Rapid and Reliable Binding Affinity Prediction of Bromodomain Inhibitors: A Computational Study
- Shunzhou Wan
- ,
- Agastya P. Bhati
- ,
- Stefan J. Zasada
- ,
- Ian Wall
- ,
- Darren Green
- ,
- Paul Bamborough
- , and
- Peter V. Coveney
Abstract

Binding free energies of bromodomain inhibitors are calculated with recently formulated approaches, namely ESMACS (enhanced sampling of molecular dynamics with approximation of continuum solvent) and TIES (thermodynamic integration with enhanced sampling). A set of compounds is provided by GlaxoSmithKline, which represents a range of chemical functionality and binding affinities. The predicted binding free energies exhibit a good Spearman correlation of 0.78 with the experimental data from the 3-trajectory ESMACS, and an excellent correlation of 0.92 from the TIES approach where applicable. Given access to suitable high end computing resources and a high degree of automation, we can compute individual binding affinities in a few hours with precisions no greater than 0.2 kcal/mol for TIES, and no larger than 0.34 and 1.71 kcal/mol for the 1- and 3-trajectory ESMACS approaches.
1 Introduction
Figure 1

Figure 1. Bromodomain inhibitor I-BET726 and its binding mode in BRD4-BD1. Two views are displayed for the binding mode (PDB ID: 4BJX (15)), in which I-BET726 (16) is represented as stick in cyan/blue/red/green, the protein is shown as cartoon in silver, the crystallographic water molecules are shown as red balls, and clipped protein surfaces are shown in orange.
2 Computational Section
Models


Compounds 1–9 are electrostatically neutral, compounds 10–12 and 16 are positively charged, and compounds 13–15 are negatively charged.
Statistical errors were calculated from repeated IC50 measurements.
There was no activity at the highest concentration (50 μM) tested.
Theoretical Basis



Simulations
3 Results
Reproducibility
Figure 2

Figure 2. Correlation and standard errors of the calculated binding free energies from two independent studies of the BRD4-ligand models performed on BlueWonder2 and ARCHER. (a) Correlation of the predictions, including all rotamers, from 1-traj calculations performed on BlueWonder2 (BW2, horizontal axis) and ARCHER (vertical axis). Solid line, regression of the data using means of the calculated free energies; dotted line, 1:1 ideal regression. (b) the averages and their standard errors from the two separate calculations. One rotamer is used for each ligand.
Choosing Rotamers
Figure 3

Figure 3. Calculated binding free energies from simulations on BlueWonder2 and ARCHER. The ligands are numbered as per Table 1. Circles with red/blue colors are the results based on studies with different rotamers. The circles with crosses are the final results with selected rotamers which are chosen on the basis of the sum of energies Gligand and ΔGbinding (see eq 1). All of the calculated binding free energies are associated with standard errors of less than 1.7 kcal/mol, and are not shown in the figures for reasons of clarity.
Comparison between ESMACS Calculations and Experiments
Figure 4

Figure 4. Spearman ranking correlations of the calculated binding free energies and the experimental data from 1-traj (left panel), 2-traj (center), and 3-traj (right panel) ESMACS approaches. The equations on the subfigures indicate the calculations used in each case. The subscripts (com/rec/lig) and the superscripts (com/lig) in the equations indicate the components (complexes, receptor, and ligands) and the simulations (complexes and free ligands), respectively. The ligands with modifications at the R2-position of the tetrahydroquinoline are marked with crosses; they are all significantly improved in the 2- and 3-trajectory version. The standard errors, which are 0.19–0.34 kcal/mol for the 1-traj and 1.02–1.71 kcal/mol for the 2- and 3-traj approaches, are not shown for reasons of clarity. They are calculated using a bootstrapping method (see Supporting Information). The 2- and 3-traj approaches have similar errors because the energy of the receptor is treated as a constant and hence the uncertainties are dominated by the energies of the complexes.
Figure 5

Figure 5. Improvement of the predictions by inclusion of the adaptation free energies of the receptor and the ligands: (a) the binding free energy changes between the 1-traj (black circles) and 2-traj (magenta circles) indicate the relative adaptation energies of the receptor; those between the 2-traj (magenta circles) and 3-traj (orange circles) show the adaptation energies of the ligands. The adaptation energies can be seen more clearly in panels b as a function of binding affinities, and in panels c for each ligand.
Figure 6

Figure 6. Correlations of free energy components and the experimental data from 3-traj approaches. Both bonded and nonbonded energy terms contribute to the ranking of binding affinities. Their combination (the MMPBSA energy) exhibits a better correlation with experimental data than the components themselves.
Comparison between TIES Calculations and Experiments
Figure 7

Figure 7. Correlations of the calculated binding free energy differences from the TIES study and from experimental measurement. The standard error bars from the TIES calculations are all no greater than 0.2 kcal/mol.
4 Discussion
Figure 8

Figure 8. Calculated vs experimental binding free energies for ligands 1, 8, and 9 which are labeled in the 1-traj subfigure.
Figure 9

Figure 9. Calculated vs experimental binding free energies for ligands 10, 11, and 12 which are labeled in the 1-traj subfigure.
Figure 10

Figure 10. Correlations of the internal energy contributions to the calculated binding free energies and experimental measurement. The internal energy changes are calculated as the differences of the binding free energies between those from the 1-traj and 3-traj approaches: ΔΔGcalc = ΔGbinding3-traj – ΔGbinding1-traj.
5 Conclusions
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jctc.6b00794.
Detailed description of the methods used, additional energetic analyses of the metadynamics, free energy calculation with inclusion of explicit water molecules, alongside the atomic coordinates of the compound-protein complexes and experimental data on compound binding (PDF)
Structures of the studied compounds (ZIP)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
References
This article references 40 other publications.
- 1Lounnas, V.; Ritschel, T.; Kelder, J.; McGuire, R.; Bywater, R. P.; Foloppe, N. Current progress in structure-based rational drug design marks a new mindset in drug discovery Comput. Struct. Biotechnol. J. 2013, 5, e201302011 DOI: 10.5936/csbj.201302011Google ScholarThere is no corresponding record for this reference.
- 2Green, D. V.; Leach, A. R.; Head, M. S. Computer-aided molecular design under the SWOTlight J. Comput.-Aided Mol. Des. 2012, 26 (1) 51– 56 DOI: 10.1007/s10822-011-9514-1Google ScholarThere is no corresponding record for this reference.
- 3Kuhn, B.; Guba, W.; Hert, J.; Banner, D.; Bissantz, C.; Ceccarelli, S.; Haap, W.; Korner, M.; Kuglstatter, A.; Lerner, C.; Mattei, P.; Neidhart, W.; Pinard, E.; Rudolph, M. G.; Schulz-Gasch, T.; Woltering, T.; Stahl, M. A real-world perspective on molecular design J. Med. Chem. 2016, 59 (9) 4087– 4102 DOI: 10.1021/acs.jmedchem.5b01875Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XisVyjtb0%253D&md5=0b81f2a0e9996c5dae44e61e5488ba79A Real-World Perspective on Molecular DesignKuhn, Bernd; Guba, Wolfgang; Hert, Jerome; Banner, David; Bissantz, Caterina; Ceccarelli, Simona; Haap, Wolfgang; Korner, Matthias; Kuglstatter, Andreas; Lerner, Christian; Mattei, Patrizio; Neidhart, Werner; Pinard, Emmanuel; Rudolph, Markus G.; Schulz-Gasch, Tanja; Woltering, Thomas; Stahl, MartinJournal of Medicinal Chemistry (2016), 59 (9), 4087-4102CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We present a series of small mol. drug discovery case studies where computational methods were prospectively employed to impact Roche research projects, with the aim of highlighting those methods that provide real added value. Our brief accounts encompass a broad range of methods and techniques applied to a variety of enzymes and receptors. Most of these are based on judicious application of knowledge about mol. conformations and interactions: filling of lipophilic pockets to gain affinity or selectivity, addn. of polar substituents, scaffold hopping, transfer of SAR, conformation anal., and mol. overlays. A case study of sequence-driven focused screening is presented to illustrate how appropriate preprocessing of information enables effective exploitation of prior knowledge. We conclude that qual. statements enabling chemists to focus on promising regions of chem. space are often more impactful than quant. prediction.
- 4Wang, L.; Wu, Y.; Deng, Y.; Kim, B.; Pierce, L.; Krilov, G.; Lupyan, D.; Robinson, S.; Dahlgren, M. K.; Greenwood, J.; Romero, D. L.; Masse, C.; Knight, J. L.; Steinbrecher, T.; Beuming, T.; Damm, W.; Harder, E.; Sherman, W.; Brewer, M.; Wester, R.; Murcko, M.; Frye, L.; Farid, R.; Lin, T.; Mobley, D. L.; Jorgensen, W. L.; Berne, B. J.; Friesner, R. A.; Abel, R. Accurate and reliable prediction of relative ligand binding potency in prospective drug discovery by way of a modern free-energy calculation protocol and force field J. Am. Chem. Soc. 2015, 137 (7) 2695– 2703 DOI: 10.1021/ja512751qGoogle Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsF2iuro%253D&md5=37a4f4a6c085f47ed531342643b6c33bAccurate and Reliable Prediction of Relative Ligand Binding Potency in Prospective Drug Discovery by Way of a Modern Free-Energy Calculation Protocol and Force FieldWang, Lingle; Wu, Yujie; Deng, Yuqing; Kim, Byungchan; Pierce, Levi; Krilov, Goran; Lupyan, Dmitry; Robinson, Shaughnessy; Dahlgren, Markus K.; Greenwood, Jeremy; Romero, Donna L.; Masse, Craig; Knight, Jennifer L.; Steinbrecher, Thomas; Beuming, Thijs; Damm, Wolfgang; Harder, Ed; Sherman, Woody; Brewer, Mark; Wester, Ron; Murcko, Mark; Frye, Leah; Farid, Ramy; Lin, Teng; Mobley, David L.; Jorgensen, William L.; Berne, Bruce J.; Friesner, Richard A.; Abel, RobertJournal of the American Chemical Society (2015), 137 (7), 2695-2703CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Designing tight-binding ligands is a primary objective of small-mol. drug discovery. Over the past few decades, free-energy calcns. have benefited from improved force fields and sampling algorithms, as well as the advent of low-cost parallel computing. However, it has proven to be challenging to reliably achieve the level of accuracy that would be needed to guide lead optimization (∼5× in binding affinity) for a wide range of ligands and protein targets. Not surprisingly, widespread com. application of free-energy simulations has been limited due to the lack of large-scale validation coupled with the tech. challenges traditionally assocd. with running these types of calcns. Here, we report an approach that achieves an unprecedented level of accuracy across a broad range of target classes and ligands, with retrospective results encompassing 200 ligands and a wide variety of chem. perturbations, many of which involve significant changes in ligand chem. structures. In addn., we have applied the method in prospective drug discovery projects and found a significant improvement in the quality of the compds. synthesized that have been predicted to be potent. Compds. predicted to be potent by this approach have a substantial redn. in false positives relative to compds. synthesized on the basis of other computational or medicinal chem. approaches. Furthermore, the results are consistent with those obtained from our retrospective studies, demonstrating the robustness and broad range of applicability of this approach, which can be used to drive decisions in lead optimization.
- 5Coveney, P. V.; Wan, S. On the calculation of equilibrium thermodynamic properties from molecular dynamics Phys. Chem. Chem. Phys. 2016, 18 (44) 30236– 30240 DOI: 10.1039/C6CP02349EGoogle Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XntVyisr8%253D&md5=f7950af8255cf494a117d46bc1b5191eOn the calculation of equilibrium thermodynamic properties from molecular dynamicsCoveney, Peter V.; Wan, ShunzhouPhysical Chemistry Chemical Physics (2016), 18 (44), 30236-30240CODEN: PPCPFQ; ISSN:1463-9076. (Royal Society of Chemistry)The purpose of statistical mechanics is to provide a route to the calcn. of macroscopic properties of matter from their constituent microscopic components. It is well known that the macrostates emerge as ensemble avs. of microstates. However, this is more often stated than implemented in computer simulation studies. Here we consider foundational aspects of statistical mechanics which are overlooked in most textbooks and research articles that purport to compute macroscopic behavior from microscopic descriptions based on classical mechanics and show how due attention to these issues leads in directions which have not been widely appreciated in the field of mol. dynamics simulation.
