1
|
Smith L, Novak B, Osato M, Mobley DL, Bowman GR. PopShift: A Thermodynamically Sound Approach to Estimate Binding Free Energies by Accounting for Ligand-Induced Population Shifts from a Ligand-Free Markov State Model. J Chem Theory Comput 2024; 20:1036-1050. [PMID: 38291966 PMCID: PMC10867841 DOI: 10.1021/acs.jctc.3c00870] [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: 08/08/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 02/01/2024]
Abstract
Obtaining accurate binding free energies from in silico screens has been a long-standing goal for the computational chemistry community. However, accuracy and computational cost are at odds with one another, limiting the utility of methods that perform this type of calculation. Many methods achieve massive scale by explicitly or implicitly assuming that the target protein adopts a single structure, or undergoes limited fluctuations around that structure, to minimize computational cost. Others simulate each protein-ligand complex of interest, accepting lower throughput in exchange for better predictions of binding affinities. Here, we present the PopShift framework for accounting for the ensemble of structures a protein adopts and their relative probabilities. Protein degrees of freedom are enumerated once, and then arbitrarily many molecules can be screened against this ensemble. Specifically, we use Markov state models (MSMs) as a compressed representation of a protein's thermodynamic ensemble. We start with a ligand-free MSM and then calculate how addition of a ligand shifts the populations of each protein conformational state based on the strength of the interaction between that protein conformation and the ligand. In this work we use docking to estimate the affinity between a given protein structure and ligand, but any estimator of binding affinities could be used in the PopShift framework. We test PopShift on the classic benchmark pocket T4 Lysozyme L99A. We find that PopShift is more accurate than common strategies, such as docking to a single structure and traditional ensemble docking─producing results that compare favorably with alchemical binding free energy calculations in terms of RMSE but not correlation─and may have a more favorable computational cost profile in some applications. In addition to predicting binding free energies and ligand poses, PopShift also provides insight into how the probability of different protein structures is shifted upon addition of various concentrations of ligand, providing a platform for predicting affinities and allosteric effects of ligand binding. Therefore, we expect PopShift will be valuable for hit finding and for providing insight into phenomena like allostery.
Collapse
Affiliation(s)
- Louis
G. Smith
- Departments
of Biochemistry & Biophysics and Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Borna Novak
- Department
of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Medical
Scientist Training Program, Washington University
in St. Louis, St. Louis, Missouri 63130, United
States
| | - Meghan Osato
- School
of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, California 92697, United States
| | - David L. Mobley
- School
of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, California 92697, United States
| | - Gregory R. Bowman
- Departments
of Biochemistry & Biophysics and Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
2
|
Smith LG, Novak B, Osato M, Mobley DL, Bowman GR. PopShift: A thermodynamically sound approach to estimate binding free energies by accounting for ligand-induced population shifts from a ligand-free MSM. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.14.549110. [PMID: 37503302 PMCID: PMC10370083 DOI: 10.1101/2023.07.14.549110] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Obtaining accurate binding free energies from in silico screens has been a longstanding goal for the computational chemistry community. However, accuracy and computational cost are at odds with one another, limiting the utility of methods that perform this type of calculation. Many methods achieve massive scale by explicitly or implicitly assuming that the target protein adopts a single structure, or undergoes limited fluctuations around that structure, to minimize computational cost. Others simulate each protein-ligand complex of interest, accepting lower throughput in exchange for better predictions of binding affinities. Here, we present the PopShift framework for accounting for the ensemble of structures a protein adopts and their relative probabilities. Protein degrees of freedom are enumerated once, and then arbitrarily many molecules can be screened against this ensemble. Specifically, we use Markov state models (MSMs) as a compressed representation of a protein's thermodynamic ensemble. We start with a ligand-free MSM and then calculate how addition of a ligand shifts the populations of each protein conformational state based on the strength of the interaction between that protein conformation and the ligand. In this work we use docking to estimate the affinity between a given protein structure and ligand, but any estimator of binding affinities could be used in the PopShift framework. We test PopShift on the classic benchmark pocket T4 Lysozyme L99A. We find that PopShift is more accurate than common strategies, such as docking to a single structure and traditional ensemble docking-producing results that compare favorably with alchemical binding free energy calculations in terms of RMSE but not correlation - and may have a more favorable computational cost profile in some applications. In addition to predicting binding free energies and ligand poses, PopShift also provides insight into how the probability of different protein structures is shifted upon addition of various concentrations of ligand, providing a platform for predicting affinities and allosteric effects of ligand binding. Therefore, we expect PopShift will be valuable for hit finding and for providing insight into phenomena like allostery.
Collapse
Affiliation(s)
- Louis G Smith
- University of Pennsylvania, Depts. of Biochemistry & Biophysics and Bioengineering
| | - Borna Novak
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics
- Medical Scientist Training Program, Washington University in St. Louis
| | - Meghan Osato
- University of California Irvine, School of Pharmacy and Pharmaceutical Sciences
| | - David L Mobley
- University of California Irvine, School of Pharmacy and Pharmaceutical Sciences
| | - Gregory R Bowman
- University of Pennsylvania, Depts. of Biochemistry & Biophysics and Bioengineering
| |
Collapse
|
3
|
Gutermuth T, Sieg J, Stohn T, Rarey M. Modeling with Alternate Locations in X-ray Protein Structures. J Chem Inf Model 2023; 63:2573-2585. [PMID: 37018549 DOI: 10.1021/acs.jcim.3c00100] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
In many molecular modeling applications, the standard procedure is still to handle proteins as single, rigid structures. While the importance of conformational flexibility is widely known, handling it remains challenging. Even the crystal structure of a protein usually contains variability exemplified in alternate side chain orientations or backbone segments. This conformational variability is encoded in PDB structure files by so-called alternate locations (AltLocs). Most modeling approaches either ignore AltLocs or resolve them with simple heuristics early on during structure import. We analyzed the occurrence and usage of AltLocs in the PDB and developed an algorithm to automatically handle AltLocs in PDB files enabling all structure-based methods using rigid structures to take the alternative protein conformations described by AltLocs into consideration. A respective software tool named AltLocEnumerator can be used as a structure preprocessor to easily exploit AltLocs. While the amount of data makes it difficult to show impact on a statistical level, handling AltLocs has a substantial impact on a case-by-case basis. We believe that the inspection and consideration of AltLocs is a very valuable approach in many modeling scenarios.
Collapse
Affiliation(s)
- Torben Gutermuth
- Universität Hamburg, ZBH - Center for Bioinformatics, Bundesstraße 43, 20146 Hamburg, Germany
| | - Jochen Sieg
- Universität Hamburg, ZBH - Center for Bioinformatics, Bundesstraße 43, 20146 Hamburg, Germany
| | - Tim Stohn
- Universität Hamburg, ZBH - Center for Bioinformatics, Bundesstraße 43, 20146 Hamburg, Germany
| | - Matthias Rarey
- Universität Hamburg, ZBH - Center for Bioinformatics, Bundesstraße 43, 20146 Hamburg, Germany
| |
Collapse
|
4
|
On the Rapid Calculation of Binding Affinities for Antigen and Antibody Design and Affinity Maturation Simulations. Antibodies (Basel) 2022; 11:antib11030051. [PMID: 35997345 PMCID: PMC9397028 DOI: 10.3390/antib11030051] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/23/2022] [Accepted: 08/01/2022] [Indexed: 02/05/2023] Open
Abstract
The accurate and efficient calculation of protein-protein binding affinities is an essential component in antibody and antigen design and optimization, and in computer modeling of antibody affinity maturation. Such calculations remain challenging despite advances in computer hardware and algorithms, primarily because proteins are flexible molecules, and thus, require explicit or implicit incorporation of multiple conformational states into the computational procedure. The astronomical size of the amino acid sequence space further compounds the challenge by requiring predictions to be computed within a short time so that many sequence variants can be tested. In this study, we compare three classes of methods for antibody/antigen (Ab/Ag) binding affinity calculations: (i) a method that relies on the physical separation of the Ab/Ag complex in equilibrium molecular dynamics (MD) simulations, (ii) a collection of 18 scoring functions that act on an ensemble of structures created using homology modeling software, and (iii) methods based on the molecular mechanics-generalized Born surface area (MM-GBSA) energy decomposition, in which the individual contributions of the energy terms are scaled to optimize agreement with the experiment. When applied to a set of 49 antibody mutations in two Ab/HIV gp120 complexes, all of the methods are found to have modest accuracy, with the highest Pearson correlations reaching about 0.6. In particular, the most computationally intensive method, i.e., MD simulation, did not outperform several scoring functions. The optimized energy decomposition methods provided marginally higher accuracy, but at the expense of requiring experimental data for parametrization. Within each method class, we examined the effect of the number of independent computational replicates, i.e., modeled structures or reinitialized MD simulations, on the prediction accuracy. We suggest using about ten modeled structures for scoring methods, and about five simulation replicates for MD simulations as a rule of thumb for obtaining reasonable convergence. We anticipate that our study will be a useful resource for practitioners working to incorporate binding affinity calculations within their protein design and optimization process.
