panova MV, Lisov AA, Abdyusheva YR, Rusina PV, Zaletskaya E, Stroganov OV, Medvedev MG, Novikov FN. Quantum Mechanical-Cluster Approach to Solve the Bioisosteric Replacement Problem in Drug Design. J Chem Inf Model 2023; 63:1239-1248. [PMID: 36763797 DOI: 10.1021/acs.jcim.2c01212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Bioisosteres are molecules that differ in substituents but still have very similar shapes. Bioisosteric replacements are ubiquitous in modern drug design, where they are used to alter metabolism, change bioavailability, or modify activity of the lead compound. Prediction of relative affinities of bioisosteres with computational methods is a long-standing task; however, the very shape closeness makes bioisosteric substitutions almost intractable for computational methods, which use standard force fields. Here, we design a quantum mechanical (QM)-cluster approach based on the GFN2-xTB semi-empirical quantum-chemical method and apply it to a set of H → F bioisosteric replacements. The proposed methodology enables advanced prediction of biological activity change upon bioisosteric substitution of -H with -F, with the standard deviation of 0.60 kcal/mol, surpassing the ChemPLP scoring function (0.83 kcal/mol), and making QM-based ΔΔG estimation comparable to ∼0.42 kcal/mol standard deviation of in vitro experiment. The speed of the method and lack of tunable parameters makes it affordable in current drug research.
Collapse
Affiliation(s)
- Timofey V Losev
- N.D. Zelinsky Institute of Organic Chemistry of Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russian Federation.,Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russian Federation.,A.N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavilov Str. 28, 119991 Moscow, Russian Federation
| | - Igor S Gerasimov
- N.D. Zelinsky Institute of Organic Chemistry of Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russian Federation.,Department of Chemistry, Kyungpook National University, Daegu 41566, South Korea
| | - Maria V Panova
- N.D. Zelinsky Institute of Organic Chemistry of Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russian Federation
| | - Alexey A Lisov
- N.D. Zelinsky Institute of Organic Chemistry of Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russian Federation
| | - Yana R Abdyusheva
- N.D. Zelinsky Institute of Organic Chemistry of Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russian Federation.,National Research University Higher School of Economics, Myasnitskaya Street 20, 101000 Moscow, Russian Federation
| | - Polina V Rusina
- N.D. Zelinsky Institute of Organic Chemistry of Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russian Federation
| | - Eugenia Zaletskaya
- N.D. Zelinsky Institute of Organic Chemistry of Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russian Federation.,National Research University Higher School of Economics, Myasnitskaya Street 20, 101000 Moscow, Russian Federation
| | - Oleg V Stroganov
- BioMolTech Corp., 226 York Mills Rd, Toronto, Ontario M2L 1L1, Canada
| | - Michael G Medvedev
- N.D. Zelinsky Institute of Organic Chemistry of Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russian Federation
| | - Fedor N Novikov
- N.D. Zelinsky Institute of Organic Chemistry of Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russian Federation.,National Research University Higher School of Economics, Myasnitskaya Street 20, 101000 Moscow, Russian Federation
| |
Collapse
|
35
|
Schieferdecker S, Vock E. Development of Pharmacophore Models for the Important Off-Target 5-HT 2B Receptor. J Med Chem 2023; 66:1509-1521. [PMID: 36621987 DOI: 10.1021/acs.jmedchem.2c01679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Toxicity is a major cause of attrition in the development of pharmaceuticals, and the off-target effects are a frequent contributor. The 5-HT2B receptor agonism is known to be responsible for a variety of safety concerns including valvular heart disease which was the cause for the withdrawal of several compounds from the market. An early detection of potential binding to this receptor is thus desirable. Herein, we present the identification of key amino acid residues in the active site of 5-HT2B by molecular dynamics simulations, the development of pharmacophore models and their performance on in-house data, and a structurally highly diverse subset of Enamine REAL labeled for 5-HT2B activity by a machine learning model. These models may be used as filters employed on screening compound sets for the early filtration of compounds with potential 5-HT2B off-target liabilities.