- 6Arrowsmith, C. H.; Bountra, C.; Fish, P. V.; Lee, K.; Schapira, M. Epigenetic protein families: a new frontier for drug discovery Nat. Rev. Drug Discovery 2012, 11 (5) 384– 400 DOI: 10.1038/nrd3674Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XlsFWjsbs%253D&md5=eb82034466a43107aa74a18ccf6d29f5Epigenetic protein families: a new frontier for drug discoveryArrowsmith, Cheryl H.; Bountra, Chas; Fish, Paul V.; Lee, Kevin; Schapira, MatthieuNature Reviews Drug Discovery (2012), 11 (5), 384-400CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Epigenetic regulation of gene expression is a dynamic and reversible process that establishes normal cellular phenotypes but also contributes to human diseases. At the mol. level, epigenetic regulation involves hierarchical covalent modification of DNA and the proteins that package DNA, such as histones. Here, we review the key protein families that mediate epigenetic signalling through the acetylation and methylation of histones, including histone deacetylases, protein methyltransferases, lysine demethylases, bromodomain-contg. proteins and proteins that bind to methylated histones. These protein families are emerging as druggable classes of enzymes and druggable classes of protein-protein interaction domains. In this article, we discuss the known links with disease, basic mol. mechanisms of action and recent progress in the pharmacol. modulation of each class of proteins.
- 7Copeland, R. A. Epigenetic medicinal chemistry ACS Med. Chem. Lett. 2016, 7 (2) 124– 127 DOI: 10.1021/acsmedchemlett.5b00462Google ScholarThere is no corresponding record for this reference.
- 8Copeland, R. A.; Olhava, E. J.; Scott, M. P. Targeting epigenetic enzymes for drug discovery Curr. Opin. Chem. Biol. 2010, 14 (4) 505– 510 DOI: 10.1016/j.cbpa.2010.06.174Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXpvVyqtrs%253D&md5=56f501166e98ff60aa6781c76650810dTargeting epigenetic enzymes for drug discoveryCopeland, Robert A.; Olhava, Edward J.; Scott, Margaret PorterCurrent Opinion in Chemical Biology (2010), 14 (4), 505-510CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review. Epigenetic control of gene transcription is the result of enzyme-mediated covalent modifications of promoter-region DNA sites and of histone proteins around which chromosomal DNA is wound. Many of the enzymes that mediate these epigenetic reactions are dysregulated in human diseases. Small mol. inhibitors against two classes of these enzymes have been approved for use in patients: DNA methyltransferase (DNMT) inhibitors and histone deacetylase inhibitors. Other classes of epigenetic enzymes have been demonstrated to have strong disease assocn. and are currently being targeted for small mol. inhibition. In this article we review these enzymes and chem. biol. approaches aimed at discovering small mol. inhibitors against them for therapeutic use.
- 9Theodoulou, N. H.; Tomkinson, N. C.; Prinjha, R. K.; Humphreys, P. G. Clinical progress and pharmacology of small molecule bromodomain inhibitors Curr. Opin. Chem. Biol. 2016, 33, 58– 66 DOI: 10.1016/j.cbpa.2016.05.028Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xps1artb8%253D&md5=4bcd42755b2dd3cebd7fae417e69e83fClinical progress and pharmacology of small molecule bromodomain inhibitorsTheodoulou, Natalie H.; Tomkinson, Nicholas C. O.; Prinjha, Rab K.; Humphreys, Philip G.Current Opinion in Chemical Biology (2016), 33 (), 58-66CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)Bromodomains have emerged as an exciting target class for drug discovery over the past decade. Research has primarily focused on the bromodomain and extra terminal (BET) family of bromodomains, which has led to the development of multiple small mol. inhibitors and an increasing no. of clin. assets. The excitement centered on the clin. potential of BET inhibition has stimulated intense interest in the broader family and the growing no. of non-BET bromodomain chem. probes has facilitated phenotypic investigations, implicating these targets in a variety of disease pathways including cancer, inflammation, embryonic development and neurol. disorders.
- 10Bamborough, P.; Diallo, H.; Goodacre, J. D.; Gordon, L.; Lewis, A.; Seal, J. T.; Wilson, D. M.; Woodrow, M. D.; Chung, C. W. Fragment-based discovery of bromodomain inhibitors part 2: optimization of phenylisoxazole sulfonamides J. Med. Chem. 2012, 55 (2) 587– 596 DOI: 10.1021/jm201283qGoogle ScholarThere is no corresponding record for this reference.
- 11Chung, C. W.; Coste, H.; White, J. H.; Mirguet, O.; Wilde, J.; Gosmini, R. L.; Delves, C.; Magny, S. M.; Woodward, R.; Hughes, S. A.; Boursier, E. V.; Flynn, H.; Bouillot, A. M.; Bamborough, P.; Brusq, J. M.; Gellibert, F. J.; Jones, E. J.; Riou, A. M.; Homes, P.; Martin, S. L.; Uings, I. J.; Toum, J.; Clement, C. A.; Boullay, A. B.; Grimley, R. L.; Blandel, F. M.; Prinjha, R. K.; Lee, K.; Kirilovsky, J.; Nicodeme, E. Discovery and characterization of small molecule inhibitors of the BET family bromodomains J. Med. Chem. 2011, 54 (11) 3827– 3838 DOI: 10.1021/jm200108tGoogle Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmtVWnurs%253D&md5=ebc0f6062321dece877259d775f1c19bDiscovery and characterization of small molecule inhibitors of the BET family bromodomainsChung, Chun-wa; Coste, Herve; White, Julia H.; Mirguet, Olivier; Wilde, Jonathan; Gosmini, Romain L.; Delves, Chris; Magny, Sylvie M.; Woodward, Robert; Hughes, Stephen A.; Boursier, Eric V.; Flynn, Helen; Bouillot, Anne M.; Bamborough, Paul; Brusq, Jean-Marie G.; Gellibert, Francoise J.; Jones, Emma J.; Riou, Alizon M.; Homes, Paul; Martin, Sandrine L.; Uings, Iain J.; Toum, Jerome; Clement, Catherine A.; Boullay, Anne-Benedicte; Grimley, Rachel L.; Blandel, Florence M.; Prinjha, Rab K.; Lee, Kevin; Kirilovsky, Jorge; Nicodeme, EdwigeJournal of Medicinal Chemistry (2011), 54 (11), 3827-3838CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Epigenetic mechanisms of gene regulation have a profound role in normal development and disease processes. An integral part of this mechanism occurs through lysine acetylation of histone tails which are recognized by bromodomains. While the biol. and structural characterization of many bromodomain contg. proteins has advanced considerably, the therapeutic tractability of this protein family is only now becoming understood. This paper describes the discovery and mol. characterization of potent (nM) small mol. inhibitors that disrupt the function of the BET family of bromodomains (Brd2, Brd3, and Brd4). By using a combination of phenotypic screening, chemoproteomics, and biophys. studies, we have discovered that the protein-protein interactions between bromodomains and acetylated histones can be antagonized by selective small mols. that bind at the acetylated lysine recognition pocket. X-ray crystal structures of compds. bound into bromodomains of Brd2 and Brd4 elucidate the mol. interactions of binding and explain the precisely defined stereochem. required for activity.
- 12Chung, C. W.; Dean, A. W.; Woolven, J. M.; Bamborough, P. Fragment-based discovery of bromodomain inhibitors part 1: inhibitor binding modes and implications for lead discovery J. Med. Chem. 2012, 55 (2) 576– 586 DOI: 10.1021/jm201320wGoogle Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsFKktrnN&md5=5b7d7239d0de57266d94beef1ea080aeFragment-Based Discovery of Bromodomain Inhibitors Part 1: Inhibitor Binding Modes and Implications for Lead DiscoveryChung, Chun-wa; Dean, Anthony W.; Woolven, James M.; Bamborough, PaulJournal of Medicinal Chemistry (2012), 55 (2), 576-586CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Bromodomain-contg. proteins are key epigenetic regulators of gene transcription and readers of the histone code. However, the therapeutic benefits of modulating this target class are largely unexplored due to the lack of suitable chem. probes. This article describes the generation of lead mols. for the BET bromodomains through screening a fragment set chosen using structural insights and computational approaches. Anal. of 40 BRD2/fragment x-ray complexes highlights both shared and disparate interaction features that may be exploited for affinity and selectivity. Six representative crystal structures are then exemplified in detail. Two of the fragments are completely new bromodomain chemotypes, and three have never before been crystd. in a bromodomain, so our results significantly extend the limited public knowledge-base of crystallog. small mol./bromodomain interactions. Certain fragments (including paracetamol) bind in a consistent mode to different bromodomains such as CREBBP, suggesting their potential to act as generic bromodomain templates. An important implication is that the bromodomains are not only a phylogenetic family but also a system in which chem. and structural knowledge of one bromodomain gives insights transferable to others.
- 13Sadiq, S. K.; Wright, D.; Watson, S. J.; Zasada, S. J.; Stoica, I.; Coveney, P. V. Automated molecular simulation based binding affinity calculator for ligand-bound HIV-1 proteases J. Chem. Inf. Model. 2008, 48 (9) 1909– 1919 DOI: 10.1021/ci8000937Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtVShtLjN&md5=c616ac9330dc1f4cb9808d68fabc7e4cAutomated Molecular Simulation Based Binding Affinity Calculator for Ligand-Bound HIV-1 ProteasesSadiq, S. Kashif; Wright, David; Watson, Simon J.; Zasada, Stefan J.; Stoica, Ileana; Coveney, Peter V.Journal of Chemical Information and Modeling (2008), 48 (9), 1909-1919CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)The successful application of high throughput mol. simulations to det. biochem. properties would be of great importance to the biomedical community if such simulations could be turned around in a clin. relevant timescale. An important example is the detn. of antiretroviral inhibitor efficacy against varying strains of HIV through calcn. of drug-protein binding affinities. We describe the Binding Affinity Calculator (BAC), a tool for the automated calcn. of HIV-1 protease-ligand binding affinities. The tool employs fully atomistic mol. simulations alongside the well established mol. mechanics Poisson-Boltzmann solvent accessible surface area (MMPBSA) free energy methodol. to enable the calcn. of the binding free energy of several ligand-protease complexes, including all nine FDA approved inhibitors of HIV-1 protease and seven of the natural substrates cleaved by the protease. This enables the efficacy of these inhibitors to be ranked across several mutant strains of the protease relative to the wildtype. BAC is a tool that utilizes the power provided by a computational grid to automate all of the stages required to compute free energies of binding: model prepn., equilibration, simulation, postprocessing, and data-marshaling around the generally widely distributed compute resources utilized. Such automation enables the mol. dynamics methodol. to be used in a high throughput manner not achievable by manual methods. This paper describes the architecture and workflow management of BAC and the function of each of its components. Given adequate compute resources, BAC can yield quant. information regarding drug resistance at the mol. level within 96 h. Such a timescale is of direct clin. relevance and can assist in decision support for the assessment of patient-specific optimal drug treatment and the subsequent response to therapy for any given genotype.
- 14Groen, D.; Bhati, A. P.; Suter, J.; Hetherington, J.; Zasada, S. J.; Coveney, P. V. FabSim: Facilitating computational research through automation on large-scale and distributed e-infrastructures Comput. Phys. Commun. 2016, 207, 375– 385 DOI: 10.1016/j.cpc.2016.05.020Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xptl2nu7s%253D&md5=4b7e50658ec8307f7fa5fcd16747cab6FabSim: Facilitating computational research through automation on large-scale and distributed e-infrastructuresGroen, Derek; Bhati, Agastya P.; Suter, James; Hetherington, James; Zasada, Stefan J.; Coveney, Peter V.Computer Physics Communications (2016), 207 (), 375-385CODEN: CPHCBZ; ISSN:0010-4655. (Elsevier B.V.)We present FabSim, a toolkit developed to simplify a range of computational tasks for researchers in diverse disciplines. FabSim is flexible, adaptable, and allows users to perform a wide range of tasks with ease. It also provides a systematic way to automate the use of resources, including HPC and distributed machines, and to make tasks easier to repeat by recording contextual information. To demonstrate this, we present three use cases where FabSim has enhanced our research productivity. These include simulating cerebrovascular bloodflow, modeling clay-polymer nanocomposites across multiple scales, and calcg. ligand-protein binding affinities.