Collapse
|
5
|
Al-Mosawi SK, Al-Hazam HA, Abbas AF, Nasif ZN, Saeed BA, Al-Masoudi N. Synthesis and QSAR of Novel Ketoprofen–Chalcone-Amide Hybrides as Acetylcholinesterase (AChE) Inhibitors for Possible Treatment of Alzheimer Disease. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1068162022040045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
6
|
Firouzi R, Ashouri M, Karimi‐Jafari MH. Structural insights into the substrate‐binding site of main protease for the structure‐based COVID‐19 drug discovery. Proteins 2022; 90:1090-1101. [DOI: 10.1002/prot.26318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/30/2022] [Accepted: 01/31/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Rohoullah Firouzi
- Department of Physical Chemistry Chemistry and Chemical Engineering Research Center of Iran Tehran Iran
| | - Mitra Ashouri
- Department of Physical Chemistry, School of Chemistry, College of Science University of Tehran Tehran Iran
| | | |
Collapse
|
7
|
Miñarro-Lleonar M, Ruiz-Carmona S, Alvarez-Garcia D, Schmidtke P, Barril X. Development of an Automatic Pipeline for Participation in the CELPP Challenge. Int J Mol Sci 2022; 23:ijms23094756. [PMID: 35563148 PMCID: PMC9105952 DOI: 10.3390/ijms23094756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 12/01/2022] Open
Abstract
The prediction of how a ligand binds to its target is an essential step for Structure-Based Drug Design (SBDD) methods. Molecular docking is a standard tool to predict the binding mode of a ligand to its macromolecular receptor and to quantify their mutual complementarity, with multiple applications in drug design. However, docking programs do not always find correct solutions, either because they are not sampled or due to inaccuracies in the scoring functions. Quantifying the docking performance in real scenarios is essential to understanding their limitations, managing expectations and guiding future developments. Here, we present a fully automated pipeline for pose prediction validated by participating in the Continuous Evaluation of Ligand Pose Prediction (CELPP) Challenge. Acknowledging the intrinsic limitations of the docking method, we devised a strategy to automatically mine and exploit pre-existing data, defining—whenever possible—empirical restraints to guide the docking process. We prove that the pipeline is able to generate predictions for most of the proposed targets as well as obtain poses with low RMSD values when compared to the crystal structure. All things considered, our pipeline highlights some major challenges in the automatic prediction of protein–ligand complexes, which will be addressed in future versions of the pipeline.
Collapse
Affiliation(s)
- Marina Miñarro-Lleonar
- Pharmacy Faculty, University of Barcelona, Av. de Joan XXIII 27-31, 08028 Barcelona, Spain;
| | | | - Daniel Alvarez-Garcia
- GAIN Therapeutics, Parc Cientific de Barcelona, Baldiri i Reixac 10, 08029 Barcelona, Spain;
| | - Peter Schmidtke
- Discngine S.A.S., 79 Avenue Ledru Rollin, 75012 Paris, France;
| | - Xavier Barril
- Pharmacy Faculty, University of Barcelona, Av. de Joan XXIII 27-31, 08028 Barcelona, Spain;
- GAIN Therapeutics, Parc Cientific de Barcelona, Baldiri i Reixac 10, 08029 Barcelona, Spain;
- Catalan Institute for Research and Advanced Studies (ICREA), Passeig de Lluis Companys 23, 08010 Barcelona, Spain
- Correspondence:
| |
Collapse
|
8
|
Ricci-Lopez J, Aguila SA, Gilson MK, Brizuela CA. Improving Structure-Based Virtual Screening with Ensemble Docking and Machine Learning. J Chem Inf Model 2021; 61:5362-5376. [PMID: 34652141 DOI: 10.1021/acs.jcim.1c00511] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
One of the main challenges of structure-based virtual screening (SBVS) is the incorporation of the receptor's flexibility, as its explicit representation in every docking run implies a high computational cost. Therefore, a common alternative to include the receptor's flexibility is the approach known as ensemble docking. Ensemble docking consists of using a set of receptor conformations and performing the docking assays over each of them. However, there is still no agreement on how to combine the ensemble docking results to obtain the final ligand ranking. A common choice is to use consensus strategies to aggregate the ensemble docking scores, but these strategies exhibit slight improvement regarding the single-structure approach. Here, we claim that using machine learning (ML) methodologies over the ensemble docking results could improve the predictive power of SBVS. To test this hypothesis, four proteins were selected as study cases: CDK2, FXa, EGFR, and HSP90. Protein conformational ensembles were built from crystallographic structures, whereas the evaluated compound library comprised up to three benchmarking data sets (DUD, DEKOIS 2.0, and CSAR-2012) and cocrystallized molecules. Ensemble docking results were processed through 30 repetitions of 4-fold cross-validation to train and validate two ML classifiers: logistic regression and gradient boosting trees. Our results indicate that the ML classifiers significantly outperform traditional consensus strategies and even the best performance case achieved with single-structure docking. We provide statistical evidence that supports the effectiveness of ML to improve the ensemble docking performance.
Collapse
Affiliation(s)
- Joel Ricci-Lopez
- Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada, Baja California C.P. 22860, Mexico.,Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México (UNAM), Ensenada, Baja California C.P. 22860, Mexico
| | - Sergio A Aguila
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México (UNAM), Ensenada, Baja California C.P. 22860, Mexico
| | - Michael K Gilson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, San Diego, California 92093, United States
| | - Carlos A Brizuela
- Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada, Baja California C.P. 22860, Mexico
| |
Collapse
|
9
|
Smith RD, Carlson HA. Identification of Cryptic Binding Sites Using MixMD with Standard and Accelerated Molecular Dynamics. J Chem Inf Model 2021; 61:1287-1299. [PMID: 33599485 DOI: 10.1021/acs.jcim.0c01002] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Protein dynamics play an important role in small molecule binding and can pose a significant challenge in the identification of potential binding sites. Cryptic binding sites have been defined as sites which require significant rearrangement of the protein structure to become physically accessible to a ligand. Mixed-solvent MD (MixMD) is a computational protocol which maps the surface of the protein using molecular dynamics (MD) of the unbound protein solvated in a 5% box of probe molecules with explicit water. This method has successfully identified known active and allosteric sites which did not require reorganization. In this study, we apply the MixMD protocol to identify known cryptic sites of 12 proteins characterized by a wide range of conformational changes. Of these 12 proteins, three require reorganization of side chains, five require loop movements, and four require movement of more significant structures such as whole helices. In five cases, we find that standard MixMD simulations are able to map the cryptic binding sites with at least one probe type. In two cases (guanylate kinase and TIE-2), accelerated MD, which increases sampling of torsional angles, was necessary to achieve mapping of portions of the cryptic binding site missed by standard MixMD. For more complex systems where movement of a helix or domain is necessary, MixMD was unable to map the binding site even with accelerated dynamics, possibly due to the limited timescale (100 ns for individual simulations). In general, similar conformational dynamics are observed in water-only simulations and those with probe molecules. This could imply that the probes are not driving opening events but rather take advantage of mapping sites that spontaneously open as part of their inherent conformational behavior. Finally, we show that docking to an ensemble of conformations from the standard MixMD simulations performs better than docking the apo crystal structure in nine cases and even better than half of the bound crystal structures. Poorer performance was seen in docking to ensembles of conformations from the accelerated MixMD simulations.
Collapse
Affiliation(s)
- Richard D Smith
- Department of Medicinal Chemistry, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109-1056, United States
| | - Heather A Carlson
- Department of Medicinal Chemistry, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109-1056, United States
| |
Collapse
|
10
|
Gazgalis D, Zaka M, Abbasi BH, Logothetis DE, Mezei M, Cui M. Protein Binding Pocket Optimization for Virtual High-Throughput Screening (vHTS) Drug Discovery. ACS OMEGA 2020; 5:14297-14307. [PMID: 32596567 PMCID: PMC7315428 DOI: 10.1021/acsomega.0c00522] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/28/2020] [Indexed: 06/11/2023]
Abstract
The virtual high-throughput screening (vHTS) approach has been widely used for large database screening to identify potential lead compounds for drug discovery. Due to its high computational demands, docking that allows receptor flexibility has been a challenging problem for virtual screening. Therefore, the selection of protein target conformations is crucial to produce useful vHTS results. Since only a single protein structure is used to screen large databases in most vHTS studies, the main challenge is to reduce false negative rates in selecting compounds for in vitro tests. False negatives are most likely to occur when using apo structures or homology models of protein targets due to the small volume of the binding pocket formed by incorrect side-chain conformations. Even holo protein structures can exhibit high false negative rates due to ligand-induced fit effects, since the shape of the binding pocket highly depends on its bound ligand. To reduce false negative rates and improve success rates for vHTS in drug discovery, we have developed a new Monte Carlo-based approach that optimizes the binding pocket of protein targets. This newly developed Monte Carlo pocket optimization (MCPO) approach was assessed on several datasets showing promising results. The binding pocket optimization approach could be a useful tool for vHTS-based drug discovery, especially in cases when only apo structures or homology models are available.