Collapse
Affiliation(s)
- Sebastian Schieferdecker
- Department of Nonclinical Drug Safety, Germany, Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach88397, Germany
| | - Esther Vock
- Department of Nonclinical Drug Safety, Germany, Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach88397, Germany
| |
Collapse
|
36
|
Souza FPS, Heinzelmann G, Caramori GF. Investigating the Solvent Effects on Binding Affinity of PAHs-ExBox 4+ Complexes: An Alchemical Approach. J Phys Chem B 2023; 127:249-260. [PMID: 36594853 DOI: 10.1021/acs.jpcb.2c06271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are polluting agents, produced naturally or artificially, widely dispersed in the environment and potentially carcinogenic and immunotoxic to humans and animals, mainly for marine life. Recently, a tetracationic box-shaped cyclophane (ExBox4+) was synthesized, fully characterized, and revealed to form host-guest complexes with PAHs in acetonitrile, demonstrating the potential ability for it to act as a PAHs scavenger. This work investigates, through Molecular Dynamics (MD) simulations, the binding affinity between different PAHs and ExBox4+ in different solvents: chloroform (nonpolar), acetonitrile (polar protic), and water (polar protic). An alchemical method of simultaneous decoupling-recoupling (SDR) was used and implemented in a newly developed Python program called GHOAT, which fully automates the calculation of binding free energies and invokes the AMBER 2020 simulation package. The results showed that the affinity between ExBox4+ and PAHs in water is much larger than in organic media, with free energies between -5 and -20 kcal/mol, being able to act as a PHAs scavenger with great potential for applications in environmental chemistry such as soil washing. The results also reveal a significant correlation with the experimental available ΔG values. The methodology employed presents itself as an important tool for the in silico determination of binding affinities, not only available for charged cyclophanes but also extensible to several other HG supramolecular systems in condensed media, aiding in the rational design of host-guest systems in a significant way.
Collapse
Affiliation(s)
- Fábio P S Souza
- Departamento de Química, Universidade Federal de Santa Catarina (UFSC), Campus Universitário Trindade, 88040-900, Florianópolis, Santa Catarina, Brazil.,Instituto Federal Catarinense, 89070-270, Blumenau, Santa Catarina, Brazil
| | - Germano Heinzelmann
- Departamento de Física, Universidade Federal de Santa Catarina (UFSC), Campus Universitário Trindade, 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Giovanni F Caramori
- Departamento de Química, Universidade Federal de Santa Catarina (UFSC), Campus Universitário Trindade, 88040-900, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
37
|
Gizzio J, Thakur A, Haldane A, Levy RM. Evolutionary divergence in the conformational landscapes of tyrosine vs serine/threonine kinases. eLife 2022; 11:83368. [PMID: 36562610 PMCID: PMC9822262 DOI: 10.7554/elife.83368] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022] Open
Abstract
Inactive conformations of protein kinase catalytic domains where the DFG motif has a "DFG-out" orientation and the activation loop is folded present a druggable binding pocket that is targeted by FDA-approved 'type-II inhibitors' in the treatment of cancers. Tyrosine kinases (TKs) typically show strong binding affinity with a wide spectrum of type-II inhibitors while serine/threonine kinases (STKs) usually bind more weakly which we suggest here is due to differences in the folded to extended conformational equilibrium of the activation loop between TKs vs. STKs. To investigate this, we use sequence covariation analysis with a Potts Hamiltonian statistical energy model to guide absolute binding free-energy molecular dynamics simulations of 74 protein-ligand complexes. Using the calculated binding free energies together with experimental values, we estimated free-energy costs for the large-scale (~17-20 Å) conformational change of the activation loop by an indirect approach, circumventing the very challenging problem of simulating the conformational change directly. We also used the Potts statistical potential to thread large sequence ensembles over active and inactive kinase states. The structure-based and sequence-based analyses are consistent; together they suggest TKs evolved to have free-energy penalties for the classical 'folded activation loop' DFG-out conformation relative to the active conformation, that is, on average, 4-6 kcal/mol smaller than the corresponding values for STKs. Potts statistical energy analysis suggests a molecular basis for this observation, wherein the activation loops of TKs are more weakly 'anchored' against the catalytic loop motif in the active conformation and form more stable substrate-mimicking interactions in the inactive conformation. These results provide insights into the molecular basis for the divergent functional properties of TKs and STKs, and have pharmacological implications for the target selectivity of type-II inhibitors.