- 15Wyce, A.; Ganji, G.; Smitheman, K. N.; Chung, C. W.; Korenchuk, S.; Bai, Y.; Barbash, O.; Le, B.; Craggs, P. D.; McCabe, M. T.; Kennedy-Wilson, K. M.; Sanchez, L. V.; Gosmini, R. L.; Parr, N.; McHugh, C. F.; Dhanak, D.; Prinjha, R. K.; Auger, K. R.; Tummino, P. J. BET inhibition silences expression of MYCN and BCL2 and induces cytotoxicity in neuroblastoma tumor models PLoS One 2013, 8 (8) e72967 DOI: 10.1371/journal.pone.0072967Google ScholarThere is no corresponding record for this reference.
- 16Gosmini, R.; Nguyen, V. L.; Toum, J.; Simon, C.; Brusq, J. M.; Krysa, G.; Mirguet, O.; Riou-Eymard, A. M.; Boursier, E. V.; Trottet, L.; Bamborough, P.; Clark, H.; Chung, C. W.; Cutler, L.; Demont, E. H.; Kaur, R.; Lewis, A. J.; Schilling, M. B.; Soden, P. E.; Taylor, S.; Walker, A. L.; Walker, M. D.; Prinjha, R. K.; Nicodeme, E. The discovery of I-BET726 (GSK1324726A), a potent tetrahydroquinoline ApoA1 up-regulator and selective BET bromodomain inhibitor J. Med. Chem. 2014, 57 (19) 8111– 8131 DOI: 10.1021/jm5010539Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1Wlur7J&md5=b1c9c9cae73c41252cf73908d561bd9bThe Discovery of I-BET726 (GSK1324726A), a Potent Tetrahydroquinoline ApoA1 Up-Regulator and Selective BET Bromodomain InhibitorGosmini, Romain; Nguyen, Van Loc; Toum, Jerome; Simon, Christophe; Brusq, Jean-Marie G.; Krysa, Gael; Mirguet, Olivier; Riou-Eymard, Alizon M.; Boursier, Eric V.; Trottet, Lionel; Bamborough, Paul; Clark, Hugh; Chung, Chun-wa; Cutler, Leanne; Demont, Emmanuel H.; Kaur, Rejbinder; Lewis, Antonia J.; Schilling, Mark B.; Soden, Peter E.; Taylor, Simon; Walker, Ann L.; Walker, Matthew D.; Prinjha, Rab K.; Nicodeme, EdwigeJournal of Medicinal Chemistry (2014), 57 (19), 8111-8131CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Through their function as epigenetic readers of the histone code, the BET family of bromodomain-contg. proteins regulate expression of multiple genes of therapeutic relevance, including those involved in tumor cell growth and inflammation. BET bromodomain inhibitors have profound antiproliferative and anti-inflammatory effects which translate into efficacy in oncol. and inflammation models, and the first compds. have now progressed into clin. trials. The exciting biol. of the BETs has led to great interest in the discovery of novel inhibitor classes. Here we describe the identification of a novel tetrahydroquinoline series through up-regulation of apolipoprotein A1 and the optimization into potent compds. active in murine models of septic shock and neuroblastoma. At the mol. level, these effects are produced by inhibition of BET bromodomains. X-ray crystallog. reveals the interactions explaining the structure-activity relationships of binding. The resulting lead mol., I-BET726, represents a new, potent, and selective class of tetrahydroquinoline-based BET inhibitors.
- 17Aldeghi, M.; Heifetz, A.; Bodkin, M. J.; Knapp, S.; Biggin, P. C. Accurate calculation of the absolute free energy of binding for drug molecules Chem. Sci. 2016, 7 (1) 207– 218 DOI: 10.1039/C5SC02678DGoogle Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFGku7%252FI&md5=d00cac3b87f1bd11837d6f292bd8c6e5Accurate calculation of the absolute free energy of binding for drug moleculesAldeghi, Matteo; Heifetz, Alexander; Bodkin, Michael J.; Knapp, Stefan; Biggin, Philip C.Chemical Science (2016), 7 (1), 207-218CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Accurate prediction of binding affinities has been a central goal of computational chem. for decades, yet remains elusive. Despite good progress, the required accuracy for use in a drug-discovery context has not been consistently achieved for drug-like mols. Here, we perform abs. free energy calcns. based on a thermodn. cycle for a set of diverse inhibitors binding to bromodomain-contg. protein 4 (BRD4) and demonstrate that a mean abs. error of 0.6 kcal mol-1 can be achieved. We also show a similar level of accuracy (1.0 kcal mol-1) can be achieved in pseudo prospective approach. Bromodomains are epigenetic mark readers that recognize acetylation motifs and regulate gene transcription, and are currently being investigated as therapeutic targets for cancer and inflammation. The unprecedented accuracy offers the exciting prospect that the binding free energy of drug-like compds. can be predicted for pharmacol. relevant targets.
- 18Lindorff-Larsen, K.; Piana, S.; Palmo, K.; Maragakis, P.; Klepeis, J. L.; Dror, R. O.; Shaw, D. E. Improved side-chain torsion potentials for the Amber ff99SB protein force field Proteins: Struct., Funct., Genet. 2010, 78 (8) 1950– 1958 DOI: 10.1002/prot.22711Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXkvFegtLo%253D&md5=447a9004026e2b93f0f7beff165daa09Improved side-chain torsion potentials for the Amber ff99SB protein force fieldLindorff-Larsen, Kresten; Piana, Stefano; Palmo, Kim; Maragakis, Paul; Klepeis, John L.; Dror, Ron O.; Shaw, David E.Proteins: Structure, Function, and Bioinformatics (2010), 78 (8), 1950-1958CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)Recent advances in hardware and software have enabled increasingly long mol. dynamics (MD) simulations of biomols., exposing certain limitations in the accuracy of the force fields used for such simulations and spurring efforts to refine these force fields. Recent modifications to the Amber and CHARMM protein force fields, for example, have improved the backbone torsion potentials, remedying deficiencies in earlier versions. Here, the authors further advance simulation accuracy by improving the amino acid side-chain torsion potentials of the Amber ff99SB force field. First, the authors used simulations of model alpha-helical systems to identify the four residue types whose rotamer distribution differed the most from expectations based on Protein Data Bank statistics. Second, the authors optimized the side-chain torsion potentials of these residues to match new, high-level quantum-mech. calcns. Finally, the authors used microsecond-timescale MD simulations in explicit solvent to validate the resulting force field against a large set of exptl. NMR measurements that directly probe side-chain conformations. The new force field, which the authors have termed Amber ff99SB-ILDN, exhibits considerably better agreement with the NMR data. Proteins 2010. © 2010 Wiley-Liss, Inc.
- 19Wang, J.; Wolf, R. M.; Caldwell, J. W.; Kollman, P. A.; Case, D. A. Development and testing of a general amber force field J. Comput. Chem. 2004, 25 (9) 1157– 1174 DOI: 10.1002/jcc.20035Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXksFakurc%253D&md5=2992017a8cf51f89290ae2562403b115Development and testing of a general Amber force fieldWang, Junmei; Wolf, Romain M.; Caldwell, James W.; Kollman, Peter A.; Case, David A.Journal of Computational Chemistry (2004), 25 (9), 1157-1174CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)We describe here a general Amber force field (GAFF) for org. mols. GAFF is designed to be compatible with existing Amber force fields for proteins and nucleic acids, and has parameters for most org. and pharmaceutical mols. that are composed of H, C, N, O, S, P, and halogens. It uses a simple functional form and a limited no. of atom types, but incorporates both empirical and heuristic models to est. force consts. and partial at. charges. The performance of GAFF in test cases is encouraging. In test I, 74 crystallog. structures were compared to GAFF minimized structures, with a root-mean-square displacement of 0.26 Å, which is comparable to that of the Tripos 5.2 force field (0.25 Å) and better than those of MMFF 94 and CHARMm (0.47 and 0.44 Å, resp.). In test II, gas phase minimizations were performed on 22 nucleic acid base pairs, and the minimized structures and intermol. energies were compared to MP2/6-31G* results. The RMS of displacements and relative energies were 0.25 Å and 1.2 kcal/mol, resp. These data are comparable to results from Parm99/RESP (0.16 Å and 1.18 kcal/mol, resp.), which were parameterized to these base pairs. Test III looked at the relative energies of 71 conformational pairs that were used in development of the Parm99 force field. The RMS error in relative energies (compared to expt.) is about 0.5 kcal/mol. GAFF can be applied to wide range of mols. in an automatic fashion, making it suitable for rational drug design and database searching.
- 20Frisch, M. J.; Trucks, G. W.; Schlegel, H. B.; Scuseria, G. E.; Robb, M. A.; Cheeseman, J. R.; Montgomery, J. A., Jr.; Vreven, T.; Kudin, K. N.; Burant, J. C.; Millam, J. M.; Iyengar, S. S.; Tomasi, J.; Barone, V.; Mennucci, B.; Cossi, M.; Scalmani, G.; Rega, N.; Petersson, G. A.; Nakatsuji, H.; Hada, M.; Ehara, M.; Toyota, K.; Fukuda, R.; Hasegawa, J.; Ishida, M.; Nakajima, T.; Honda, Y.; Kitao, O.; Nakai, H.; Klene, M.; Li, X.; Knox, J. E.; Hratchian, H. P.; Cross, J. B.; Bakken, V.; Adamo, C.; Jaramillo, J.; Gomperts, R.; Stratmann, R. E.; Yazyev, O.; Austin, A. J.; Cammi, R.; Pomelli, C.; Ochterski, J. W.; Ayala, P. Y.; Morokuma, K.; Voth, G. A.; Salvador, P.; Dannenberg, J. J.; Zakrzewski, V. G.; Dapprich, S.; Daniels, A. D.; Strain, M. C.; Farkas, O.; Malick, D. K.; Rabuck, A. D.; Raghavachari, K.; Foresman, J. B.; Ortiz, J. V.; Cui, Q.; Baboul, A. G.; Clifford, S.; Cioslowski, J.; Stefanov, B. B.; Liu, G.; Liashenko, A.; Piskorz, P.; Komaromi, I.; Martin, R. L.; Fox, D. J.; Keith, T.; Al-Laham, M. A.; Peng, C. Y.; Nanayakkara, A.; Challacombe, M.; Gill, P. M. W.; Johnson, B.; Chen, W.; Wong, M. W.; Gonzalez, C.; Pople, J. A., Gaussian 03; Gaussian, Inc.: Wallingford, CT, 2004.Google ScholarThere is no corresponding record for this reference.
- 21Case, D. A.; Cheatham, T. E., 3rd; Darden, T.; Gohlke, H.; Luo, R.; Merz, K. M., Jr.; Onufriev, A.; Simmerling, C.; Wang, B.; Woods, R. J. The Amber biomolecular simulation programs J. Comput. Chem. 2005, 26 (16) 1668– 1688 DOI: 10.1002/jcc.20290Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1SlsbbM&md5=93be29ff894bab96c783d24e9886c7d0The amber biomolecular simulation programsCase, David A.; Cheatham, Thomas E., III; Darden, Tom; Gohlke, Holger; Luo, Ray; Merz, Kenneth M., Jr.; Onufriev, Alexey; Simmerling, Carlos; Wang, Bing; Woods, Robert J.Journal of Computational Chemistry (2005), 26 (16), 1668-1688CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)The authors describe the development, current features, and some directions for future development of the Amber package of computer programs. This package evolved from a program that was constructed in the late 1970s to do Assisted Model Building with Energy Refinement, and now contains a group of programs embodying a no. of powerful tools of modern computational chem., focused on mol. dynamics and free energy calcns. of proteins, nucleic acids, and carbohydrates.