Collapse
Affiliation(s)
- Dimitris Gazgalis
- Department
of Pharmaceutical Sciences, Northeastern
University School of Pharmacy, Boston, Massachusetts 02115, United States
| | - Mehreen Zaka
- Department
of Pharmaceutical Sciences, Northeastern
University School of Pharmacy, Boston, Massachusetts 02115, United States
- Department
of Biotechnology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Bilal Haider Abbasi
- Department
of Biotechnology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Diomedes E. Logothetis
- Department
of Pharmaceutical Sciences, Northeastern
University School of Pharmacy, Boston, Massachusetts 02115, United States
| | - Mihaly Mezei
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Meng Cui
- Department
of Pharmaceutical Sciences, Northeastern
University School of Pharmacy, Boston, Massachusetts 02115, United States
| |
Collapse
|
11
|
Chandak T, Mayginnes JP, Mayes H, Wong CF. Using machine learning to improve ensemble docking for drug discovery. Proteins 2020; 88:1263-1270. [PMID: 32401384 DOI: 10.1002/prot.25899] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 04/09/2020] [Accepted: 05/07/2020] [Indexed: 01/26/2023]
Abstract
Ensemble docking has provided an inexpensive method to account for receptor flexibility in molecular docking for virtual screening. Unfortunately, as there is no rigorous theory to connect the docking scores from multiple structures to measured activity, researchers have not yet come up with effective ways to use these scores to classify compounds into actives and inactives. This shortcoming has led to the decrease, rather than an increase in the performance of classifying compounds when more structures are added to the ensemble. Previously, we suggested machine learning, implemented in the form of a naïve Bayesian model could alleviate this problem. However, the naïve Bayesian model assumed that the probabilities of observing the docking scores to different structures to be independent. This approximation might prevent it from achieving even higher performance. In the work presented in this paper, we have relaxed this approximation when using several other machine learning methods-k nearest neighbor, logistic regression, support vector machine, and random forest-to improve ensemble docking. We found significant improvement.
Collapse
Affiliation(s)
- Tanay Chandak
- Department of Chemistry and Biochemistry and Center for Nanoscience, University of Missouri-St. Louis, Saint Louis, Missouri, USA
| | - John P Mayginnes
- Department of Chemistry and Biochemistry and Center for Nanoscience, University of Missouri-St. Louis, Saint Louis, Missouri, USA
| | - Howard Mayes
- Department of Chemistry and Biochemistry and Center for Nanoscience, University of Missouri-St. Louis, Saint Louis, Missouri, USA
| | - Chung F Wong
- Department of Chemistry and Biochemistry and Center for Nanoscience, University of Missouri-St. Louis, Saint Louis, Missouri, USA
| |
Collapse
|
12
|
Vazquez J, Deplano A, Herrero A, Gibert E, Herrero E, Luque FJ. Assessing the Performance of Mixed Strategies To Combine Lipophilic Molecular Similarity and Docking in Virtual Screening. J Chem Inf Model 2020; 60:4231-4245. [PMID: 32364713 DOI: 10.1021/acs.jcim.9b01191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The accuracy of structure-based (SB) virtual screening (VS) is heavily affected by the scoring function used to rank a library of screened compounds. Even in cases where the docked pose agrees with the experimental binding mode of the ligand, the limitations of current scoring functions may lead to sensible inaccuracies in the ability to discriminate between actives and inactives. In this context, the combination of SB and ligand-based (LB) molecular similarity may be a promising strategy to increase the hit rates in VS. This study explores different strategies that aim to exploit the synergy between LB and SB methods in order to mitigate the limitations of these techniques, and to enhance the performance of VS studies by means of a balanced combination between docking scores and three-dimensional (3D) similarity. Particularly, attention is focused to the use of measurements of molecular similarity with PharmScreen, which exploits the 3D distribution of atomic lipophilicity determined from quantum mechanical-based continuum solvation calculations performed with the MST model, in conjunction with three docking programs: Glide, rDock, and GOLD. Different strategies have been explored to combine the information provided by docking and similarity measurements for re-ranking the screened ligands. For a benchmarking of 44 datasets, including 41 targets, the hybrid methods increase the identification of active compounds, according to the early (ROCe%) and total (AUC) enrichment metrics of VS, compared to pure LB and SB methods. Finally, the hybrid approaches are also more effective in enhancing the chemical diversity of active compounds. The datasets employed in this work are available in https://github.com/Pharmacelera/Molecular-Similarity-and-Docking.
Collapse
Affiliation(s)
- Javier Vazquez
- Pharmacelera, Plaça Pau Vila, 1, Sector C 2a, Edificio Palau de Mar, Barcelona 08039, Spain.,Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), and Institute of Theoretical and Computational Chemistry (IQTC-UB), University of Barcelona, Av. Prat de la Riba 171, Santa Coloma de Gramanet E-08921, Spain
| | - Alessandro Deplano
- Pharmacelera, Plaça Pau Vila, 1, Sector C 2a, Edificio Palau de Mar, Barcelona 08039, Spain
| | - Albert Herrero
- Pharmacelera, Plaça Pau Vila, 1, Sector C 2a, Edificio Palau de Mar, Barcelona 08039, Spain
| | - Enric Gibert
- Pharmacelera, Plaça Pau Vila, 1, Sector C 2a, Edificio Palau de Mar, Barcelona 08039, Spain
| | - Enric Herrero
- Pharmacelera, Plaça Pau Vila, 1, Sector C 2a, Edificio Palau de Mar, Barcelona 08039, Spain
| | - F Javier Luque
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), and Institute of Theoretical and Computational Chemistry (IQTC-UB), University of Barcelona, Av. Prat de la Riba 171, Santa Coloma de Gramanet E-08921, Spain
| |
Collapse
|
13
|
Rauf A, Kashif MK, Saeed BA, Al-Masoudi NA, Hameed S. Synthesis, anti-HIV activity, molecular modeling study and QSAR of new designed 2-(2-arylidenehydrazinyl)-4-arylthiazoles. J Mol Struct 2019. [DOI: 10.1016/j.molstruc.2019.07.113] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
14
|
Fan N, Bauer CA, Stork C, de Bruyn Kops C, Kirchmair J. ALADDIN: Docking Approach Augmented by Machine Learning for Protein Structure Selection Yields Superior Virtual Screening Performance. Mol Inform 2019; 39:e1900103. [PMID: 31663691 PMCID: PMC7187304 DOI: 10.1002/minf.201900103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/28/2019] [Indexed: 01/16/2023]
Abstract
Protein flexibility and solvation pose major challenges to docking algorithms and scoring functions. One established strategy for addressing these challenges is to use multiple protein conformations for docking (all-against-all ensemble docking). Recent studies have shown that the performance of ensemble docking can be improved by selecting the most relevant protein structures for docking. In search for a robust approach to protein structure selection, we have come up with an integrated mAchine Learning AnD DockINg approach (ALADDIN). ALADDIN employs a battery of random forest classifiers to select, individually for each compound of interest, from an ensemble of protein structures, the single most suitable protein structure for docking. ALADDIN outperformed the best single-structure docking runs, ensemble docking and a similarity-based docking approach on three out of four investigated targets, with up to 0.15, 0.11 and 0.16 higher area under the receiver operating characteristic curve (AUC) values, respectively. Only in the case of cytochrome P450 3A4, ALADDIN, like any of the other tested approaches, failed to obtain decent performance. ALADDIN can be particularly useful for structure-based virtual screening of malleable proteins, including kinases, some viral enzymes and anti-targets.
Collapse
Affiliation(s)
- Ningning Fan
- Universität Hamburg, Faculty of Mathematics, Informatics and Natural Sciences, Department of Informatics, Center for Bioinformatics, 20146, Hamburg, Germany
| | - Christoph A Bauer
- University of Bergen, Department of Chemistry, N-5020, Bergen, Norway.,University of Bergen, Computational Biology Unit (CBU), N-5020, Bergen, Norway
| | - Conrad Stork
- Universität Hamburg, Faculty of Mathematics, Informatics and Natural Sciences, Department of Informatics, Center for Bioinformatics, 20146, Hamburg, Germany
| | - Christina de Bruyn Kops
- Universität Hamburg, Faculty of Mathematics, Informatics and Natural Sciences, Department of Informatics, Center for Bioinformatics, 20146, Hamburg, Germany
| | - Johannes Kirchmair
- Universität Hamburg, Faculty of Mathematics, Informatics and Natural Sciences, Department of Informatics, Center for Bioinformatics, 20146, Hamburg, Germany.,University of Bergen, Department of Chemistry, N-5020, Bergen, Norway.,University of Bergen, Computational Biology Unit (CBU), N-5020, Bergen, Norway
| |
Collapse
|
15
|
Wong CF. Improving ensemble docking for drug discovery by machine learning. JOURNAL OF THEORETICAL & COMPUTATIONAL CHEMISTRY 2019. [DOI: 10.1142/s0219633619200013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Ensemble docking has provided an inexpensive method to account for receptor flexibility in molecular docking. However, it is still unclear how best to use the docking scores from multiple structures to classify compounds into actives and inactives. Previous studies have also found that the performance of classification could decrease rather than increase with the number of structures included in the ensemble. Machine learning could help to alleviate these problems.