Collapse
Affiliation(s)
- Joan Gizzio
- Center for Biophysics and Computational Biology, Temple University, Philadelphia, United States.,Department of Chemistry, Temple University, Philadelphia, United States
| | - Abhishek Thakur
- Center for Biophysics and Computational Biology, Temple University, Philadelphia, United States.,Department of Chemistry, Temple University, Philadelphia, United States
| | - Allan Haldane
- Center for Biophysics and Computational Biology, Temple University, Philadelphia, United States.,Department of Physics, Temple University, Philadelphia, United States
| | - Ronald M Levy
- Center for Biophysics and Computational Biology, Temple University, Philadelphia, United States.,Department of Chemistry, Temple University, Philadelphia, United States
| |
Collapse
|
38
|
Zhang H, Kim S, Im W. Practical Guidance for Consensus Scoring and Force Field Selection in Protein-Ligand Binding Free Energy Simulations. J Chem Inf Model 2022; 62:6084-6093. [PMID: 36399655 PMCID: PMC9772090 DOI: 10.1021/acs.jcim.2c01115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The advances in ligand binding affinity prediction have been fostered by system generation tools and improved force fields (FFs). CHARMM-GUI Free Energy Calculator provides input and postprocessing scripts for AMBER-TI free energy calculations with various FFs. In this study, we used 12 different FF combinations (ff14SB and ff19SB for protein, GAFF2.2 and OpenFF for ligand, and TIP3P, TIP4PEW, and OPC for water) to calculate relative binding free energies (ΔΔGbind) for 80 alchemical transformations (among the JACS benchmark set) with different numbers of λ windows (12 or 21) and simulation times (1, 5, or 10 ns). Our results show that 12 λ windows and 5 ns simulation time for each window are sufficient to obtain reliable ΔΔGbind with 4 independent runs for the current benchmark set. While there is no statistically noticeable performance difference among 12 different FF combinations compared to the experimental values, a combination of ff14SB + GAFF2.2 + TIP3P FFs appears to be best with a mean unsigned error of 0.87 [0.69, 1.07] kcal/mol, a root-mean-square error of 1.22 [0.94, 1.50] kcal/mol, a Pearson's correlation of 0.64 [0.52, 0.76], a Spearman's correlation of 0.73 [0.58, 0.83], and a Kendell's correlation of 0.54 [0.42, 0.64]. This large-scale ΔΔGbind calculation study provides useful information about ΔΔGbind prediction with different AMBER FF combinations and presents valuable suggestions for FF selection in AMBER-TI ΔΔGbind calculations.
Collapse
Affiliation(s)
- Han Zhang
- Departments of Biological Sciences, Chemistry, Bioengineering, and Computer Science and Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, USA
| | - Seonghoon Kim
- School of Computational Sciences, Korea Institute for Advanced Study, Seoul 02455, Republic of Korea
| | - Wonpil Im
- Departments of Biological Sciences, Chemistry, Bioengineering, and Computer Science and Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, USA,Corresponding Author:
| |
Collapse
|
39
|
Ge Y, Baumann HM, Mobley DL. Absolute Binding Free Energy Calculations for Buried Water Molecules. J Chem Theory Comput 2022; 18:6482-6499. [PMID: 36197451 PMCID: PMC9873352 DOI: 10.1021/acs.jctc.2c00658] [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: 01/27/2023]
Abstract
Water often plays a key role in mediating protein-ligand interactions. Understanding contributions from active-site water molecules to binding thermodynamics of a ligand is important in predicting binding free energies for ligand optimization. In this work, we tested a non-equilibrium switching method for absolute binding free energy calculations on water molecules in binding sites of 13 systems. We discuss the lessons we learned about identified issues that affected our calculations and ways to address them. This work fits with our larger focus on how to do accurate ligand binding free energy calculations when water rearrangements are very slow, such as rearrangements due to ligand modification (as in relative free energy calculations) or ligand binding (as in absolute free energy calculations). The method studied in this work can potentially be used to account for limited water sampling via providing endpoint corrections to free energy calculations using our calculated binding free energy of water.