- 22Laio, A.; Gervasio, F. L. Metadynamics: a method to simulate rare events and reconstruct the free energy in biophysics, chemistry and material science Rep. Prog. Phys. 2008, 71 (12) 126601 DOI: 10.1088/0034-4885/71/12/126601Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFyntrk%253D&md5=cd84cfc103f97c7d7ccf09fc434e2478Metadynamics: a method to stimulate rare events and reconstruct the free energy in biophysics, chemistry and material scienceLaio, Alessandro; Gervasio, Francesco L.Reports on Progress in Physics (2008), 71 (12), 126601/1-126601/22CODEN: RPPHAG; ISSN:0034-4885. (Institute of Physics Publishing)A review. Metadynamics is a powerful algorithm that can be used both for reconstructing the free energy and for accelerating rare events in systems described by complex Hamiltonians, at the classical or at the quantum level. In the algorithm the normal evolution of the system is biased by a history-dependent potential constructed as a sum of Gaussians centered along the trajectory followed by a suitably chosen set of collective variables. The sum of Gaussians is exploited for reconstructing iteratively an estimator of the free energy and forcing the system to escape from local min. This review is intended to provide a comprehensive description of the algorithm, with a focus on the practical aspects that need to be addressed when one attempts to apply metadynamics to a new system: (i) the choice of the appropriate set of collective variables; (ii) the optimal choice of the metadynamics parameters and (iii) how to control the error and ensure convergence of the algorithm.
- 23Lin, Y. L.; Aleksandrov, A.; Simonson, T.; Roux, B. An overview of electrostatic free energy computations for solutions and proteins J. Chem. Theory Comput. 2014, 10 (7) 2690– 2709 DOI: 10.1021/ct500195pGoogle Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXnslaksLc%253D&md5=6aa50d77dc3326ae518f682336b9f4a8An Overview of Electrostatic Free Energy Computations for Solutions and ProteinsLin, Yen-Lin; Aleksandrov, Alexey; Simonson, Thomas; Roux, BenoitJournal of Chemical Theory and Computation (2014), 10 (7), 2690-2709CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)A review. Free energy simulations for electrostatic and charging processes in complex mol. systems encounter specific difficulties owing to the long-range, 1/r Coulomb interaction. To calc. the solvation free energy of a simple ion, it is essential to take into account the polarization of nearby solvent but also the electrostatic potential drop across the liq.-gas boundary, however distant. The latter does not exist in a simulation model based on periodic boundary conditions because there is no phys. boundary to the system. An important consequence is that the ref. value of the electrostatic potential is not an ion in a vacuum. Also, in an infinite system, the electrostatic potential felt by a perturbing charge is conditionally convergent and dependent on the choice of computational conventions. Furthermore, with Ewald lattice summation and tinfoil conducting boundary conditions, the charges experience a spurious shift in the potential that depends on the details of the simulation system such as the vol. fraction occupied by the solvent. All these issues can be handled with established computational protocols, as reviewed here and illustrated for several small ions and three solvated proteins.
- 24Wan, S.; Knapp, B.; Wright, D. W.; Deane, C. M.; Coveney, P. V. Rapid, precise, and reproducible prediction of peptide-MHC binding affinities from molecular dynamics that correlate well with experiment J. Chem. Theory Comput. 2015, 11 (7) 3346– 3356 DOI: 10.1021/acs.jctc.5b00179Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVaksLjN&md5=860aa35a6e8e4013e2b0c7f0ae99170fRapid, precise, and reproducible prediction of peptide-MHC binding affinities from molecular dynamics that correlate well with experimentWan, Shunzhou; Knapp, Bernhard; Wright, David W.; Deane, Charlotte M.; Coveney, Peter V.Journal of Chemical Theory and Computation (2015), 11 (7), 3346-3356CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The presentation of potentially pathogenic peptides by major histocompatibility complex (MHC) mols. is one of the most important processes in adaptive immune defense. Prediction of peptide-MHC (pMHC) binding affinities is therefore a principal objective of theor. immunol. Machine learning techniques achieve good results if substantial exptl. training data are available. Approaches based on structural information become necessary if sufficiently similar training data are unavailable for a specific MHC allele, although they have often been deemed to lack accuracy. In this study, we use a free energy method to rank the binding affinities of 12 diverse peptides bound by a class I MHC mol. HLA-A*02:01. The method is based on enhanced sampling of mol. dynamics calcns. in combination with a continuum solvent approxn. and includes ests. of the configurational entropy based on either a one or a three trajectory protocol. It produces precise and reproducible free energy ests. which correlate well with exptl. measurements. If the results are combined with an amino acid hydrophobicity scale, then an extremely good ranking of peptide binding affinities emerges. Our approach is rapid, robust, and applicable to a wide range of ligand-receptor interactions without further adjustment.
- 25Bhati, A. P.; Wan, S.; Wright, D. W.; Coveney, P. V. Rapid, accurate, precise and reliable relative free energy prediction using ensemble based thermodynamic integration J. Chem. Theory Comput. 2016, DOI: 10.1021/acs.jctc.6b00979Google ScholarThere is no corresponding record for this reference.
- 26Kollman, P. A.; Massova, I.; Reyes, C.; Kuhn, B.; Huo, S.; Chong, L.; Lee, M.; Lee, T.; Duan, Y.; Wang, W.; Donini, O.; Cieplak, P.; Srinivasan, J.; Case, D. A.; Cheatham, T. E., III Calculating structures and free energies of complex molecules: combining molecular mechanics and continuum models Acc. Chem. Res. 2000, 33 (12) 889– 897 DOI: 10.1021/ar000033jGoogle Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXmvFGiu7g%253D&md5=8436ee610ae145894428db1a1deff73cCalculating Structures and Free Energies of Complex Molecules: Combining Molecular Mechanics and Continuum ModelsKollman, Peter A.; Massova, Irina; Reyes, Carolina; Kuhn, Bernd; Huo, Shuanghong; Chong, Lillian; Lee, Matthew; Lee, Taisung; Duan, Yong; Wang, Wei; Donini, Oreola; Cieplak, Piotr; Srinivasan, Jaysharee; Case, David A.; Cheatham, Thomas E., IIIAccounts of Chemical Research (2000), 33 (12), 889-897CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review, with 63 refs. A historical perspective on the application of mol. dynamics (MD) to biol. macromols. is presented. Recent developments combining state-of-the-art force fields with continuum solvation calcns. have allowed us to reach the fourth era of MD applications in which one can often derive both accurate structure and accurate relative free energies from mol. dynamics trajectories. We illustrate such applications on nucleic acid duplexes, RNA hairpins, protein folding trajectories, and protein-ligand, protein-protein, and protein-nucleic acid interactions.
- 27Wright, D. W.; Hall, B. A.; Kenway, O. A.; Jha, S.; Coveney, P. V. Computing Clinically Relevant Binding Free Energies of HIV-1 Protease Inhibitors J. Chem. Theory Comput. 2014, 10 (3) 1228– 1241 DOI: 10.1021/ct4007037Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXht12ltrk%253D&md5=56bc7e7c8f6bbdd694bfc76c20dcec63Computing Clinically Relevant Binding Free Energies of HIV-1 Protease InhibitorsWright, David W.; Hall, Benjamin A.; Kenway, Owain A.; Jha, Shantenu; Coveney, Peter V.Journal of Chemical Theory and Computation (2014), 10 (3), 1228-1241CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The use of mol. simulation to est. the strength of macromol. binding free energies is becoming increasingly widespread, with goals ranging from lead optimization and enrichment in drug discovery to personalizing or stratifying treatment regimes. To realize the potential of such approaches to predict new results, not merely to explain previous exptl. findings, it is necessary that the methods used are reliable and accurate, and that their limitations are thoroughly understood. However, the computational cost of atomistic simulation techniques such as mol. dynamics (MD) has meant that until recently little work has focused on validating and verifying the available free energy methodologies, with the consequence that many of the results published in the literature are not reproducible. Here, we present a detailed anal. of two of the most popular approx. methods for calcg. binding free energies from mol. simulations, mol. mechanics Poisson-Boltzmann surface area (MMPBSA) and mol. mechanics generalized Born surface area (MMGBSA), applied to the nine FDA-approved HIV-1 protease inhibitors. Our results show that the values obtained from replica simulations of the same protease-drug complex, differing only in initially assigned atom velocities, can vary by as much as 10 kcal mol-1, which is greater than the difference between the best and worst binding inhibitors under investigation. Despite this, anal. of ensembles of simulations producing 50 trajectories of 4 ns duration leads to well converged free energy ests. For seven inhibitors, we find that with correctly converged normal mode ests. of the configurational entropy, we can correctly distinguish inhibitors in agreement with exptl. data for both the MMPBSA and MMGBSA methods and thus have the ability to rank the efficacy of binding of this selection of drugs to the protease (no account is made for free energy penalties assocd. with protein distortion leading to the over estn. of the binding strength of the two largest inhibitors ritonavir and atazanavir). We obtain improved rankings and ests. of the relative binding strengths of the drugs by using a novel combination of MMPBSA/MMGBSA with normal mode entropy ests. and the free energy of assocn. calcd. directly from simulation trajectories. Our work provides a thorough assessment of what is required to produce converged and hence reliable free energies for protein-ligand binding.
- 28Genheden, S.; Ryde, U. The MM/PBSA and MM/GBSA methods to estimate ligand-binding affinities Expert Opin. Drug Discovery 2015, 10 (5) 449– 461 DOI: 10.1517/17460441.2015.1032936Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXntFGktr8%253D&md5=b123b88809f275564f95a2271ebd159fThe MM/PBSA and MM/GBSA methods to estimate ligand-binding affinitiesGenheden, Samuel; Ryde, UlfExpert Opinion on Drug Discovery (2015), 10 (5), 449-461CODEN: EODDBX; ISSN:1746-0441. (Informa Healthcare)Introduction: The mol. mechanics energies combined with the Poisson-Boltzmann or generalized Born and surface area continuum solvation (MM/PBSA and MM/GBSA) methods are popular approaches to est. the free energy of the binding of small ligands to biol. macromols. They are typically based on mol. dynamics simulations of the receptor-ligand complex and are therefore intermediate in both accuracy and computational effort between empirical scoring and strict alchem. perturbation methods. They have been applied to a large no. of systems with varying success. Areas covered: The authors review the use of MM/PBSA and MM/GBSA methods to calc. ligand-binding affinities, with an emphasis on calibration, testing and validation, as well as attempts to improve the methods, rather than on specific applications. Expert opinion: MM/PBSA and MM/GBSA are attractive approaches owing to their modular nature and that they do not require calcns. on a training set. They have been used successfully to reproduce and rationalize exptl. findings and to improve the results of virtual screening and docking. However, they contain several crude and questionable approxns., for example, the lack of conformational entropy and information about the no. and free energy of water mols. in the binding site. Moreover, there are many variants of the method and their performance varies strongly with the tested system. Likewise, most attempts to ameliorate the methods with more accurate approaches, for example, quantum-mech. calcns., polarizable force fields or improved solvation have deteriorated the results.
- 29Swanson, J. M.; Henchman, R. H.; McCammon, J. A. Revisiting free energy calculations: a theoretical connection to MM/PBSA and direct calculation of the association free energy Biophys. J. 2004, 86, 67– 74 DOI: 10.1016/S0006-3495(04)74084-9Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXlsV2hug%253D%253D&md5=720b2ec6ef0462edcda34f7be768bc44Revisiting free energy calculations: A theoretical connection to MM/PBSA and direct calculation of the association free energySwanson, Jessica M. J.; Henchman, Richard H.; McCammon, J. AndrewBiophysical Journal (2004), 86 (1, Pt. 1), 67-74CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)The prediction of abs. ligand-receptor binding affinities is essential in a wide range of biophys. queries, from the study of protein-protein interactions to structure-based drug design. End-point free energy methods, such as the Mol. Mechanics Poisson-Boltzmann Surface Area (MM/PBSA) model, have received much attention and widespread application in recent literature. These methods benefit from computational efficiency as only the initial and final states of the system are evaluated, yet there remains a need for strengthening their theor. foundation. Here a clear connection between statistical thermodn. and end-point free energy models is presented. The importance of the assocn. free energy, arising from one mol.'s loss of translational and rotational freedom from the std. state concn., is addressed. A novel method for calcg. this quantity directly from a mol. dynamics simulation is described. The challenges of accounting for changes in the protein conformation and its fluctuations from sep. simulations are discussed. A simple first-order approxn. of the configuration integral is presented to lay the groundwork for future efforts. This model has been applied to FKBP12, a small immunophilin that has been widely studied in the drug industry for its potential immunosuppressive and neuroregenerative effects.