Collapse
Affiliation(s)
- Chung F. Wong
- Department of Chemistry and Biochemistry and Center for Nanoscience, University of Missouri-St. Louis, Saint Louis, MO 63121, USA
| |
Collapse
|
16
|
Matthews N, Kitao A, Laycock S, Hayward S. Haptic-Assisted Interactive Molecular Docking Incorporating Receptor Flexibility. J Chem Inf Model 2019; 59:2900-2912. [DOI: 10.1021/acs.jcim.9b00112] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Nick Matthews
- School of Computing Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - Akio Kitao
- School of Life Science and Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, M6-13, Meguro, Tokyo 152-8550, Japan
| | - Stephen Laycock
- School of Computing Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - Steven Hayward
- School of Computing Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| |
Collapse
|
17
|
Palacio-Rodríguez K, Lans I, Cavasotto CN, Cossio P. Exponential consensus ranking improves the outcome in docking and receptor ensemble docking. Sci Rep 2019; 9:5142. [PMID: 30914702 PMCID: PMC6435795 DOI: 10.1038/s41598-019-41594-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 03/04/2019] [Indexed: 12/21/2022] Open
Abstract
Consensus-scoring methods are commonly used with molecular docking in virtual screening campaigns to filter potential ligands for a protein target. Traditional consensus methods combine results from different docking programs by averaging the score or rank of each molecule obtained from individual programs. Unfortunately, these methods fail if one of the docking programs has poor performance, which is likely to occur due to training-set dependencies and scoring-function parameterization. In this work, we introduce a novel consensus method that overcomes these limitations. We combine the results from individual docking programs using a sum of exponential distributions as a function of the molecule rank for each program. We test the method over several benchmark systems using individual and ensembles of target structures from diverse protein families with challenging decoy/ligand datasets. The results demonstrate that the novel method outperforms the best traditional consensus strategies over a wide range of systems. Moreover, because the novel method is based on the rank rather than the score, it is independent of the score units, scales and offsets, which can hinder the combination of results from different structures or programs. Our method is simple and robust, providing a theoretical basis not only for molecular docking but also for any consensus strategy in general.
Collapse
Affiliation(s)
- Karen Palacio-Rodríguez
- Biophysics of Tropical Diseases Max Planck Tandem Group, University of Antioquia, Medellín, Colombia
| | - Isaias Lans
- Biophysics of Tropical Diseases Max Planck Tandem Group, University of Antioquia, Medellín, Colombia
| | - Claudio N Cavasotto
- Computational Drug Design and Drug Discovery Informatics Laboratory, Translational Medicine Research Institute (IIMT), CONICET-Universidad Austral, Pilar-Derqui, Buenos Aires, Argentina. .,Facultad de Ciencias Biomédicas, Universidad Austral, Pilar-Derqui, Buenos Aires, Argentina. .,Facultad de Ingeniería, Universidad Austral, Pilar-Derqui, Buenos Aires, Argentina.
| | - Pilar Cossio
- Biophysics of Tropical Diseases Max Planck Tandem Group, University of Antioquia, Medellín, Colombia. .,Department of Theoretical Biophysics, Max Planck Institute of Biophysics, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
18
|
Abstract
Computational methods, applied at the early stages of the drug design process, use current technology to provide valuable insights into the understanding of chemical systems in a virtual manner, complementing experimental analysis. Molecular docking is an in silico method employed to foresee binding modes of small compounds or macromolecules in contact with a receptor and to predict their molecular interactions. Moreover, the methodology opens up the possibility of ranking these compounds according to a hierarchy determined using particular scoring functions. Docking protocols assign many approximations, and most of them lack receptor flexibility. Therefore, the reliability of the resulting protein-ligand complexes is uncertain. The association with the costly but more accurate MD techniques provides significant complementary with docking. MD simulations can be used before docking since a series of "new" and broader protein conformations can be extracted from the processing of the resulting trajectory and employed as targets for docking. They also can be utilized a posteriori to optimize the structures of the final complexes from docking, calculate more detailed interaction energies, and provide information about the ligand binding mechanism. Here, we focus on protocols that offer the docking-MD combination as a logical approach to improving the drug discovery process.
Collapse
|
19
|
Ouf SA, Gomha SM, Eweis M, Ouf AS, Sharawy IA. Efficiency of newly prepared thiazole derivatives against some cutaneous fungi. Bioorg Med Chem 2018; 26:3287-3295. [DOI: 10.1016/j.bmc.2018.04.056] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/01/2018] [Accepted: 04/27/2018] [Indexed: 01/25/2023]
|
20
|
Strecker C, Meyer B. Plasticity of the Binding Site of Renin: Optimized Selection of Protein Structures for Ensemble Docking. J Chem Inf Model 2018; 58:1121-1131. [PMID: 29683661 DOI: 10.1021/acs.jcim.8b00010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Protein flexibility poses a major challenge to docking of potential ligands in that the binding site can adopt different shapes. Docking algorithms usually keep the protein rigid and only allow the ligand to be treated as flexible. However, a wrong assessment of the shape of the binding pocket can prevent a ligand from adapting a correct pose. Ensemble docking is a simple yet promising method to solve this problem: Ligands are docked into multiple structures, and the results are subsequently merged. Selection of protein structures is a significant factor for this approach. In this work we perform a comprehensive and comparative study evaluating the impact of structure selection on ensemble docking. We perform ensemble docking with several crystal structures and with structures derived from molecular dynamics simulations of renin, an attractive target for antihypertensive drugs. Here, 500 ns of MD simulations revealed binding site shapes not found in any available crystal structure. We evaluate the importance of structure selection for ensemble docking by comparing binding pose prediction, ability to rank actives above nonactives (screening utility), and scoring accuracy. As a result, for ensemble definition k-means clustering appears to be better suited than hierarchical clustering with average linkage. The best performing ensemble consists of four crystal structures and is able to reproduce the native ligand poses better than any individual crystal structure. Moreover this ensemble outperforms 88% of all individual crystal structures in terms of screening utility as well as scoring accuracy. Similarly, ensembles of MD-derived structures perform on average better than 75% of any individual crystal structure in terms of scoring accuracy at all inspected ensembles sizes.
Collapse
Affiliation(s)
- Claas Strecker
- Department of Chemistry , University of Hamburg , Martin-Luther-King-Platz 6 , 20146 Hamburg , Germany
| | - Bernd Meyer
- Department of Chemistry , University of Hamburg , Martin-Luther-King-Platz 6 , 20146 Hamburg , Germany
| |
Collapse
|
21
|
Ganser LR, Lee J, Rangadurai A, Merriman DK, Kelly ML, Kansal AD, Sathyamoorthy B, Al-Hashimi HM. High-performance virtual screening by targeting a high-resolution RNA dynamic ensemble. Nat Struct Mol Biol 2018; 25:425-434. [PMID: 29728655 PMCID: PMC5942591 DOI: 10.1038/s41594-018-0062-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/27/2018] [Indexed: 12/22/2022]
Abstract
Dynamic ensembles hold great promise in advancing RNA-targeted drug discovery. Here we subjected the transactivation response element (TAR) RNA from human immunodeficiency virus type-1 to experimental high-throughput screening against ~100,000 drug-like small molecules. Results were augmented with 170 known TAR-binding molecules and used to generate sublibraries optimized for evaluating enrichment when virtually screening a dynamic ensemble of TAR determined by combining NMR spectroscopy data and molecular dynamics simulations. Ensemble-based virtual screening scores molecules with an area under the receiver operator characteristic curve of ~0.85-0.94 and with ~40-75% of all hits falling within the top 2% of scored molecules. The enrichment decreased significantly for ensembles generated from the same molecular dynamics simulations without input NMR data and for other control ensembles. The results demonstrate that experimentally determined RNA ensembles can significantly enrich libraries with true hits and that the degree of enrichment is dependent on the accuracy of the ensemble.
Collapse
Affiliation(s)
- Laura R Ganser
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Janghyun Lee
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Atul Rangadurai
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | | | - Megan L Kelly
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Aman D Kansal
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | | | - Hashim M Al-Hashimi
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA.
- Department of Chemistry, Duke University, Durham, NC, USA.
| |
Collapse
|
22
|
Shehu Z, Uzairu A, Sagagi B. Quantitative Structure Activity Relationship (QSAR) and Molecular Docking Study of Some Pyrrolones Antimalarial Agents against Plasmodium Falciparum. JOURNAL OF THE TURKISH CHEMICAL SOCIETY, SECTION A: CHEMISTRY 2018. [DOI: 10.18596/jotcsa.346661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
23
|
Nnadi CI, Jenkins ML, Gentile DR, Bateman LA, Zaidman D, Balius TE, Nomura DK, Burke JE, Shokat KM, London N. Novel K-Ras G12C Switch-II Covalent Binders Destabilize Ras and Accelerate Nucleotide Exchange. J Chem Inf Model 2018; 58:464-471. [PMID: 29320178 DOI: 10.1021/acs.jcim.7b00399] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The success of targeted covalent inhibitors in the global pharmaceutical industry has led to a resurgence of covalent drug discovery. However, covalent inhibitor design for flexible binding sites remains a difficult task due to a lack of methodological development. Here, we compared covalent docking to empirical electrophile screening against the highly dynamic target K-RasG12C. While the overall hit rate of both methods was comparable, we were able to rapidly progress a docking hit to a potent irreversible covalent binder that modifies the inactive, GDP-bound state of K-RasG12C. Hydrogen-deuterium exchange mass spectrometry was used to probe the protein dynamics of compound binding to the switch-II pocket and subsequent destabilization of the nucleotide-binding region. SOS-mediated nucleotide exchange assays showed that, contrary to prior switch-II pocket inhibitors, these new compounds appear to accelerate nucleotide exchange. This study highlights the efficiency of covalent docking as a tool for the discovery of chemically novel hits against challenging targets.