Collapse
Affiliation(s)
- Yunhui Ge
- Department of Pharmaceutical Sciences, University of California, Irvine, California92697, United States
| | - Hannah M Baumann
- Department of Pharmaceutical Sciences, University of California, Irvine, California92697, United States
| | - David L Mobley
- Department of Pharmaceutical Sciences, University of California, Irvine, California92697, United States
- Department of Chemistry, University of California, Irvine, California92697, United States
| |
Collapse
|
40
|
Fu H, Zhou Y, Jing X, Shao X, Cai W. Meta-Analysis Reveals That Absolute Binding Free-Energy Calculations Approach Chemical Accuracy. J Med Chem 2022; 65:12970-12978. [PMID: 36179112 DOI: 10.1021/acs.jmedchem.2c00796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Systematic and quantitative analysis of the reliability of formally exact methods that in silico calculate absolute protein-ligand binding free energies remains lacking. Here, we provide, for the first time, evidence-based information on the reliability of these methods by statistically studying 853 cases from 34 different research groups through meta-analysis. The results show that formally exact methods approach chemical accuracy (error = 1.58 kcal/mol), even if people are challenging difficult tasks such as blind drug screening in recent years. The geometrical-pathway-based methods prove to possess a better convergence ability than the alchemical ones, while the latter have a larger application range. We also reveal the importance of always using the latest force fields to guarantee reliability and discuss the pros and cons of turning to an implicit solvent model in absolute binding free-energy calculations. Moreover, based on the meta-analysis, an evidence-based guideline for in silico binding free-energy calculations is provided.
Collapse
Affiliation(s)
- Haohao Fu
- Research Center for Analytical Sciences, Frontiers Science Center for New Organic Matter, College of Chemistry, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin300071, China.,Haihe Laboratory of Sustainable Chemical Transformations, Tianjin300192, China
| | - Yan Zhou
- School of Medicine, Nankai University, Tianjin300071, China.,Department of Ultrasound, Tianjin Third Central Hospital, Tianjin300170, China
| | - Xiang Jing
- Department of Ultrasound, Tianjin Third Central Hospital, Tianjin300170, China
| | - Xueguang Shao
- Research Center for Analytical Sciences, Frontiers Science Center for New Organic Matter, College of Chemistry, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin300071, China.,Haihe Laboratory of Sustainable Chemical Transformations, Tianjin300192, China
| | - Wensheng Cai
- Research Center for Analytical Sciences, Frontiers Science Center for New Organic Matter, College of Chemistry, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin300071, China.,Haihe Laboratory of Sustainable Chemical Transformations, Tianjin300192, China
| |
Collapse
|
41
|
Fernández-Bachiller MI, Hwang S, Schembri ME, Lindemann P, Guberman M, Herziger S, Specker E, Matter H, Will DW, Czech J, Wagner M, Bauer A, Schreuder H, Ritter K, Urmann M, Wehner V, Sun H, Nazaré M. Probing Factor Xa Protein-Ligand Interactions: Accurate Free Energy Calculations and Experimental Validations of Two Series of High-Affinity Ligands. J Med Chem 2022; 65:13013-13028. [PMID: 36178213 DOI: 10.1021/acs.jmedchem.2c00865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The accurate prediction of protein-ligand binding affinity belongs to one of the central goals in computer-based drug design. Molecular dynamics (MD)-based free energy calculations have become increasingly popular in this respect due to their accuracy and solid theoretical basis. Here, we present a combined study which encompasses experimental and computational studies on two series of factor Xa ligands, which enclose a broad chemical space including large modifications of the central scaffold. Using this integrated approach, we identified several new ligands with different heterocyclic scaffolds different from the previously identified indole-2-carboxamides that show superior or similar affinity. Furthermore, the so far underexplored terminal alkyne moiety proved to be a suitable non-classical bioisosteric replacement for the higher halogen-π aryl interactions. With this challenging example, we demonstrated the ability of the MD-based non-equilibrium free energy calculation approach for guiding crucial modifications in the lead optimization process, such as scaffold replacement and single-site modifications at molecular interaction hot spots.