- 30Genheden, S.; Ryde, U. How to obtain statistically converged MM/GBSA results J. Comput. Chem. 2010, 31 (4) 837– 846 DOI: 10.1002/jcc.21366Google ScholarThere is no corresponding record for this reference.
- 31Luo, R.; David, L.; Gilson, M. K. Accelerated Poisson-Boltzmann calculations for static and dynamic systems J. Comput. Chem. 2002, 23 (13) 1244– 1253 DOI: 10.1002/jcc.10120Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XmsVKgsLo%253D&md5=7ce67345061bdd6fcfae91be1650a578Accelerated Poisson-Boltzmann calculations for static and dynamic systemsLuo, Ray; David, Laurent; Gilson, Michael K.Journal of Computational Chemistry (2002), 23 (13), 1244-1253CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)We report an efficient implementation of the finite-difference Poisson-Boltzmann solvent model based on the Modified Incomplete Cholsky Conjugate Gradient algorithm, which gives rather impressive performance for both static and dynamic systems. This is achieved by implementing the algorithm with Eisenstat's two optimizations, utilizing the electrostatic update in simulations, and applying prudent approxns., including: relaxing the convergence criterion, not updating Poisson-Boltzmann-related forces every step, and using electrostatic focusing. It is also possible to markedly accelerate the supporting routines that are used to set up the calcns. and to obtain energies and forces. The resulting finite difference Poisson-Boltzmann method delivers efficiency comparable to the distance-dependent dielec. model for a system tested, HIV Protease, making it a strong candidate for soln.-phase mol. dynamics simulations. Further, the finite difference method includes all intra-solute electrostatic interactions, whereas the distance dependent dielec. calcns. use a 15-Å cutoff. The speed of our numerical finite difference method is comparable to that of the pair-wise Generalized Born approxn. to the Poisson-Boltzmann method.
- 32Beveridge, D. L.; Dicapua, F. M. Free-energy via molecular simulation - applications to chemical and biomolecular systems Annu. Rev. Biophys. Biophys. Chem. 1989, 18, 431– 492 DOI: 10.1146/annurev.bb.18.060189.002243Google ScholarThere is no corresponding record for this reference.
- 33Bunney, T. D.; Wan, S.; Thiyagarajan, N.; Sutto, L.; Williams, S. V.; Ashford, P.; Koss, H.; Knowles, M. A.; Gervasio, F. L.; Coveney, P. V.; Katan, M. The effect of mutations on drug sensitivity and kinase activity of fibroblast growth factor receptors: A combined experimental and theoretical study EBioMedicine 2015, 2 (3) 194– 204 DOI: 10.1016/j.ebiom.2015.02.009Google ScholarThere is no corresponding record for this reference.
- 34Phillips, J. C.; Braun, R.; Wang, W.; Gumbart, J.; Tajkhorshid, E.; Villa, E.; Chipot, C.; Skeel, R. D.; Kale, L.; Schulten, K. Scalable molecular dynamics with NAMD J. Comput. Chem. 2005, 26 (16) 1781– 1802 DOI: 10.1002/jcc.20289Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1SlsbbJ&md5=189051128443b547f4300a1b8fb0e034Scalable molecular dynamics with NAMDPhillips, James C.; Braun, Rosemary; Wang, Wei; Gumbart, James; Tajkhorshid, Emad; Villa, Elizabeth; Chipot, Christophe; Skeel, Robert D.; Kale, Laxmikant; Schulten, KlausJournal of Computational Chemistry (2005), 26 (16), 1781-1802CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)NAMD is a parallel mol. dynamics code designed for high-performance simulation of large biomol. systems. NAMD scales to hundreds of processors on high-end parallel platforms, as well as tens of processors on low-cost commodity clusters, and also runs on individual desktop and laptop computers. NAMD works with AMBER and CHARMM potential functions, parameters, and file formats. This article, directed to novices as well as experts, first introduces concepts and methods used in the NAMD program, describing the classical mol. dynamics force field, equations of motion, and integration methods along with the efficient electrostatics evaluation algorithms employed and temp. and pressure controls used. Features for steering the simulation across barriers and for calcg. both alchem. and conformational free energy differences are presented. The motivations for and a roadmap to the internal design of NAMD, implemented in C++ and based on Charm++ parallel objects, are outlined. The factors affecting the serial and parallel performance of a simulation are discussed. Finally, typical NAMD use is illustrated with representative applications to a small, a medium, and a large biomol. system, highlighting particular features of NAMD, for example, the Tcl scripting language. The article also provides a list of the key features of NAMD and discusses the benefits of combining NAMD with the mol. graphics/sequence anal. software VMD and the grid computing/collab. software BioCoRE. NAMD is distributed free of charge with source code at www.ks.uiuc.edu.
- 35Sadiq, S. K.; Wright, D. W.; Kenway, O. A.; Coveney, P. V. Accurate ensemble molecular dynamics binding free energy ranking of multidrug-resistant HIV-1 proteases J. Chem. Inf. Model. 2010, 50 (5) 890– 905 DOI: 10.1021/ci100007wGoogle Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksFeqtb4%253D&md5=4a9c06d974aad014a9a5e027a08a76acAccurate Ensemble Molecular Dynamics Binding Free Energy Ranking of Multidrug-Resistant HIV-1 ProteasesSadiq, S. Kashif; Wright, David W.; Kenway, Owain A.; Coveney, Peter V.Journal of Chemical Information and Modeling (2010), 50 (5), 890-905CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Accurate calcn. of important thermodn. properties, such as macromol. binding free energies, is one of the principal goals of mol. dynamics simulations. However, single long simulation frequently produces incorrectly converged quant. results due to inadequate sampling of conformational space in a feasible wall-clock time. Multiple short (ensemble) simulations have been shown to explore conformational space more effectively than single long simulations, but the two methods have not yet been thermodynamically compared. Here we show that, for end-state binding free energy detn. methods, ensemble simulations exhibit significantly enhanced thermodn. sampling over single long simulations and result in accurate and converged relative binding free energies that are reproducible to within 0.5 kcal/mol. Completely correct ranking is obtained for six HIV-1 protease variants bound to lopinavir with a correlation coeff. of 0.89 and a mean relative deviation from expt. of 0.9 kcal/mol. Multidrug resistance to lopinavir is enthalpically driven and increases through a decrease in the protein-ligand van der Waals interaction, principally due to the V82A/I84V mutation, and an increase in net electrostatic repulsion due to water-mediated disruption of protein-ligand interactions in the catalytic region. Furthermore, we correctly rank, to within 1 kcal/mol of expt., the substantially increased chem. potency of lopinavir binding to the wild-type protease compared to saquinavir and show that lopinavir takes advantage of a decreased net electrostatic repulsion to confer enhanced binding. Our approach is dependent on the combined use of petascale computing resources and on an automated simulation workflow to attain the required level of sampling and turn around time to obtain the results, which can be as little as three days. This level of performance promotes integration of such methodol. with clin. decision support systems for the optimization of patient-specific therapy.
- 36Norman, G. E.; Stegailov, V. V. Stochastic theory of the classical molecular dynamics method Math. Models Comput. Simul. 2013, 5 (4) 305– 333 DOI: 10.1134/S2070048213040108Google ScholarThere is no corresponding record for this reference.
- 37Chodera, J. D.; Mobley, D. L. Entropy-enthalpy compensation: role and ramifications in biomolecular ligand recognition and design Annu. Rev. Biophys. 2013, 42, 121– 142 DOI: 10.1146/annurev-biophys-083012-130318Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFGrs7bP&md5=52a30a1d0f4f9ae49128e87d2a23e3faEntropy-enthalpy compensation: role and ramifications in biomolecular ligand recognition and designChodera, John D.; Mobley, David L.Annual Review of Biophysics (2013), 42 (), 121-142CODEN: ARBNCV; ISSN:1936-122X. (Annual Reviews)A review. Recent calorimetric studies of interactions between small mols. and biomol. targets have generated renewed interest in the phenomenon of entropy-enthalpy compensation. In these studies, entropic and enthalpic contributions to binding are obsd. to vary substantially and in an opposing manner as the ligand or protein is modified, whereas the binding free energy varies little. In severe examples, engineered enthalpic gains can lead to completely compensating entropic penalties, frustrating ligand design. Here, we examine the evidence for compensation, as well as its potential origins, prevalence, severity, and ramifications for ligand engineering. We find the evidence for severe compensation to be weak in light of the large magnitude of and correlation between errors in exptl. measurements of entropic and enthalpic contributions to binding, though a limited form of compensation may be common. Given the difficulty of predicting or measuring entropic and enthalpic changes to useful precision, or using this information in design, we recommend ligand engineering efforts instead focus on computational and exptl. methodologies to directly assess changes in binding free energy.
- 38Zhu, Y. L.; Beroza, P.; Artis, D. R. Including explicit water molecules as part of the protein structure in MM/PBSA calculations J. Chem. Inf. Model. 2014, 54 (2) 462– 469 DOI: 10.1021/ci4001794Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXitVWnsb7J&md5=285d8818f326df9f889912aaf0313e7dIncluding Explicit Water Molecules as Part of the Protein Structure in MM/PBSA CalculationsZhu, Yong-Liang; Beroza, Paul; Artis, Dean R.Journal of Chemical Information and Modeling (2014), 54 (2), 462-469CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Water is the natural medium of mols. in the cell and plays an important role in protein structure, function and interaction with small mol. ligands. However, the widely used mol. mechanics Poisson-Boltzmann surface area (MM/PBSA) method for binding energy calcn. does not explicitly take account of water mols. that mediate key protein-ligand interactions. We have developed a protocol to include water mols. that mediate ligand-protein interactions as part of the protein structure in calcn. of MM/PBSA binding energies (a method we refer to as water-MM/PBSA) for a series of JNK3 kinase inhibitors. Improved correlation between water-MM/PBSA binding energies and exptl. IC50 values was obtained compared to that obtained from classical MM/PBSA binding energy. This improved correlation was further validated using sets of neuraminidase and avidin inhibitors. The obsd. improvement, however, appears to be limited to systems in which there are water-mediated ligand-protein hydrogen bond interactions. We conclude that the water-MM/PBSA method performs better than classical MM/PBSA in predicting binding affinities when water mols. play a direct role in mediating ligand-protein hydrogen bond interactions.
- 39Rastelli, G.; Del Rio, A.; Degliesposti, G.; Sgobba, M. Fast and accurate predictions of binding free energies using MM-PBSA and MM-GBSA J. Comput. Chem. 2010, 31 (4) 797– 810 DOI: 10.1002/jcc.21372Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtlenuro%253D&md5=786af541a9e19b7d8c61c210c3aa96b5Fast and accurate predictions of binding free energies using MM-PBSA and MM-GBSARastelli, Giulio; Del Rio, Alberto; Degliesposti, Gianluca; Sgobba, MiriamJournal of Computational Chemistry (2010), 31 (4), 797-810CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)In the drug discovery process, accurate methods of computing the affinity of small mols. with a biol. target are strongly needed. This is particularly true for mol. docking and virtual screening methods, which use approximated scoring functions and struggle in estg. binding energies in correlation with exptl. values. Among the various methods, MM-PBSA (mol. mechanics Poisson-Boltzmann surface area) and MM-GBSA (mol. mechanics-generalized Born surface area) are emerging as useful and effective approaches. Although these methods are typically applied to large collections of equilibrated structures of protein-ligand complexes sampled during mol. dynamics in water, the possibility to reliably est. ligand affinity using a single energy-minimized structure and implicit solvation models has not been explored in sufficient detail. Herein, the authors thoroughly investigate this hypothesis by comparing different methods for the generation of protein-ligand complexes and diverse methods for free energy prediction for their ability to correlate with exptl. values. The methods were tested on a series of structurally diverse inhibitors of Plasmodium falciparum DHFR (dihydrofolate reductase) with known binding mode and measured affinities. The results showed that correlations between MM-PBSA or MM-GBSA binding free energies with exptl. affinities were in most cases excellent. Importantly, the authors found that correlations obtained with the use of a single protein-ligand minimized structure and with implicit solvation models were similar to those obtained after averaging over multiple MD snapshots with explicit water mols., with consequent save of computing time without loss of accuracy. When applied to a virtual screening expt., such an approach proved to discriminate between true binders and decoy mols. and yielded significantly better enrichment curves. © 2009 Wiley Periodicals, Inc. J Comput Chem, 31: 797-810, 2010.