Collapse
Affiliation(s)
- Chimno I Nnadi
- Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco , San Francisco, California 94158, United States
| | - Meredith L Jenkins
- Department of Biochemistry and Microbiology. University of Victoria , Victoria, BC V8W 2Y2, Canada
| | - Daniel R Gentile
- Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco , San Francisco, California 94158, United States
| | - Leslie A Bateman
- Departments of Chemistry, Molecular and Cell Biology, and Nutritional Sciences and Toxicology, University of California, Berkeley , Berkeley, California 94720, United States
| | - Daniel Zaidman
- Department of Organic Chemistry, The Weizmann Institute of Science , Rehovot, 7610001, Israel
| | - Trent E Balius
- Department of Pharmaceutical Chemistry, University of California, San Francisco , San Francisco, California 94158, United States
| | - Daniel K Nomura
- Departments of Chemistry, Molecular and Cell Biology, and Nutritional Sciences and Toxicology, University of California, Berkeley , Berkeley, California 94720, United States
| | - John E Burke
- Department of Biochemistry and Microbiology. University of Victoria , Victoria, BC V8W 2Y2, Canada
| | - Kevan M Shokat
- Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco , San Francisco, California 94158, United States
| | - Nir London
- Department of Organic Chemistry, The Weizmann Institute of Science , Rehovot, 7610001, Israel
| |
Collapse
|
24
|
Chaput L, Mouawad L. Efficient conformational sampling and weak scoring in docking programs? Strategy of the wisdom of crowds. J Cheminform 2017; 9:37. [PMID: 29086077 PMCID: PMC5468358 DOI: 10.1186/s13321-017-0227-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/28/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In drug design, an efficient structure-based optimization of a ligand needs the precise knowledge of the protein-ligand interactions. In the absence of experimental information, docking programs are necessary for ligand positioning, and the choice of a reliable program is essential for the success of such an optimization. The performances of four popular docking programs, Gold, Glide, Surflex and FlexX, were investigated using 100 crystal structures of complexes taken from the Directory of Useful Decoys-Enhanced database. RESULTS The ligand conformational sampling was rather efficient, with a correct pose found for a maximum of 84 complexes, obtained by Surflex. However, the ranking of the correct poses was not as efficient, with a maximum of 68 top-rank or 75 top-4 rank correct poses given by Glidescore. No relationship was found between either the sampling or the scoring performance of the four programs and the properties of either the targets or the small molecules, except for the number of ligand rotatable bonds. As well, no exploitable relationship was found between each program performance in docking and in virtual screening; a wrong top-rank pose may obtain a good score that allows it to be ranked among the most active compounds and vice versa. Also, to improve the results of docking, the strengths of the programs were combined either by using a rescoring procedure or the United Subset Consensus (USC). Oddly, positioning with Surflex and rescoring with Glidescore did not improve the results. However, USC based on docking allowed us to obtain a correct pose in the top-4 rank for 87 complexes. Finally, nine complexes were scrutinized, because a correct pose was found by at least one program but poorly ranked by all four programs. Contrarily to what was expected, except for one case, this was not due to weaknesses of the scoring functions. CONCLUSIONS We conclude that the scoring functions should be improved to detect the correct poses, but sometimes their failure may be due to other varied considerations. To increase the chances of success, we recommend to use several programs and combine their results. Graphical abstract Summary of the results obtained by semi-rigid docking of crystallographic ligands. The docking was done on 100 protein-ligand X-ray structures, taken from the DUD-E database, and using four programs, Glide, Gold, Surflex and FlexX. Based on the docking results, we applied our United Subset Consensus method (USC), for which only the top4-rank poses are relevant. The number of complexes for which the best pose is correct, is represented by the gray boxes, the blue and red boxes correspond to the number of complexes with a correct pose ranked as the top 1 or within the top 4. A pose is considered correct when its root-mean-square deviation from the crystal structure is less than 2 Å.
Collapse
Affiliation(s)
- Ludovic Chaput
- Chemistry, Modelling and Imaging for Biology (CMIB), Institut Curie - PSL Research University, Bât 112, Centre Universitaire, 91405, Orsay Cedex, France.,Paris-Sud University, Orsay, France.,Inserm, U1196, Orsay, France.,CNRS, UMR 9187, Orsay, France.,Selebio SAS, 17 rue de la Barauderie, 77140, Darvault, France
| | - Liliane Mouawad
- Chemistry, Modelling and Imaging for Biology (CMIB), Institut Curie - PSL Research University, Bât 112, Centre Universitaire, 91405, Orsay Cedex, France. .,Paris-Sud University, Orsay, France. .,Inserm, U1196, Orsay, France. .,CNRS, UMR 9187, Orsay, France.
| |
Collapse
|
25
|
Uehara S, Tanaka S. Cosolvent-Based Molecular Dynamics for Ensemble Docking: Practical Method for Generating Druggable Protein Conformations. J Chem Inf Model 2017; 57:742-756. [PMID: 28388074 DOI: 10.1021/acs.jcim.6b00791] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Protein flexibility is a major hurdle in current structure-based virtual screening (VS). In spite of the recent advances in high-performance computing, protein-ligand docking methods still demand tremendous computational cost to take into account the full degree of protein flexibility. In this context, ensemble docking has proven its utility and efficiency for VS studies, but it still needs a rational and efficient method to select and/or generate multiple protein conformations. Molecular dynamics (MD) simulations are useful to produce distinct protein conformations without abundant experimental structures. In this study, we present a novel strategy that makes use of cosolvent-based molecular dynamics (CMD) simulations for ensemble docking. By mixing small organic molecules into a solvent, CMD can stimulate dynamic protein motions and induce partial conformational changes of binding pocket residues appropriate for the binding of diverse ligands. The present method has been applied to six diverse target proteins and assessed by VS experiments using many actives and decoys of DEKOIS 2.0. The simulation results have revealed that the CMD is beneficial for ensemble docking. Utilizing cosolvent simulation allows the generation of druggable protein conformations, improving the VS performance compared with the use of a single experimental structure or ensemble docking by standard MD with pure water as the solvent.
Collapse
Affiliation(s)
- Shota Uehara
- Department of Computational Science, Graduate School of System Informatics, Kobe University , 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Shigenori Tanaka
- Department of Computational Science, Graduate School of System Informatics, Kobe University , 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| |
Collapse
|
26
|
Bohlooli F, Sepehri S, Razzaghi-Asl N. Response surface methodology in drug design: A case study on docking analysis of a potent antifungal fluconazole. Comput Biol Chem 2017; 67:158-173. [DOI: 10.1016/j.compbiolchem.2017.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 11/01/2016] [Accepted: 01/16/2017] [Indexed: 10/20/2022]
|
27
|
Abdulfatai U, Uzairu A, Uba S. Quantitative structure-activity relationship and molecular docking studies of a series of quinazolinonyl analogues as inhibitors of gamma amino butyric acid aminotransferase. J Adv Res 2016; 8:33-43. [PMID: 27942417 PMCID: PMC5137336 DOI: 10.1016/j.jare.2016.10.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/11/2016] [Accepted: 10/15/2016] [Indexed: 11/25/2022] Open
Abstract
Quantitative structure-activity relationship and molecular docking studies were carried out on a series of quinazolinonyl analogues as anticonvulsant inhibitors. Density Functional Theory (DFT) quantum chemical calculation method was used to find the optimized geometry of the anticonvulsants inhibitors. Four types of molecular descriptors were used to derive a quantitative relation between anticonvulsant activity and structural properties. The relevant molecular descriptors were selected by Genetic Function Algorithm (GFA). The best model was validated and found to be statistically significant with squared correlation coefficient (R2) of 0.934, adjusted squared correlation coefficient (R2adj) value of 0.912, Leave one out (LOO) cross validation coefficient (Q2) value of 0.8695 and the external validation (R2pred) of 0.72. Docking analysis revealed that the best compound with the docking scores of −9.5 kcal/mol formed hydrophobic interaction and H-bonding with amino acid residues of gamma aminobutyric acid aminotransferase (GABAAT). This research has shown that the binding affinity generated was found to be better than the commercially sold anti-epilepsy drug, vigabatrin. Also, it was found to be better than the one reported by other researcher. Our QSAR model and molecular docking results corroborate with each other and propose the directions for the design of new inhibitors with better activity against GABAAT. The present study will help in rational drug design and synthesis of new selective GABAAT inhibitors with predetermined affinity and activity and provides valuable information for the understanding of interactions between GABAAT and the anticonvulsants inhibitors.