Collapse
Affiliation(s)
| | - Songhwan Hwang
- Structural Chemistry and Computational Biophysics, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle Str. 10, 13125Berlin, Germany
| | - María Elena Schembri
- Medizinische Chemie, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle Str. 10, 13125Berlin, Germany
| | - Peter Lindemann
- Medizinische Chemie, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle Str. 10, 13125Berlin, Germany
| | - Mónica Guberman
- Medizinische Chemie, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle Str. 10, 13125Berlin, Germany
| | - Svenja Herziger
- Medizinische Chemie, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle Str. 10, 13125Berlin, Germany
| | - Edgar Specker
- Medizinische Chemie, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle Str. 10, 13125Berlin, Germany
| | - Hans Matter
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark-Höchst, Building G878, 65926Frankfurt am Main, Germany
| | - David W Will
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark-Höchst, Building G878, 65926Frankfurt am Main, Germany
| | - Jörg Czech
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark-Höchst, Building G878, 65926Frankfurt am Main, Germany
| | - Michael Wagner
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark-Höchst, Building G878, 65926Frankfurt am Main, Germany
| | - Armin Bauer
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark-Höchst, Building G878, 65926Frankfurt am Main, Germany
| | - Herman Schreuder
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark-Höchst, Building G878, 65926Frankfurt am Main, Germany
| | - Kurt Ritter
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark-Höchst, Building G878, 65926Frankfurt am Main, Germany
| | - Matthias Urmann
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark-Höchst, Building G878, 65926Frankfurt am Main, Germany
| | - Volkmar Wehner
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark-Höchst, Building G878, 65926Frankfurt am Main, Germany
| | - Han Sun
- Structural Chemistry and Computational Biophysics, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle Str. 10, 13125Berlin, Germany.,Institute of Chemistry, Technische Universität Berlin, Strasse des 17. Juni 135, 10623Berlin, Germany
| | - Marc Nazaré
- Medizinische Chemie, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle Str. 10, 13125Berlin, Germany
| |
Collapse
|
42
|
Sandoval-Pérez A, Winger BA, Jacobson MP. Assessing the Activation of Tyrosine Kinase KIT through Free Energy Calculations. J Chem Theory Comput 2022; 18:6251-6258. [PMID: 36166736 PMCID: PMC9558371 DOI: 10.1021/acs.jctc.2c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
KIT is a type 3 receptor tyrosine kinase that plays a crucial role in cellular growth and proliferation. Mutations in KIT can dysregulate its active-inactive equilibrium. Activating mutations drive cancer growth, while deactivating mutations result in the loss of skin and hair pigmentation in a disease known as piebaldism. Here, we propose a method based on molecular dynamics and free energy calculations to predict the functional effect of KIT mutations. Our calculations may have important clinical implications by defining the functional significance of previously uncharacterized KIT mutations and guiding targeted therapy.
Collapse
Affiliation(s)
- Angélica Sandoval-Pérez
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco 94158, California, United States
| | - Beth Apsel Winger
- Department of Pediatrics, Division of Hematology and Oncology, University of California, San Francisco, San Francisco 94158, California, United States
| | - Matthew P Jacobson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco 94158, California, United States
| |
Collapse
|
43
|
Alibay I, Magarkar A, Seeliger D, Biggin PC. Evaluating the use of absolute binding free energy in the fragment optimisation process. Commun Chem 2022; 5:105. [PMID: 36697714 PMCID: PMC9814858 DOI: 10.1038/s42004-022-00721-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/10/2022] [Indexed: 02/01/2023] Open
Abstract
Key to the fragment optimisation process within drug design is the need to accurately capture the changes in affinity that are associated with a given set of chemical modifications. Due to the weakly binding nature of fragments, this has proven to be a challenging task, despite recent advancements in leveraging experimental and computational methods. In this work, we evaluate the use of Absolute Binding Free Energy (ABFE) calculations in guiding fragment optimisation decisions, retrospectively calculating binding free energies for 59 ligands across 4 fragment elaboration campaigns. We first demonstrate that ABFEs can be used to accurately rank fragment-sized binders with an overall Spearman's r of 0.89 and a Kendall τ of 0.67, although often deviating from experiment in absolute free energy values with an RMSE of 2.75 kcal/mol. We then also show that in several cases, retrospective fragment optimisation decisions can be supported by the ABFE calculations. Comparing against cheaper endpoint methods, namely Nwat-MM/GBSA, we find that ABFEs offer better ranking power and correlation metrics. Our results indicate that ABFE calculations can usefully guide fragment elaborations to maximise affinity.