- 40Reynolds, C. H.; Holloway, M. K. Thermodynamics of ligand binding and efficiency ACS Med. Chem. Lett. 2011, 2 (6) 433– 437 DOI: 10.1021/ml200010kGoogle Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXjs1Churk%253D&md5=e44c416392ab548a53d96802d030e12bThermodynamics of Ligand Binding and EfficiencyReynolds, Charles H.; Holloway, M. KatharineACS Medicinal Chemistry Letters (2011), 2 (6), 433-437CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Anal. of the exptl. binding thermodn. for approx. 100 protein-ligand complexes provides important insights into the factors governing ligand affinity and efficiency. The commonly accepted correlation between enthalpy and -TΔS is clearly obsd. for this relatively diverse data set. It is also clear that affinity (i.e., ΔG) is not generally correlated to either enthalpy or -TΔS. This is a worrisome trend since the vast majority of computational structure-based design is carried out using interaction energies for one, or at most a few, ligand poses. As such, these energies are most closely comparable to enthalpies not free energies. Closer inspection of the data shows that in a few cases the enthalpy (or -TΔS) is correlated with free energy. It is tempting to speculate that this could be an important consideration as to why some targets are readily amenable to modeling and others are not. Addnl., anal. of the enthalpy and -TΔS efficiencies shows that the trends obsd. for ligand efficiencies with respect to mol. size are primarily a consequence of enthalpic, not entropic, effects.
Cited By
This article is cited by 54 publications.
- Shunzhou Wan, Agastya P. Bhati, Peter V. Coveney. Comparison of Equilibrium and Nonequilibrium Approaches for Relative Binding Free Energy Predictions. Journal of Chemical Theory and Computation 2023, 19
(21)
, 7846-7860. https://doi.org/10.1021/acs.jctc.3c00842
- Agastya P. Bhati, Art Hoti, Andrew Potterton, Mateusz K. Bieniek, Peter V. Coveney. Long Time Scale Ensemble Methods in Molecular Dynamics: Ligand–Protein Interactions and Allostery in SARS-CoV-2 Targets. Journal of Chemical Theory and Computation 2023, 19
(11)
, 3359-3378. https://doi.org/10.1021/acs.jctc.3c00020
- Margarita Stampelou, Anna Suchankova, Efpraxia Tzortzini, Lakshiv Dhingra, Kerry Barkan, Nikolaos Lougiakis, Panagiotis Marakos, Nicole Pouli, Graham Ladds, Antonios Kolocouris. Dual A1/A3 Adenosine Receptor Antagonists: Binding Kinetics and Structure−Activity Relationship Studies Using Mutagenesis and Alchemical Binding Free Energy Calculations. Journal of Medicinal Chemistry 2022, 65
(19)
, 13305-13327. https://doi.org/10.1021/acs.jmedchem.2c01123
- Alexander D. Wade, Agastya P. Bhati, Shunzhou Wan, Peter V. Coveney. Alchemical Free Energy Estimators and Molecular Dynamics Engines: Accuracy, Precision, and Reproducibility. Journal of Chemical Theory and Computation 2022, 18
(6)
, 3972-3987. https://doi.org/10.1021/acs.jctc.2c00114
- Shunzhou Wan, Agastya P. Bhati, David W. Wright, Ian D. Wall, Alan P. Graves, Darren Green, Peter V. Coveney. Ensemble Simulations and Experimental Free Energy Distributions: Evaluation and Characterization of Isoxazole Amides as SMYD3 Inhibitors. Journal of Chemical Information and Modeling 2022, 62
(10)
, 2561-2570. https://doi.org/10.1021/acs.jcim.2c00255
- Agastya P. Bhati, Peter V. Coveney. Large Scale Study of Ligand–Protein Relative Binding Free Energy Calculations: Actionable Predictions from Statistically Robust Protocols. Journal of Chemical Theory and Computation 2022, 18
(4)
, 2687-2702. https://doi.org/10.1021/acs.jctc.1c01288
- Ellen E. Guest, Luis F. Cervantes, Stephen D. Pickett, Charles L. Brooks III, Jonathan D. Hirst. Alchemical Free Energy Methods Applied to Complexes of the First Bromodomain of BRD4. Journal of Chemical Information and Modeling 2022, 62
(6)
, 1458-1470. https://doi.org/10.1021/acs.jcim.1c01229
- Rory M. Crean, Christopher R. Pudney, David K. Cole, Marc W. van der Kamp. Reliable In Silico Ranking of Engineered Therapeutic TCR Binding Affinities with MMPB/GBSA. Journal of Chemical Information and Modeling 2022, 62
(3)
, 577-590. https://doi.org/10.1021/acs.jcim.1c00765
- Maxime Vassaux, Shunzhou Wan, Wouter Edeling, Peter V. Coveney. Ensembles Are Required to Handle Aleatoric and Parametric Uncertainty in Molecular Dynamics Simulation. Journal of Chemical Theory and Computation 2021, 17
(8)
, 5187-5197. https://doi.org/10.1021/acs.jctc.1c00526
- Mateusz K. Bieniek, Agastya P. Bhati, Shunzhou Wan, Peter V. Coveney. TIES 20: Relative Binding Free Energy with a Flexible Superimposition Algorithm and Partial Ring Morphing. Journal of Chemical Theory and Computation 2021, 17
(2)
, 1250-1265. https://doi.org/10.1021/acs.jctc.0c01179
- Alexander D. Wade, David J. Huggins. Identification of Optimal Ligand Growth Vectors Using an Alchemical Free-Energy Method. Journal of Chemical Information and Modeling 2020, 60
(11)
, 5580-5594. https://doi.org/10.1021/acs.jcim.0c00610
- Masakazu Nakadai, Shuta Tomida. Diameter Is a Key 3D Characteristic for Assessments of Efficient Inhibitors of Protein–Protein Interactions. Journal of Chemical Information and Modeling 2020, 60
(10)
, 4785-4790. https://doi.org/10.1021/acs.jcim.0c00607
- Andrew Potterton, Fouad S. Husseini, Michelle W. Y. Southey, Mike J. Bodkin, Alexander Heifetz, Peter V. Coveney, Andrea Townsend-Nicholson. Ensemble-Based Steered Molecular Dynamics Predicts Relative Residence Time of A2A Receptor Binders. Journal of Chemical Theory and Computation 2019, 15
(5)
, 3316-3330. https://doi.org/10.1021/acs.jctc.8b01270
- Agastya
P. Bhati, Shunzhou Wan, Peter V. Coveney. Ensemble-Based Replica Exchange Alchemical Free Energy Methods: The Effect of Protein Mutations on Inhibitor Binding. Journal of Chemical Theory and Computation 2019, 15
(2)
, 1265-1277. https://doi.org/10.1021/acs.jctc.8b01118
- William
M. Menzer, Chen Li, Wenji Sun, Bing Xie, David D. L. Minh. Simple Entropy Terms for End-Point Binding Free Energy Calculations. Journal of Chemical Theory and Computation 2018, 14
(11)
, 6035-6049. https://doi.org/10.1021/acs.jctc.8b00418
- Hannes H. Loeffler, Stefano Bosisio, Guilherme Duarte Ramos Matos, Donghyuk Suh, Benoit Roux, David L. Mobley, Julien Michel. Reproducibility of Free Energy Calculations across Different Molecular Simulation Software Packages. Journal of Chemical Theory and Computation 2018, 14
(11)
, 5567-5582. https://doi.org/10.1021/acs.jctc.8b00544
- Kathryn
A. Giblin, Samantha J. Hughes, Helen Boyd, Pia Hansson, Andreas Bender. Prospectively Validated Proteochemometric Models for the Prediction of Small-Molecule Binding to Bromodomain Proteins. Journal of Chemical Information and Modeling 2018, 58
(9)
, 1870-1888. https://doi.org/10.1021/acs.jcim.8b00400
- Agastya
P. Bhati, Shunzhou Wan, Yuan Hu, Brad Sherborne, Peter V. Coveney. Uncertainty Quantification in Alchemical Free Energy Methods. Journal of Chemical Theory and Computation 2018, 14
(6)
, 2867-2880. https://doi.org/10.1021/acs.jctc.7b01143
- Nojood A. Altwaijry, Michael Baron, David W. Wright, Peter V. Coveney, and Andrea Townsend-Nicholson . An Ensemble-Based Protocol for the Computational Prediction of Helix–Helix Interactions in G Protein-Coupled Receptors using Coarse-Grained Molecular Dynamics. Journal of Chemical Theory and Computation 2017, 13
(5)
, 2254-2270. https://doi.org/10.1021/acs.jctc.6b01246
- Shunzhou Wan, Agastya P. Bhati, Sarah Skerratt, Kiyoyuki Omoto, Veerabahu Shanmugasundaram, Sharan K. Bagal, and Peter V. Coveney . Evaluation and Characterization of Trk Kinase Inhibitors for the Treatment of Pain: Reliable Binding Affinity Predictions from Theory and Computation. Journal of Chemical Information and Modeling 2017, 57
(4)
, 897-909. https://doi.org/10.1021/acs.jcim.6b00780
- Agastya P. Bhati, Shunzhou Wan, David W. Wright, and Peter V. Coveney . Rapid, Accurate, Precise, and Reliable Relative Free Energy Prediction Using Ensemble Based Thermodynamic Integration. Journal of Chemical Theory and Computation 2017, 13
(1)
, 210-222. https://doi.org/10.1021/acs.jctc.6b00979
- Alexander Heifetz. Accelerating COVID-19 Drug Discovery with High-Performance Computing. 2024, 405-411. https://doi.org/10.1007/978-1-0716-3449-3_19
- Katya Ahmad, Abid Javed, Conor Lanphere, Peter V. Coveney, Elena V. Orlova, Stefan Howorka. Structure and dynamics of an archetypal DNA nanoarchitecture revealed via cryo-EM and molecular dynamics simulations. Nature Communications 2023, 14
(1)
https://doi.org/10.1038/s41467-023-38681-5
- Somayeh Zare, Leila Emami, Zahra Faghih, Farshid Zargari, Zeinab Faghih, Soghra Khabnadideh. Design, synthesis, computational study and cytotoxic evaluation of some new quinazoline derivatives containing pyrimidine moiety. Scientific Reports 2023, 13
(1)
https://doi.org/10.1038/s41598-023-41530-6
- Süleyman Selim Çınaroğlu, Philip C. Biggin. The role of loop dynamics in the prediction of ligand–protein binding enthalpy. Chemical Science 2023, 14
(24)
, 6792-6805. https://doi.org/10.1039/D2SC06471E
- Arnaldo F. Silva, Ellen E. Guest, Bruno N. Falcone, Stephen D. Pickett, David M. Rogers, Jonathan D. Hirst. Free energy perturbation calculations of tetrahydroquinolines complexed to the first bromodomain of BRD4. Molecular Physics 2023, 121
(9-10)
https://doi.org/10.1080/00268976.2022.2124201
- Darren V. S. Green, Colin M. Edge. Useful Computational Chemistry Tools for Medicinal Chemistry. 2023, 94-136. https://doi.org/10.1039/9781788018982-00094
- Shunzhou Wan, Agastya P. Bhati, David W. Wright, Alexander D. Wade, Gary Tresadern, Herman van Vlijmen, Peter V. Coveney. The performance of ensemble-based free energy protocols in computing binding affinities to ROS1 kinase. Scientific Reports 2022, 12
(1)
https://doi.org/10.1038/s41598-022-13319-6
- Manas Bandyopadhyay, Utsav Sengupta, Muthaimanoj Periyasamy, Sudipta Mukhopadhyay, Avantika Hasija, Deepak Chopra, Namık Özdemir, Musa A. Said, Mrinal K. Bera. Cu(II)(PhOMe-Salophen) Complex: Greener Pasture Biological Study, XRD/HAS Interactions, and MEP. Russian Journal of Inorganic Chemistry 2022, 67
(S2)
, S114-S127. https://doi.org/10.1134/S0036023623700274
- Shunzhou Wan, Agastya P. Bhati, Alexander D. Wade, Dario Alfè, Peter V. Coveney. Thermodynamic and structural insights into the repurposing of drugs that bind to SARS-CoV-2 main protease. Molecular Systems Design & Engineering 2022, 7
(2)
, 123-131. https://doi.org/10.1039/D1ME00124H