Collapse
Affiliation(s)
- Usman Abdulfatai
- Department of Chemistry, Ahmadu Bello University, P.M.B. 1044, Zaria, Nigeria
| | - Adamu Uzairu
- Department of Chemistry, Ahmadu Bello University, P.M.B. 1044, Zaria, Nigeria
| | - Sani Uba
- Department of Chemistry, Ahmadu Bello University, P.M.B. 1044, Zaria, Nigeria
| |
Collapse
|
28
|
Docking-undocking combination applied to the D3R Grand Challenge 2015. J Comput Aided Mol Des 2016; 30:805-815. [DOI: 10.1007/s10822-016-9979-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/24/2016] [Indexed: 11/30/2022]
|
29
|
Offutt TL, Swift RV, Amaro RE. Enhancing Virtual Screening Performance of Protein Kinases with Molecular Dynamics Simulations. J Chem Inf Model 2016; 56:1923-1935. [PMID: 27662181 DOI: 10.1021/acs.jcim.6b00261] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In silico virtual screening (VS) is a powerful hit identification technique used in drug discovery projects that aims to effectively distinguish true actives from inactive or decoy molecules. To better capture the dynamic behavior of protein drug targets, compound databases may be screened against an ensemble of protein conformations, which may be experimentally determined or generated computationally, i.e. via molecular dynamics (MD) simulations. Several studies have shown that conformations generated by MD are useful in identifying novel hit compounds, in part because structural rearrangements sampled during MD can provide novel targetable areas. However, it remains difficult to predict a priori when an MD conformation will outperform a VS against the crystal structure alone. Here, we assess whether MD conformations result in improved VS performance for six protein kinases. MD conformations are selected using three different methods, and their VS performances are compared to the corresponding crystal structures. Additionally, these conformations are used to train ensembles, and their VS performance is compared to the individual MD conformations and the corresponding crystal structures using receiver operating characteristic curve (ROC) metrics. We show that performing MD results in at least one conformation that offers better VS performance than the crystal structure, and that, while it is possible to train ensembles to outperform the crystal structure alone, the extent of this enhancement is target dependent. Lastly, we show that the optimal structural selection method is also target dependent and recommend optimizing virtual screens on a kinase-by-kinase basis to improve the likelihood of success.
Collapse
Affiliation(s)
- Tavina L Offutt
- Department of Chemistry and Biochemistry, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92092-0340, United States
| | - Robert V Swift
- Department of Chemistry and Biochemistry, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92092-0340, United States
| | - Rommie E Amaro
- Department of Chemistry and Biochemistry, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92092-0340, United States
| |
Collapse
|
30
|
Blinded predictions of binding modes and energies of HSP90-α ligands for the 2015 D3R grand challenge. Bioorg Med Chem 2016; 24:4890-4899. [DOI: 10.1016/j.bmc.2016.07.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/19/2016] [Accepted: 07/20/2016] [Indexed: 01/14/2023]
|
31
|
Santos-Martins D. Interaction with specific HSP90 residues as a scoring function: validation in the D3R Grand Challenge 2015. J Comput Aided Mol Des 2016; 30:731-742. [PMID: 27549813 DOI: 10.1007/s10822-016-9943-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/17/2016] [Indexed: 01/19/2023]
Abstract
Here is reported the development of a novel scoring function that performs remarkably well at identifying the native binding pose of a subset of HSP90 inhibitors containing aminopyrimidine or resorcinol based scaffolds. This scoring function is called PocketScore, and consists of the interaction energy between a ligand and three residues in the binding pocket: Asp93, Thr184 and a water molecule. We integrated PocketScore into a molecular docking workflow, and used it to participate in the Drug Design Data Resource (D3R) Grand Challenge 2015 (GC2015). PocketScore was able to rank 180 molecules of the GC2015 according to their binding affinity with satisfactory performance. These results indicate that the specific residues considered by PocketScore are determinant to properly model the interaction between HSP90 and its subset of inhibitors containing aminopyrimidine or resorcinol based scaffolds. Moreover, the development of PocketScore aimed at improving docking power while neglecting the prediction of binding affinities, suggesting that accurate identification of native binding poses is a determinant factor for the performance of virtual screens.
Collapse
Affiliation(s)
- Diogo Santos-Martins
- UCIBIO, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, 4169-007, Porto, Portugal.
| |
Collapse
|
32
|
Melvin RL, Salsbury FR. Visualizing ensembles in structural biology. J Mol Graph Model 2016; 67:44-53. [PMID: 27179343 PMCID: PMC5954827 DOI: 10.1016/j.jmgm.2016.05.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/26/2016] [Accepted: 05/02/2016] [Indexed: 10/21/2022]
Abstract
Displaying a single representative conformation of a biopolymer rather than an ensemble of states mistakenly conveys a static nature rather than the actual dynamic personality of biopolymers. However, there are few apparent options due to the fixed nature of print media. Here we suggest a standardized methodology for visually indicating the distribution width, standard deviation and uncertainty of ensembles of states with little loss of the visual simplicity of displaying a single representative conformation. Of particular note is that the visualization method employed clearly distinguishes between isotropic and anisotropic motion of polymer subunits. We also apply this method to ligand binding, suggesting a way to indicate the expected error in many high throughput docking programs when visualizing the structural spread of the output. We provide several examples in the context of nucleic acids and proteins with particular insights gained via this method. Such examples include investigating a therapeutic polymer of FdUMP (5-fluoro-2-deoxyuridine-5-O-monophosphate) - a topoisomerase-1 (Top1), apoptosis-inducing poison - and nucleotide-binding proteins responsible for ATP hydrolysis from Bacillus subtilis. We also discuss how these methods can be extended to any macromolecular data set with an underlying distribution, including experimental data such as NMR structures.
Collapse
Affiliation(s)
- Ryan L Melvin
- Department of Physics, Wake Forest University, NC, United States
| | | |
Collapse
|
33
|
Bietz S, Fährrolfes R, Rarey M. The Art of Compiling Protein Binding Site Ensembles. Mol Inform 2016; 35:593-598. [PMID: 27870245 DOI: 10.1002/minf.201600043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 04/25/2016] [Indexed: 01/24/2023]
Abstract
Structure-based drug design starts with the collection, preparation, and initial analysis of protein structures. With more than 115,000 structures publically available in the Protein Data Bank (PDB), fully automated processes reliably performing these important preprocessing steps are needed. Several tools are available for these tasks, however, most of them do not address the special needs of scientists interested in protein-ligand interactions. In this paper, we summarize our research activities towards an automated processing pipeline from raw PDB data towards ready-to-use protein binding site ensembles. Starting from a single protein structure, the pipeline covers the following phases: Extracting structurally related binding sites from the PDB, aligning disconnected binding site sequences, resolving tautomeric forms and protonation, orienting hydrogens and flippable side-chains, structurally aligning the multitude of binding sites, and performing a reasonable reduction of ensemble structures. The pipeline, named SIENA, creates protein-structural ensembles for the analysis of protein flexibility, molecular design efforts like docking or de novo design within seconds. For the first time, we are able to process the whole PDB in order to create a large collection of protein binding site ensembles. SIENA is available as part of the ZBH ProteinsPlus webserver under http://proteinsplus.zbh.uni-hamburg.de.
Collapse
Affiliation(s)
- Stefan Bietz
- University of Hamburg, ZBH -, Center for Bioinformatics, Bundesstraße 43, 20146, Hamburg, Germany
| | - Rainer Fährrolfes
- University of Hamburg, ZBH -, Center for Bioinformatics, Bundesstraße 43, 20146, Hamburg, Germany
| | - Matthias Rarey
- University of Hamburg, ZBH -, Center for Bioinformatics, Bundesstraße 43, 20146, Hamburg, Germany
| |
Collapse
|
34
|
Bolia A, Ozkan SB. Adaptive BP-Dock: An Induced Fit Docking Approach for Full Receptor Flexibility. J Chem Inf Model 2016; 56:734-46. [PMID: 26971620 DOI: 10.1021/acs.jcim.5b00587] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We present an induced fit docking approach called Adaptive BP-Dock that integrates perturbation response scanning (PRS) with the flexible docking protocol of RosettaLigand in an adaptive manner. We first perturb the binding pocket residues of a receptor and obtain a new conformation based on the residue response fluctuation profile using PRS. Next, we dock a ligand to this new conformation by RosettaLigand, where we repeat these steps for several iterations. We test this approach on several protein test sets including difficult unbound docking cases such as HIV-1 reverse transcriptase and HIV-1 protease. Adaptive BP-Dock results show better correlation with experimental binding affinities compared to other docking protocols. Overall, the results imply that Adaptive BP-Dock can easily capture binding induced conformational changes by simultaneous sampling of protein and ligand conformations. This can provide faster and efficient docking of novel targets for rational drug design.
Collapse
Affiliation(s)
- Ashini Bolia
- Department of Chemistry and Biochemistry, Arizona State University , Tempe, Arizona 85287, United States
| | - S Banu Ozkan
- Department of Physics, Center for Biological Physics, Arizona State University , Tempe, Arizona 85287, United States
| |
Collapse
|
35
|
De Vivo M, Masetti M, Bottegoni G, Cavalli A. Role of Molecular Dynamics and Related Methods in Drug Discovery. J Med Chem 2016; 59:4035-61. [DOI: 10.1021/acs.jmedchem.5b01684] [Citation(s) in RCA: 538] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Marco De Vivo
- Laboratory
of Molecular Modeling and Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
- IAS-5/INM-9 Computational
Biomedicine Forschungszentrum Jülich, Wilhelm-Johnen-Straße, 52428 Jülich, Germany
| | - Matteo Masetti
- Department
of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro
6, I-40126 Bologna, Italy
| | - Giovanni Bottegoni
- CompuNet, Istituto
Italiano di Tecnologia, Via Morego
30, 16163 Genova, Italy
- BiKi Technologies
srl, Via XX Settembre 33/10, 16121 Genova, Italy
| | - Andrea Cavalli
- Department
of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro
6, I-40126 Bologna, Italy
- CompuNet, Istituto
Italiano di Tecnologia, Via Morego
30, 16163 Genova, Italy
| |
Collapse
|
36
|
Bietz S, Rarey M. SIENA: Efficient Compilation of Selective Protein Binding Site Ensembles. J Chem Inf Model 2016; 56:248-59. [PMID: 26759067 DOI: 10.1021/acs.jcim.5b00588] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Structural flexibility of proteins has an important influence on molecular recognition and enzymatic function. In modeling, structure ensembles are therefore often applied as a valuable source of alternative protein conformations. However, their usage is often complicated by structural artifacts and inconsistent data annotation. Here, we present SIENA, a new computational approach for the automated assembly and preprocessing of protein binding site ensembles. Starting with an arbitrarily defined binding site in a single protein structure, SIENA searches for alternative conformations of the same or sequentially closely related binding sites. The method is based on an indexed database for identifying perfect k-mer matches and a recently published algorithm for the alignment of protein binding site conformations. Furthermore, SIENA provides a new algorithm for the interaction-based selection of binding site conformations which aims at covering all known ligand-binding geometries. Various experiments highlight that SIENA is able to generate comprehensive and well selected binding site ensembles improving the compatibility to both known and unconsidered ligand molecules. Starting with the whole PDB as data source, the computation time of the whole ensemble generation takes only a few seconds. SIENA is available via a Web service at www.zbh.uni-hamburg.de/siena .