Collapse
Affiliation(s)
- Irfan Alibay
- Department of Biochemistry, The University of Oxford, South Parks Road, OX1 3QU, Oxford, UK
| | - Aniket Magarkar
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach an de Riß, Germany
| | - Daniel Seeliger
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach an de Riß, Germany
- Exscientia Inc, Office 400E, 2125 Biscayne Blvd, Miami, FL, 33137, USA
| | - Philip Charles Biggin
- Department of Biochemistry, The University of Oxford, South Parks Road, OX1 3QU, Oxford, UK.
| |
Collapse
|
44
|
Vakali V, Papadourakis M, Georgiou N, Zoupanou N, Diamantis DA, Javornik U, Papakyriakopoulou P, Plavec J, Valsami G, Tzakos AG, Tzeli D, Cournia Z, Mauromoustakos T. Comparative Interaction Studies of Quercetin with 2-Hydroxyl-propyl-β-cyclodextrin and 2,6-Methylated-β-cyclodextrin. Molecules 2022; 27:5490. [PMID: 36080258 PMCID: PMC9458201 DOI: 10.3390/molecules27175490] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/06/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Quercetin (QUE) is a well-known natural product that can exert beneficial properties on human health. However, due to its low solubility its bioavailability is limited. In the present study, we examine whether its formulation with two cyclodextrins (CDs) may enhance its pharmacological profile. Comparative interaction studies of quercetin with 2-hydroxyl-propyl-β-cyclodextrin (2HP-β-CD) and 2,6-methylated cyclodextrin (2,6Me-β-CD) were performed using NMR spectroscopy, DFT calculations, and in silico molecular dynamics (MD) simulations. Using T1 relaxation experiments and 2D DOSY it was illustrated that both cyclodextrin vehicles can host quercetin. Quantum mechanical calculations showed the formation of hydrogen bonds between QUE with 2HP-β-CD and 2,6Μe-β-CD. Six hydrogen bonds are formed ranging between 2 to 2.8 Å with 2HP-β-CD and four hydrogen bonds within 2.8 Å with 2,6Μe-β-CD. Calculations of absolute binding free energies show that quercetin binds favorably to both 2,6Me-β-CD and 2HP-β-CD. MM/GBSA results show equally favorable binding of quercetin in the two CDs. Fluorescence spectroscopy shows moderate binding of quercetin in 2HP-β-CD (520 M-1) and 2,6Me-β-CD (770 M-1). Thus, we propose that both formulations (2HP-β-CD:quercetin, 2,6Me-β-CD:quercetin) could be further explored and exploited as small molecule carriers in biological studies.
Collapse
Affiliation(s)
- Vasiliki Vakali
- Organic Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopollis Zografou, 11571 Athens, Greece
| | - Michail Papadourakis
- Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou, 11527 Athens, Greece
| | - Nikitas Georgiou
- Organic Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopollis Zografou, 11571 Athens, Greece
| | - Nikoletta Zoupanou
- Organic Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopollis Zografou, 11571 Athens, Greece
| | - Dimitrios A. Diamantis
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, 45110 Ioannina, Greece
| | - Uroš Javornik
- Slovenian NMR Centre, National Institute of Chemistry, SI-1001 Ljubljana, Slovenia
| | - Paraskevi Papakyriakopoulou
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Janez Plavec
- Slovenian NMR Centre, National Institute of Chemistry, SI-1001 Ljubljana, Slovenia
| | - Georgia Valsami
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Andreas G. Tzakos
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, 45110 Ioannina, Greece
- Institute of Materials Science and Computing, University Research Center of Ioannina (URCI), 45110 Ioannina, Greece
| | - Demeter Tzeli
- Laboratory of Physical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 11571 Athens, Greece
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Zoe Cournia
- Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou, 11527 Athens, Greece
| | - Thomas Mauromoustakos
- Organic Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopollis Zografou, 11571 Athens, Greece
| |
Collapse
|
45
|
Akkus E, Tayfuroglu O, Yildiz M, Kocak A. Accurate Binding Free Energy Method from End-State MD Simulations. J Chem Inf Model 2022; 62:4095-4106. [PMID: 35972783 PMCID: PMC9472276 DOI: 10.1021/acs.jcim.2c00601] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
![]()
Herein, we introduce a new strategy to estimate binding
free energies
using end-state molecular dynamics simulation trajectories. The method
is adopted from linear interaction energy (LIE) and ANI-2x neural
network potentials (machine learning) for the atomic simulation environment
(ASE). It predicts the single-point interaction energies between ligand–protein
and ligand–solvent pairs at the accuracy of the wb97x/6-31G*
level for the conformational space that is sampled by molecular dynamics
(MD) simulations. Our results on 54 protein–ligand complexes
show that the method can be accurate and have a correlation of R = 0.87–0.88 to the experimental binding free energies,
outperforming current end-state methods with reduced computational
cost. The method also allows us to compare BFEs of ligands with different
scaffolds. The code is available free of charge (documentation and
test files) at https://github.com/otayfuroglu/deepQM.