- Agastya P. Bhati, Shunzhou Wan, Dario Alfè, Austin R. Clyde, Mathis Bode, Li Tan, Mikhail Titov, Andre Merzky, Matteo Turilli, Shantenu Jha, Roger R. Highfield, Walter Rocchia, Nicola Scafuri, Sauro Succi, Dieter Kranzlmüller, Gerald Mathias, David Wifling, Yann Donon, Alberto Di Meglio, Sofia Vallecorsa, Heng Ma, Anda Trifan, Arvind Ramanathan, Tom Brettin, Alexander Partin, Fangfang Xia, Xiaotan Duan, Rick Stevens, Peter V. Coveney. Pandemic drugs at pandemic speed: infrastructure for accelerating COVID-19 drug discovery with hybrid machine learning- and physics-based simulations on high-performance computers. Interface Focus 2021, 11
(6)
https://doi.org/10.1098/rsfs.2021.0018
- Shunzhou Wan, Deepak Kumar, Valentin Ilyin, Ussama Al Homsi, Gulab Sher, Alexander Knuth, Peter V. Coveney. The effect of protein mutations on drug binding suggests ensuing personalised drug selection. Scientific Reports 2021, 11
(1)
https://doi.org/10.1038/s41598-021-92785-w
- Dawei Zhang, Rui Duan. Understanding the avidin–biotin binding based on polarized protein-specific charge. Physical Chemistry Chemical Physics 2021, 23
(38)
, 21951-21958. https://doi.org/10.1039/D1CP02752B
- Qianqian Wang, Xiaomin Shao, Elaine Lai Han Leung, Yingqing Chen, Xiaojun Yao. Selectively targeting individual bromodomain: Drug discovery and molecular mechanisms. Pharmacological Research 2021, 172 , 105804. https://doi.org/10.1016/j.phrs.2021.105804
- Aymen Al Saadi, Dario Alfe, Yadu Babuji, Agastya Bhati, Ben Blaiszik, Alexander Brace, Thomas Brettin, Kyle Chard, Ryan Chard, Austin Clyde, Peter Coveney, Ian Foster, Tom Gibbs, Shantenu Jha, Kristopher Keipert, Dieter Kranzlmüller, Thorsten Kurth, Hyungro Lee, Zhuozhao Li, Heng Ma, Gerald Mathias, Andre Merzky, Alexander Partin, Arvind Ramanathan, Ashka Shah, Abraham Stern, Rick Stevens, Li Tan, Mikhail Titov, Anda Trifan, Aristeidis Tsaris, Matteo Turilli, Huub Van Dam, Shunzhou Wan, David Wifling, Junqi Yin. IMPECCABLE: Integrated Modeling PipelinE for COVID Cure by Assessing Better LEads. 2021, 1-12. https://doi.org/10.1145/3472456.3473524
- Hyungro Lee, Andre Merzky, Li Tan, Mikhail Titov, Matteo Turilli, Dario Alfe, Agastya Bhati, Alex Brace, Austin Clyde, Peter Coveney, Heng Ma, Arvind Ramanathan, Rick Stevens, Anda Trifan, Hubertus Van Dam, Shunzhou Wan, Sean Wilkinson, Shantenu Jha. Scalable HPC & AI infrastructure for COVID-19 therapeutics. 2021, 1-13. https://doi.org/10.1145/3468267.3470573
- Alya A Arabi. Artificial intelligence in drug design: algorithms, applications, challenges and ethics. Future Drug Discovery 2021, 3
(2)
https://doi.org/10.4155/fdd-2020-0028
- Shunzhou Wan, Robert C. Sinclair, Peter V. Coveney. Uncertainty quantification in classical molecular dynamics. Philosophical Transactions of the Royal Society A: Mathematical, Physical and Engineering Sciences 2021, 379
(2197)
https://doi.org/10.1098/rsta.2020.0082
- Shunzhou Wan, Andrew Potterton, Fouad S. Husseini, David W. Wright, Alexander Heifetz, Maciej Malawski, Andrea Townsend-Nicholson, Peter V. Coveney. Hit-to-lead and lead optimization binding free energy calculations for G protein-coupled receptors. Interface Focus 2020, 10
(6)
, 20190128. https://doi.org/10.1098/rsfs.2019.0128
- S. J. Zasada, D. W. Wright, P. V. Coveney. Large-scale binding affinity calculations on commodity compute clouds. Interface Focus 2020, 10
(6)
, 20190133. https://doi.org/10.1098/rsfs.2019.0133
- Shunzhou Wan, Agastya P. Bhati, Stefan J. Zasada, Peter V. Coveney. Rapid, accurate, precise and reproducible ligand–protein binding free energy prediction. Interface Focus 2020, 10
(6)
, 20200007. https://doi.org/10.1098/rsfs.2020.0007
- William M. Menzer, Bing Xie, David D. L. Minh. On Restraints in End‐Point Protein–Ligand Binding Free Energy Calculations. Journal of Computational Chemistry 2020, 41
(6)
, 573-586. https://doi.org/10.1002/jcc.26119
- David W. Wright, Fouad Husseini, Shunzhou Wan, Christophe Meyer, Herman van Vlijmen, Gary Tresadern, Peter V. Coveney. Application of the ESMACS Binding Free Energy Protocol to a Multi‐Binding Site Lactate Dehydogenase A Ligand Dataset. Advanced Theory and Simulations 2020, 3
(1)
https://doi.org/10.1002/adts.201900194
- Shunzhou Wan, Gary Tresadern, Laura Pérez‐Benito, Herman van Vlijmen, Peter V. Coveney. Accuracy and Precision of Alchemical Relative Free Energy Predictions with and without Replica‐Exchange. Advanced Theory and Simulations 2020, 3
(1)
https://doi.org/10.1002/adts.201900195
- Derek Groen, Robin A. Richardson, David W. Wright, Vytautas Jancauskas, Robert Sinclair, Paul Karlshoefer, Maxime Vassaux, Hamid Arabnejad, Tomasz Piontek, Piotr Kopta, Bartosz Bosak, Jalal Lakhlili, Olivier Hoenen, Diana Suleimenova, Wouter Edeling, Daan Crommelin, Anna Nikishova, Peter V. Coveney. Introducing VECMAtk - Verification, Validation and Uncertainty Quantification for Multiscale and HPC Simulations. 2019, 479-492. https://doi.org/10.1007/978-3-030-22747-0_36
- Vassilios Myrianthopoulos, Emmanuel Mikros. From bench to bedside, via desktop. Recent advances in the application of cutting-edge in silico tools in the research of drugs targeting bromodomain modules. Biochemical Pharmacology 2019, 159 , 40-51. https://doi.org/10.1016/j.bcp.2018.11.007
- Laura Pérez-Benito, Henrik Keränen, Herman van Vlijmen, Gary Tresadern. Predicting Binding Free Energies of PDE2 Inhibitors. The Difficulties of Protein Conformation. Scientific Reports 2018, 8
(1)
https://doi.org/10.1038/s41598-018-23039-5
- Jumana Dakka, Matteo Turilli, David W. Wright, Stefan J. Zasada, Vivek Balasubramanian, Shunzhou Wan, Peter V. Coveney, Shantenu Jha. High-throughput binding affinity calculations at extreme scales. BMC Bioinformatics 2018, 19
(S18)
https://doi.org/10.1186/s12859-018-2506-6
- Robert Abel, Eric S Manas, Richard A Friesner, Ramy S Farid, Lingle Wang. Modeling the value of predictive affinity scoring in preclinical drug discovery. Current Opinion in Structural Biology 2018, 52 , 103-110. https://doi.org/10.1016/j.sbi.2018.09.002
- Jumana Dakka, Kristof Farkas-Pall, Matteo Turilli, David W. Wright, Peter V. Coveney, Shantenu Jha. Concurrent and Adaptive Extreme Scale Binding Free Energy Calculations. 2018, 189-200. https://doi.org/10.1109/eScience.2018.00034
- Jumana Dakka, Kristof Farkas-Pall, Vivek Balasubramanian, Matteo Turilli, Shunzhou Wan, David W. Wright, Stefan Zasada, Peter V. Coveney, Shantenu Jha. Enabling Trade-offs Between Accuracy and Computational Cost: Adaptive Algorithms to Reduce Time to Clinical Insight. 2018, 572-577. https://doi.org/10.1109/CCGRID.2018.00005
- Trung Hai Nguyen, Huan‐Xiang Zhou, David D. L. Minh. Using the fast fourier transform in binding free energy calculations. Journal of Computational Chemistry 2018, 39
(11)
, 621-636. https://doi.org/10.1002/jcc.25139
- Trung Hai Nguyen, David D. L. Minh. Implicit ligand theory for relative binding free energies. The Journal of Chemical Physics 2018, 148
(10)
https://doi.org/10.1063/1.5017136
- R. Charlotte Eccleston, Shunzhou Wan, Neil Dalchau, Peter V. Coveney. The Role of Multiscale Protein Dynamics in Antigen Presentation and T Lymphocyte Recognition. Frontiers in Immunology 2017, 8 https://doi.org/10.3389/fimmu.2017.00797
Abstract
Figure 1
Figure 1. Bromodomain inhibitor I-BET726 and its binding mode in BRD4-BD1. Two views are displayed for the binding mode (PDB ID: 4BJX (15)), in which I-BET726 (16) is represented as stick in cyan/blue/red/green, the protein is shown as cartoon in silver, the crystallographic water molecules are shown as red balls, and clipped protein surfaces are shown in orange.
Figure 2
Figure 2. Correlation and standard errors of the calculated binding free energies from two independent studies of the BRD4-ligand models performed on BlueWonder2 and ARCHER. (a) Correlation of the predictions, including all rotamers, from 1-traj calculations performed on BlueWonder2 (BW2, horizontal axis) and ARCHER (vertical axis). Solid line, regression of the data using means of the calculated free energies; dotted line, 1:1 ideal regression. (b) the averages and their standard errors from the two separate calculations. One rotamer is used for each ligand.
Figure 3
Figure 3. Calculated binding free energies from simulations on BlueWonder2 and ARCHER. The ligands are numbered as per Table 1. Circles with red/blue colors are the results based on studies with different rotamers. The circles with crosses are the final results with selected rotamers which are chosen on the basis of the sum of energies Gligand and ΔGbinding (see eq 1). All of the calculated binding free energies are associated with standard errors of less than 1.7 kcal/mol, and are not shown in the figures for reasons of clarity.
Figure 4
Figure 4. Spearman ranking correlations of the calculated binding free energies and the experimental data from 1-traj (left panel), 2-traj (center), and 3-traj (right panel) ESMACS approaches. The equations on the subfigures indicate the calculations used in each case. The subscripts (com/rec/lig) and the superscripts (com/lig) in the equations indicate the components (complexes, receptor, and ligands) and the simulations (complexes and free ligands), respectively. The ligands with modifications at the R2-position of the tetrahydroquinoline are marked with crosses; they are all significantly improved in the 2- and 3-trajectory version. The standard errors, which are 0.19–0.34 kcal/mol for the 1-traj and 1.02–1.71 kcal/mol for the 2- and 3-traj approaches, are not shown for reasons of clarity. They are calculated using a bootstrapping method (see Supporting Information). The 2- and 3-traj approaches have similar errors because the energy of the receptor is treated as a constant and hence the uncertainties are dominated by the energies of the complexes.
Figure 5
Figure 5. Improvement of the predictions by inclusion of the adaptation free energies of the receptor and the ligands: (a) the binding free energy changes between the 1-traj (black circles) and 2-traj (magenta circles) indicate the relative adaptation energies of the receptor; those between the 2-traj (magenta circles) and 3-traj (orange circles) show the adaptation energies of the ligands. The adaptation energies can be seen more clearly in panels b as a function of binding affinities, and in panels c for each ligand.
Figure 6
Figure 6. Correlations of free energy components and the experimental data from 3-traj approaches. Both bonded and nonbonded energy terms contribute to the ranking of binding affinities. Their combination (the MMPBSA energy) exhibits a better correlation with experimental data than the components themselves.