Collapse
Affiliation(s)
- Stefan Bietz
- Center for Bioinformatics, University of Hamburg , Bundesstrasse 43, 20146 Hamburg, Germany
| | - Matthias Rarey
- Center for Bioinformatics, University of Hamburg , Bundesstrasse 43, 20146 Hamburg, Germany
| |
Collapse
|
37
|
Convertino M, Dokholyan NV. Computational Modeling of Small Molecule Ligand Binding Interactions and Affinities. Methods Mol Biol 2016; 1414:23-32. [PMID: 27094283 DOI: 10.1007/978-1-4939-3569-7_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Understanding and controlling biological phenomena via structure-based drug screening efforts often critically rely on accurate description of protein-ligand interactions. However, most of the currently available computational techniques are affected by severe deficiencies in both protein and ligand conformational sampling as well as in the scoring of the obtained docking solutions. To overcome these limitations, we have recently developed MedusaDock, a novel docking methodology, which simultaneously models ligand and receptor flexibility. Coupled with MedusaScore, a physical force field-based scoring function that accounts for the protein-ligand interaction energy, MedusaDock, has reported the highest success rate in the CSAR 2011 exercise. Here, we present a standard computational protocol to evaluate the binding properties of the two enantiomers of the non-selective β-blocker propanolol in the β2 adrenergic receptor's binding site. We describe details of our protocol, which have been successfully applied to several other targets.
Collapse
Affiliation(s)
- Marino Convertino
- Department of Biochemistry and Biophysics, University of North Carolina, 120 Mason Farm Road, 27599, Chapel Hill, NC, USA
| | - Nikolay V Dokholyan
- Department of Biochemistry and Biophysics, University of North Carolina, 120 Mason Farm Road, 27599, Chapel Hill, NC, USA.
| |
Collapse
|
38
|
Huang Z, Wong CF. Inexpensive Method for Selecting Receptor Structures for Virtual Screening. J Chem Inf Model 2015; 56:21-34. [PMID: 26651874 DOI: 10.1021/acs.jcim.5b00299] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This article introduces a screening performance index (SPI) to help select from a number of experimental structures one or a few that are more likely to identify more actives among its top hits from virtual screening of a compound library. It achieved this by docking only known actives to the experimental structures without considering a large number of decoys to reduce computational costs. The SPI is calculated by using the docking energies of the actives to all the receptor structures. We evaluated the performance of the SPI by applying it to study eight protein systems: fatty acid binding protein adipocyte FABP4, serine/threonine-protein kinase BRAF, beta-1 adrenergic receptor ADRB1, TGF-beta receptor type I TGFR1, adenosylhomocysteinase SAHH, thyroid hormone receptor beta-1 THB, phospholipase A2 group IIA PA2GA, and cytochrome P450 3a4 CP3A4. We found that the SPI agreed with the results from other popular performance metrics such as Boltzmann-Enhanced Discrimination Receiver Operator Characteristics (BEDROC), Robust Initial Enhancement (RIE), Area Under Accumulation Curve (AUAC), and Enrichment Factor (EF) but is less expensive to calculate. SPI also performed better than the best docking energy, the molecular volume of the bound ligand, and the resolution of crystal structure in selecting good receptor structures for virtual screening. The implications of these findings were further discussed in the context of ensemble docking, in situations when no experimental structure for the targeted protein was available, or under circumstances when quick choices of receptor structures need to be made before quantitative indexes such as the SPI and BEDROC can be calculated.
Collapse
Affiliation(s)
- Zunnan Huang
- China-America Cancer Research Institute, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Scientific Research Center, Guangdong Medical University , Dongguan, Guangdong Province, P. R. China , 523808
| | - Chung F Wong
- Department of Chemistry and Biochemistry and Center for Nanoscience, University of Missouri-Saint Louis , One University Boulevard, St. Louis, Missouri 63121, United States
| |
Collapse
|
39
|
Fong P, Tong HHY, Ng KH, Lao CK, Chong CI, Chao CM. In silico prediction of prostaglandin D2 synthase inhibitors from herbal constituents for the treatment of hair loss. JOURNAL OF ETHNOPHARMACOLOGY 2015; 175:470-80. [PMID: 26456343 DOI: 10.1016/j.jep.2015.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 09/16/2015] [Accepted: 10/02/2015] [Indexed: 05/22/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Many herbal topical formulations have been marketed worldwide to prevent hair loss or promote hair growth. Certain in vivo studies have shown promising results among them; however, the effectiveness of their bioactive constituents remains unknown. AIM OF THE STUDY Recently, prostaglandin D2 (PGD2) inhibition has been discovered as a pharmacological mechanism for treating androgenic alopecia (AGA). This present study was aimed to identify prostaglandin D2 synthase (PTGDS) inhibitors in traditional Chinese medicines (TCMs) for treating AGA. MATERIALS AND METHODS In this study, 389 constituents of 12 selected herbs were docked into 6 different crystal structures of PTGDS. The accuracy of the docking methods was successfully validated with experimental data from the ZINC In Man (Zim) database using receiver operating characteristic (ROC) studies. Seven essential drug properties were predicted for topical formulation: skin permeability, sensitisation, irritation, corrosion, mutagenicity, tumorigenicity and reproductive effects. RESULTS Many constituents of the twelve herbs were found to have more advanced binding energies than the experimentally proved PTGDS inhibitors, but many of them were indicative of at least one type of skin adverse reactions, and exhibited poor skin permeability. CONCLUSIONS Overall, ricinoleic acid, acteoside, amentoflavone, quercetin-3-O-rutinoside and hinokiflavone were predicted to be PTGDS inhibitors with good pharmacokinetic properties and minimal adverse skin reactions. These compounds have the highest potential for further in vitro and in vivo investigation with the aim of developing safe and high-efficacy hair loss treatment.
Collapse
Affiliation(s)
- Pedro Fong
- School of Health Sciences, Macao Polytechnic Institute, Macao, China.
| | - Henry H Y Tong
- School of Health Sciences, Macao Polytechnic Institute, Macao, China
| | - Kin H Ng
- School of Health Sciences, Macao Polytechnic Institute, Macao, China
| | - Cheng K Lao
- School of Health Sciences, Macao Polytechnic Institute, Macao, China
| | - Chon I Chong
- School of Health Sciences, Macao Polytechnic Institute, Macao, China
| | - Chi M Chao
- School of Health Sciences, Macao Polytechnic Institute, Macao, China
| |
Collapse
|
40
|
Sampling of conformational ensemble for virtual screening using molecular dynamics simulations and normal mode analysis. Future Med Chem 2015; 7:2317-31. [DOI: 10.4155/fmc.15.150] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aim: Molecular dynamics simulations and normal mode analysis are well-established approaches to generate receptor conformational ensembles (RCEs) for ligand docking and virtual screening. Here, we report new fast molecular dynamics-based and normal mode analysis-based protocols combined with conformational pocket classifications to efficiently generate RCEs. Materials & Methods: We assessed our protocols on two well-characterized protein targets showing local active site flexibility, dihydrofolate reductase and large collective movements, CDK2. The performance of the RCEs was validated by distinguishing known ligands of dihydrofolate reductase and CDK2 among a dataset of diverse chemical decoys. Results & discussion: Our results show that different simulation protocols can be efficient for generation of RCEs depending on different kind of protein flexibility.[Formula: see text]
Collapse
|
41
|
Spyrakis F, Cavasotto CN. Open challenges in structure-based virtual screening: Receptor modeling, target flexibility consideration and active site water molecules description. Arch Biochem Biophys 2015; 583:105-19. [DOI: 10.1016/j.abb.2015.08.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 08/03/2015] [Accepted: 08/03/2015] [Indexed: 01/05/2023]
|
42
|
Antunes DA, Devaurs D, Kavraki LE. Understanding the challenges of protein flexibility in drug design. Expert Opin Drug Discov 2015; 10:1301-13. [DOI: 10.1517/17460441.2015.1094458] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
43
|
Ultrafast protein structure-based virtual screening with Panther. J Comput Aided Mol Des 2015; 29:989-1006. [PMID: 26407559 DOI: 10.1007/s10822-015-9870-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 09/19/2015] [Indexed: 12/31/2022]
Abstract
Molecular docking is by far the most common method used in protein structure-based virtual screening. This paper presents Panther, a novel ultrafast multipurpose docking tool. In Panther, a simple shape-electrostatic model of the ligand-binding area of the protein is created by utilizing the protein crystal structure. The features of the possible ligands are then compared to the model by using a similarity search algorithm. On average, one ligand can be processed in a few minutes by using classical docking methods, whereas using Panther processing takes <1 s. The presented Panther protocol can be used in several applications, such as speeding up the early phases of drug discovery projects, reducing the number of failures in the clinical phase of the drug development process, and estimating the environmental toxicity of chemicals. Panther-code is available in our web pages (http://www.jyu.fi/panther) free of charge after registration.