Collapse
Affiliation(s)
- Ebru Akkus
- Department of Bioengineering, Gebze Technical University, 41400 Gebze, Kocaeli, Turkey
| | - Omer Tayfuroglu
- Department of Chemistry, Gebze Technical University, 41400 Gebze, Kocaeli, Turkey
| | - Muslum Yildiz
- Department of Molecular Biology and Genetics, Gebze Technical University, 41400 Gebze, Kocaeli, Turkey
| | - Abdulkadir Kocak
- Department of Chemistry, Gebze Technical University, 41400 Gebze, Kocaeli, Turkey
| |
Collapse
|
46
|
Feng M, Heinzelmann G, Gilson MK. Absolute binding free energy calculations improve enrichment of actives in virtual compound screening. Sci Rep 2022; 12:13640. [PMID: 35948614 PMCID: PMC9365818 DOI: 10.1038/s41598-022-17480-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/26/2022] [Indexed: 12/04/2022] Open
Abstract
We determined the effectiveness of absolute binding free energy (ABFE) calculations to refine the selection of active compounds in virtual compound screening, a setting where the more commonly used relative binding free energy approach is not readily applicable. To do this, we conducted baseline docking calculations of structurally diverse compounds in the DUD-E database for three targets, BACE1, CDK2 and thrombin, followed by ABFE calculations for compounds with high docking scores. The docking calculations alone achieved solid enrichment of active compounds over decoys. Encouragingly, the ABFE calculations then improved on this baseline. Analysis of the results emphasizes the importance of establishing high quality ligand poses as starting points for ABFE calculations, a nontrivial goal when processing a library of diverse compounds without informative co-crystal structures. Overall, our results suggest that ABFE calculations can play a valuable role in the drug discovery process.
Collapse
Affiliation(s)
- Mudong Feng
- Department of Chemistry and Biochemistry, and Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA, 92093, USA
| | - Germano Heinzelmann
- Departamento de Física, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Michael K Gilson
- Department of Chemistry and Biochemistry, and Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
47
|
Patel LA, Chau P, Debesai S, Darwin L, Neale C. Drug Discovery by Automated Adaptation of Chemical Structure and Identity. J Chem Theory Comput 2022; 18:5006-5024. [PMID: 35834740 DOI: 10.1021/acs.jctc.1c01271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Computer-aided drug design offers the potential to dramatically reduce the cost and effort required for drug discovery. While screening-based methods are valuable in the early stages of hit identification, they are frequently succeeded by iterative, hypothesis-driven computations that require recurrent investment of human time and intuition. To increase automation, we introduce a computational method for lead refinement that combines concerted dynamics of the ligand/protein complex via molecular dynamics simulations with integrated Monte Carlo-based changes in the chemical formula of the ligand. This approach, which we refer to as ligand-exchange Monte Carlo molecular dynamics, accounts for solvent- and entropy-based contributions to competitive binding free energies by coupling the energetics of bound and unbound states during the ligand-exchange attempt. Quantitative comparison of relative binding free energies to reference values from free energy perturbation, conducted in vacuum, indicates that ligand-exchange Monte Carlo molecular dynamics simulations sample relevant conformational ensembles and are capable of identifying strongly binding compounds. Additional simulations demonstrate the use of an implicit solvent model. We speculate that the use of chemical graphs in which exchanges are only permitted between ligands with sufficient similarity may enable an automated search to capture some of the benefits provided by human intuition during hypothesis-guided lead refinement.
Collapse
|
48
|
Bhati A, Coveney PV. Large Scale Study of Ligand-Protein Relative Binding Free Energy Calculations: Actionable Predictions from Statistically Robust Protocols. J Chem Theory Comput 2022; 18:2687-2702. [PMID: 35293737 PMCID: PMC9009079 DOI: 10.1021/acs.jctc.1c01288] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Indexed: 12/28/2022]
Abstract
The accurate and reliable prediction of protein-ligand binding affinities can play a central role in the drug discovery process as well as in personalized medicine. Of considerable importance during lead optimization are the alchemical free energy methods that furnish an estimation of relative binding free energies (RBFE) of similar molecules. Recent advances in these methods have increased their speed, accuracy, and precision. This is evident from the increasing number of retrospective as well as prospective studies employing them. However, such methods still have limited applicability in real-world scenarios due to a number of important yet unresolved issues. Here, we report the findings from a large data set comprising over 500 ligand transformations spanning over 300 ligands binding to a diverse set of 14 different protein targets which furnish statistically robust results on the accuracy, precision, and reproducibility of RBFE calculations. We use ensemble-based methods which are the only way to provide reliable uncertainty quantification given that the underlying molecular dynamics is chaotic. These are implemented using TIES (Thermodynamic Integration with Enhanced Sampling). Results achieve chemical accuracy in all cases. Ensemble simulations also furnish information on the statistical distributions of the free energy calculations which exhibit non-normal behavior. We find that the "enhanced sampling" method known as replica exchange with solute tempering degrades RBFE predictions. We also report definitively on numerous associated alchemical factors including the choice of ligand charge method, flexibility in ligand structure, and the size of the alchemical region including the number of atoms involved in transforming one ligand into another. Our findings provide a key set of recommendations that should be adopted for the reliable application of RBFE methods.
Collapse
Affiliation(s)
- Agastya
P. Bhati
- Centre
for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, United Kingdom
| | - Peter V. Coveney
- Centre
for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, United Kingdom
- Informatics
Institute, University of Amsterdam, P.O. Box 94323, 1090 GH Amsterdam, Netherlands
| |
Collapse
|
49
|
Kutzner C, Kniep C, Cherian A, Nordstrom L, Grubmüller H, de Groot BL, Gapsys V. GROMACS in the Cloud: A Global Supercomputer to Speed Up Alchemical Drug Design. J Chem Inf Model 2022; 62:1691-1711. [PMID: 35353508 PMCID: PMC9006219 DOI: 10.1021/acs.jcim.2c00044] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Indexed: 12/16/2022]
Abstract
We assess costs and efficiency of state-of-the-art high-performance cloud computing and compare the results to traditional on-premises compute clusters. Our use case is atomistic simulations carried out with the GROMACS molecular dynamics (MD) toolkit with a particular focus on alchemical protein-ligand binding free energy calculations. We set up a compute cluster in the Amazon Web Services (AWS) cloud that incorporates various different instances with Intel, AMD, and ARM CPUs, some with GPU acceleration. Using representative biomolecular simulation systems, we benchmark how GROMACS performs on individual instances and across multiple instances. Thereby we assess which instances deliver the highest performance and which are the most cost-efficient ones for our use case. We find that, in terms of total costs, including hardware, personnel, room, energy, and cooling, producing MD trajectories in the cloud can be about as cost-efficient as an on-premises cluster given that optimal cloud instances are chosen. Further, we find that high-throughput ligand-screening can be accelerated dramatically by using global cloud resources. For a ligand screening study consisting of 19 872 independent simulations or ∼200 μs of combined simulation trajectory, we made use of diverse hardware available in the cloud at the time of the study. The computations scaled-up to reach peak performance using more than 4 000 instances, 140 000 cores, and 3 000 GPUs simultaneously. Our simulation ensemble finished in about 2 days in the cloud, while weeks would be required to complete the task on a typical on-premises cluster consisting of several hundred nodes.
Collapse
Affiliation(s)
- Carsten Kutzner
- Department
of Theoretical and Computational Biophysics, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany
| | - Christian Kniep
- Amazon
Development Center Germany, Amazon Web Services, Krausenstr. 38, 10117 Berlin, Germany
| | - Austin Cherian
- Amazon
Web Services Singapore Pte Ltd, 23 Church St, #10-01, Singapore 049481
| | - Ludvig Nordstrom
- Amazon
Web Services, 60 Holborn
Viaduct, London EC1A 2FD, United Kingdom
| | - Helmut Grubmüller
- Department
of Theoretical and Computational Biophysics, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany
| | - Bert L. de Groot
- Computational
Biomolecular Dynamics Group, Max Planck
Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany
| | - Vytautas Gapsys
- Computational
Biomolecular Dynamics Group, Max Planck
Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany
| |
Collapse
|
50
|
At least three xenon binding sites in the glycine binding domain of the N-methyl D-aspartate receptor. Arch Biochem Biophys 2022; 724:109265. [DOI: 10.1016/j.abb.2022.109265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 11/21/2022]
|