Collapse
|
44
|
Spyrakis F, Benedetti P, Decherchi S, Rocchia W, Cavalli A, Alcaro S, Ortuso F, Baroni M, Cruciani G. A Pipeline To Enhance Ligand Virtual Screening: Integrating Molecular Dynamics and Fingerprints for Ligand and Proteins. J Chem Inf Model 2015; 55:2256-74. [DOI: 10.1021/acs.jcim.5b00169] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Francesca Spyrakis
- Department of Life
Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Paolo Benedetti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| | - Sergio Decherchi
- CONCEPT Lab, Italian Institute of Technology, via Morego 30, 16163 Genova, Italy
- BiKi Technologies s.r.l., via XX Settembre 33, 16121 Genova, Italy
| | - Walter Rocchia
- CONCEPT Lab, Italian Institute of Technology, via Morego 30, 16163 Genova, Italy
| | - Andrea Cavalli
- CompuNet, Italian Institute of Technology, via Morego 30, 16163 Genova, Italy
- Department of Pharmacy
and Biotechnology, University of Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Stefano Alcaro
- Department of Health Science, University Magna Graecia of Catanzaro, Campus “S Venuta”, Viale Europa 88100, Catanzaro, Italy
| | - Francesco Ortuso
- Department of Health Science, University Magna Graecia of Catanzaro, Campus “S Venuta”, Viale Europa 88100, Catanzaro, Italy
| | - Massimo Baroni
- Molecular Discovery Limited, 215
Marsh Road, Pinner Middlesex, London HA5-5NE, United Kingdom
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| |
Collapse
|
45
|
Hou X, Li K, Yu X, Sun JP, Fang H. Protein Flexibility in Docking-Based Virtual Screening: Discovery of Novel Lymphoid-Specific Tyrosine Phosphatase Inhibitors Using Multiple Crystal Structures. J Chem Inf Model 2015; 55:1973-83. [DOI: 10.1021/acs.jcim.5b00344] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Xuben Hou
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology
of Natural Products (MOE), School of Pharmacy, ‡Department of Physiology, School
of Medicine, and §Key Laboratory Experimental Teratology of the Ministry of Education
and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Kangshuai Li
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology
of Natural Products (MOE), School of Pharmacy, ‡Department of Physiology, School
of Medicine, and §Key Laboratory Experimental Teratology of the Ministry of Education
and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiao Yu
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology
of Natural Products (MOE), School of Pharmacy, ‡Department of Physiology, School
of Medicine, and §Key Laboratory Experimental Teratology of the Ministry of Education
and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jin-peng Sun
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology
of Natural Products (MOE), School of Pharmacy, ‡Department of Physiology, School
of Medicine, and §Key Laboratory Experimental Teratology of the Ministry of Education
and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Hao Fang
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology
of Natural Products (MOE), School of Pharmacy, ‡Department of Physiology, School
of Medicine, and §Key Laboratory Experimental Teratology of the Ministry of Education
and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
46
|
Abstract
INTRODUCTION Molecular docking has become a popular method for virtual screening. Docking small molecules to a rigid biological receptor is fast but could produce many false negatives and identify less diverse compounds. Flexible receptor docking has alleviated this problem. AREAS COVERED This article focuses on reviewing ensemble docking as an approximate but inexpensive method to incorporate receptor flexibility in molecular docking. It outlines key features and recent advances of this method and points out problem areas that need to be addressed to make it even more useful in drug discovery. EXPERT OPINION Among the different methods introduced for flexible receptor docking, ensemble docking represents one of the most popular approaches, especially for high-throughput virtual screening. One can generate structural ensembles by using experimental structures, by structural modeling and by various types of molecular simulations. In building a structural ensemble, a judicious choice of the structures to be included can improve performance. Furthermore, reducing the size of the structural ensemble can cut computational costs, and removing the structures that can bind few ligands well could enrich the number of true actives identified by ensemble docking. The ability of ensemble docking to identify more true positives at the top of a rank-ordered list also depends on the choice of the methods to score and rank compounds, an area that needs further research.
Collapse
Affiliation(s)
- Chung F Wong
- a University of Missouri-St. Louis, Department of Chemistry and Biochemistry , 1 University Boulevard, St. Louis, MO 63121, USA +1 31 4516 5318 ;
| |
Collapse
|
47
|
Nedumpully-Govindan P, Jemec DB, Ding F. CSAR Benchmark of Flexible MedusaDock in Affinity Prediction and Nativelike Binding Pose Selection. J Chem Inf Model 2015; 56:1042-52. [PMID: 26252196 DOI: 10.1021/acs.jcim.5b00303] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
While molecular docking with both ligand and receptor flexibilities can help capture conformational changes upon binding, correct ranking of nativelike binding poses and accurate estimation of binding affinities remains a major challenge. In addition to the commonly used scoring approach with intermolecular interaction energies, we included the contribution of intramolecular energies changes upon binding in our flexible docking method, MedusaDock. In CSAR 2013-2014 binding prediction benchmark exercises, the new scoring function MScomplex was found to better recapitulate experimental binding affinities and correctly identify ligand-binding sequences from decoy receptors. Our further analysis with the DUD data sets indicates significant improvement of virtual screening enrichment using the new scoring function when compared to the previous intermolecular energy based scoring method. Our postanalysis also suggests a new approach to select nativelike poses in the clustering-based pose ranking approach by MedusaDock. Since the calculation of intramolecular energy changes and clustering-based pose ranking and selection are not MedusaDock specific, we expect a broad application in force-field based estimation of binding affinities and pose ranking using flexible ligand-receptor docking.
Collapse
Affiliation(s)
- Praveen Nedumpully-Govindan
- Department of Physics and Astronomy and ‡Department of Genetics and Biochemistry, Clemson University , Clemson, South Carolina 29634, United States
| | - Domen B Jemec
- Department of Physics and Astronomy and ‡Department of Genetics and Biochemistry, Clemson University , Clemson, South Carolina 29634, United States
| | - Feng Ding
- Department of Physics and Astronomy and ‡Department of Genetics and Biochemistry, Clemson University , Clemson, South Carolina 29634, United States
| |
Collapse
|
48
|
Husby J, Bottegoni G, Kufareva I, Abagyan R, Cavalli A. Structure-based predictions of activity cliffs. J Chem Inf Model 2015; 55:1062-76. [PMID: 25918827 DOI: 10.1021/ci500742b] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
In drug discovery, it is generally accepted that neighboring molecules in a given descriptor's space display similar activities. However, even in regions that provide strong predictability, structurally similar molecules can occasionally display large differences in potency. In QSAR jargon, these discontinuities in the activity landscape are known as "activity cliffs". In this study, we assessed the reliability of ligand docking and virtual ligand screening schemes in predicting activity cliffs. We performed our calculations on a diverse, independently collected database of cliff-forming cocrystals. Starting from ideal situations, which allowed us to establish our baseline, we progressively moved toward simulating more realistic scenarios. Ensemble- and template-docking achieved a significant level of accuracy, suggesting that, despite the well-known limitations of empirical scoring schemes, activity cliffs can be accurately predicted by advanced structure-based methods.
Collapse
Affiliation(s)
- Jarmila Husby
- †Department of Drug Discovery and Development-Computation, IIT-Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Giovanni Bottegoni
- †Department of Drug Discovery and Development-Computation, IIT-Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Irina Kufareva
- ‡Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California-San Diego, La Jolla, California 92161, United States
| | - Ruben Abagyan
- ‡Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California-San Diego, La Jolla, California 92161, United States
| | - Andrea Cavalli
- †Department of Drug Discovery and Development-Computation, IIT-Istituto Italiano di Tecnologia, 16163 Genova, Italy.,§Department of Pharmacy and Biotechnology, Università di Bologna, 40126 Bologna, Italy
| |
Collapse
|
49
|
Kooistra AJ, Leurs R, de Esch IJP, de Graaf C. Structure-Based Prediction of G-Protein-Coupled Receptor Ligand Function: A β-Adrenoceptor Case Study. J Chem Inf Model 2015; 55:1045-61. [DOI: 10.1021/acs.jcim.5b00066] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Albert J. Kooistra
- Amsterdam Institute for Molecules,
Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty
of Science, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Rob Leurs
- Amsterdam Institute for Molecules,
Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty
of Science, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Iwan J. P. de Esch
- Amsterdam Institute for Molecules,
Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty
of Science, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Chris de Graaf
- Amsterdam Institute for Molecules,
Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty
of Science, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
50
|
Synthesis, biological evaluation and molecular docking studies of thiazole-based pyrrolidinones and isoindolinediones as anticonvulsant agents. Med Chem Res 2015. [DOI: 10.1007/s00044-015-1371-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|