Figure 7
Figure 7. Correlations of the calculated binding free energy differences from the TIES study and from experimental measurement. The standard error bars from the TIES calculations are all no greater than 0.2 kcal/mol.
Figure 8
Figure 8. Calculated vs experimental binding free energies for ligands 1, 8, and 9 which are labeled in the 1-traj subfigure.
Figure 9
Figure 9. Calculated vs experimental binding free energies for ligands 10, 11, and 12 which are labeled in the 1-traj subfigure.
Figure 10
Figure 10. Correlations of the internal energy contributions to the calculated binding free energies and experimental measurement. The internal energy changes are calculated as the differences of the binding free energies between those from the 1-traj and 3-traj approaches: ΔΔGcalc = ΔGbinding3-traj – ΔGbinding1-traj.
References
ARTICLE SECTIONSThis article references 40 other publications.
- 1Lounnas, V.; Ritschel, T.; Kelder, J.; McGuire, R.; Bywater, R. P.; Foloppe, N. Current progress in structure-based rational drug design marks a new mindset in drug discovery Comput. Struct. Biotechnol. J. 2013, 5, e201302011 DOI: 10.5936/csbj.201302011Google ScholarThere is no corresponding record for this reference.
- 2Green, D. V.; Leach, A. R.; Head, M. S. Computer-aided molecular design under the SWOTlight J. Comput.-Aided Mol. Des. 2012, 26 (1) 51– 56 DOI: 10.1007/s10822-011-9514-1Google ScholarThere is no corresponding record for this reference.
- 3Kuhn, B.; Guba, W.; Hert, J.; Banner, D.; Bissantz, C.; Ceccarelli, S.; Haap, W.; Korner, M.; Kuglstatter, A.; Lerner, C.; Mattei, P.; Neidhart, W.; Pinard, E.; Rudolph, M. G.; Schulz-Gasch, T.; Woltering, T.; Stahl, M. A real-world perspective on molecular design J. Med. Chem. 2016, 59 (9) 4087– 4102 DOI: 10.1021/acs.jmedchem.5b01875Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XisVyjtb0%253D&md5=0b81f2a0e9996c5dae44e61e5488ba79A Real-World Perspective on Molecular DesignKuhn, Bernd; Guba, Wolfgang; Hert, Jerome; Banner, David; Bissantz, Caterina; Ceccarelli, Simona; Haap, Wolfgang; Korner, Matthias; Kuglstatter, Andreas; Lerner, Christian; Mattei, Patrizio; Neidhart, Werner; Pinard, Emmanuel; Rudolph, Markus G.; Schulz-Gasch, Tanja; Woltering, Thomas; Stahl, MartinJournal of Medicinal Chemistry (2016), 59 (9), 4087-4102CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We present a series of small mol. drug discovery case studies where computational methods were prospectively employed to impact Roche research projects, with the aim of highlighting those methods that provide real added value. Our brief accounts encompass a broad range of methods and techniques applied to a variety of enzymes and receptors. Most of these are based on judicious application of knowledge about mol. conformations and interactions: filling of lipophilic pockets to gain affinity or selectivity, addn. of polar substituents, scaffold hopping, transfer of SAR, conformation anal., and mol. overlays. A case study of sequence-driven focused screening is presented to illustrate how appropriate preprocessing of information enables effective exploitation of prior knowledge. We conclude that qual. statements enabling chemists to focus on promising regions of chem. space are often more impactful than quant. prediction.
- 4Wang, L.; Wu, Y.; Deng, Y.; Kim, B.; Pierce, L.; Krilov, G.; Lupyan, D.; Robinson, S.; Dahlgren, M. K.; Greenwood, J.; Romero, D. L.; Masse, C.; Knight, J. L.; Steinbrecher, T.; Beuming, T.; Damm, W.; Harder, E.; Sherman, W.; Brewer, M.; Wester, R.; Murcko, M.; Frye, L.; Farid, R.; Lin, T.; Mobley, D. L.; Jorgensen, W. L.; Berne, B. J.; Friesner, R. A.; Abel, R. Accurate and reliable prediction of relative ligand binding potency in prospective drug discovery by way of a modern free-energy calculation protocol and force field J. Am. Chem. Soc. 2015, 137 (7) 2695– 2703 DOI: 10.1021/ja512751qGoogle Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsF2iuro%253D&md5=37a4f4a6c085f47ed531342643b6c33bAccurate and Reliable Prediction of Relative Ligand Binding Potency in Prospective Drug Discovery by Way of a Modern Free-Energy Calculation Protocol and Force FieldWang, Lingle; Wu, Yujie; Deng, Yuqing; Kim, Byungchan; Pierce, Levi; Krilov, Goran; Lupyan, Dmitry; Robinson, Shaughnessy; Dahlgren, Markus K.; Greenwood, Jeremy; Romero, Donna L.; Masse, Craig; Knight, Jennifer L.; Steinbrecher, Thomas; Beuming, Thijs; Damm, Wolfgang; Harder, Ed; Sherman, Woody; Brewer, Mark; Wester, Ron; Murcko, Mark; Frye, Leah; Farid, Ramy; Lin, Teng; Mobley, David L.; Jorgensen, William L.; Berne, Bruce J.; Friesner, Richard A.; Abel, RobertJournal of the American Chemical Society (2015), 137 (7), 2695-2703CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Designing tight-binding ligands is a primary objective of small-mol. drug discovery. Over the past few decades, free-energy calcns. have benefited from improved force fields and sampling algorithms, as well as the advent of low-cost parallel computing. However, it has proven to be challenging to reliably achieve the level of accuracy that would be needed to guide lead optimization (∼5× in binding affinity) for a wide range of ligands and protein targets. Not surprisingly, widespread com. application of free-energy simulations has been limited due to the lack of large-scale validation coupled with the tech. challenges traditionally assocd. with running these types of calcns. Here, we report an approach that achieves an unprecedented level of accuracy across a broad range of target classes and ligands, with retrospective results encompassing 200 ligands and a wide variety of chem. perturbations, many of which involve significant changes in ligand chem. structures. In addn., we have applied the method in prospective drug discovery projects and found a significant improvement in the quality of the compds. synthesized that have been predicted to be potent. Compds. predicted to be potent by this approach have a substantial redn. in false positives relative to compds. synthesized on the basis of other computational or medicinal chem. approaches. Furthermore, the results are consistent with those obtained from our retrospective studies, demonstrating the robustness and broad range of applicability of this approach, which can be used to drive decisions in lead optimization.
- 5Coveney, P. V.; Wan, S. On the calculation of equilibrium thermodynamic properties from molecular dynamics Phys. Chem. Chem. Phys. 2016, 18 (44) 30236– 30240 DOI: 10.1039/C6CP02349EGoogle Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XntVyisr8%253D&md5=f7950af8255cf494a117d46bc1b5191eOn the calculation of equilibrium thermodynamic properties from molecular dynamicsCoveney, Peter V.; Wan, ShunzhouPhysical Chemistry Chemical Physics (2016), 18 (44), 30236-30240CODEN: PPCPFQ; ISSN:1463-9076. (Royal Society of Chemistry)The purpose of statistical mechanics is to provide a route to the calcn. of macroscopic properties of matter from their constituent microscopic components. It is well known that the macrostates emerge as ensemble avs. of microstates. However, this is more often stated than implemented in computer simulation studies. Here we consider foundational aspects of statistical mechanics which are overlooked in most textbooks and research articles that purport to compute macroscopic behavior from microscopic descriptions based on classical mechanics and show how due attention to these issues leads in directions which have not been widely appreciated in the field of mol. dynamics simulation.
- 6Arrowsmith, C. H.; Bountra, C.; Fish, P. V.; Lee, K.; Schapira, M. Epigenetic protein families: a new frontier for drug discovery Nat. Rev. Drug Discovery 2012, 11 (5) 384– 400 DOI: 10.1038/nrd3674Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XlsFWjsbs%253D&md5=eb82034466a43107aa74a18ccf6d29f5Epigenetic protein families: a new frontier for drug discoveryArrowsmith, Cheryl H.; Bountra, Chas; Fish, Paul V.; Lee, Kevin; Schapira, MatthieuNature Reviews Drug Discovery (2012), 11 (5), 384-400CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Epigenetic regulation of gene expression is a dynamic and reversible process that establishes normal cellular phenotypes but also contributes to human diseases. At the mol. level, epigenetic regulation involves hierarchical covalent modification of DNA and the proteins that package DNA, such as histones. Here, we review the key protein families that mediate epigenetic signalling through the acetylation and methylation of histones, including histone deacetylases, protein methyltransferases, lysine demethylases, bromodomain-contg. proteins and proteins that bind to methylated histones. These protein families are emerging as druggable classes of enzymes and druggable classes of protein-protein interaction domains. In this article, we discuss the known links with disease, basic mol. mechanisms of action and recent progress in the pharmacol. modulation of each class of proteins.
- 7Copeland, R. A. Epigenetic medicinal chemistry ACS Med. Chem. Lett. 2016, 7 (2) 124– 127 DOI: 10.1021/acsmedchemlett.5b00462Google ScholarThere is no corresponding record for this reference.
- 8Copeland, R. A.; Olhava, E. J.; Scott, M. P. Targeting epigenetic enzymes for drug discovery Curr. Opin. Chem. Biol. 2010, 14 (4) 505– 510 DOI: 10.1016/j.cbpa.2010.06.174Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXpvVyqtrs%253D&md5=56f501166e98ff60aa6781c76650810dTargeting epigenetic enzymes for drug discoveryCopeland, Robert A.; Olhava, Edward J.; Scott, Margaret PorterCurrent Opinion in Chemical Biology (2010), 14 (4), 505-510CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review. Epigenetic control of gene transcription is the result of enzyme-mediated covalent modifications of promoter-region DNA sites and of histone proteins around which chromosomal DNA is wound. Many of the enzymes that mediate these epigenetic reactions are dysregulated in human diseases. Small mol. inhibitors against two classes of these enzymes have been approved for use in patients: DNA methyltransferase (DNMT) inhibitors and histone deacetylase inhibitors. Other classes of epigenetic enzymes have been demonstrated to have strong disease assocn. and are currently being targeted for small mol. inhibition. In this article we review these enzymes and chem. biol. approaches aimed at discovering small mol. inhibitors against them for therapeutic use.
- 9Theodoulou, N. H.; Tomkinson, N. C.; Prinjha, R. K.; Humphreys, P. G. Clinical progress and pharmacology of small molecule bromodomain inhibitors Curr. Opin. Chem. Biol. 2016, 33, 58– 66 DOI: 10.1016/j.cbpa.2016.05.028Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xps1artb8%253D&md5=4bcd42755b2dd3cebd7fae417e69e83fClinical progress and pharmacology of small molecule bromodomain inhibitorsTheodoulou, Natalie H.; Tomkinson, Nicholas C. O.; Prinjha, Rab K.; Humphreys, Philip G.Current Opinion in Chemical Biology (2016), 33 (), 58-66CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)Bromodomains have emerged as an exciting target class for drug discovery over the past decade. Research has primarily focused on the bromodomain and extra terminal (BET) family of bromodomains, which has led to the development of multiple small mol. inhibitors and an increasing no. of clin. assets. The excitement centered on the clin. potential of BET inhibition has stimulated intense interest in the broader family and the growing no. of non-BET bromodomain chem. probes has facilitated phenotypic investigations, implicating these targets in a variety of disease pathways including cancer, inflammation, embryonic development and neurol. disorders.
- 10Bamborough, P.; Diallo, H.; Goodacre, J. D.; Gordon, L.; Lewis, A.; Seal, J. T.; Wilson, D. M.; Woodrow, M. D.; Chung, C. W. Fragment-based discovery of bromodomain inhibitors part 2: optimization of phenylisoxazole sulfonamides J. Med. Chem. 2012, 55 (2) 587– 596 DOI: 10.1021/jm201283qGoogle ScholarThere is no corresponding record for this reference.
- 11Chung, C. W.; Coste, H.; White, J. H.; Mirguet, O.; Wilde, J.; Gosmini, R. L.; Delves, C.; Magny, S. M.; Woodward, R.; Hughes, S. A.; Boursier, E. V.; Flynn, H.; Bouillot, A. M.; Bamborough, P.; Brusq, J. M.; Gellibert, F. J.; Jones, E. J.; Riou, A. M.; Homes, P.; Martin, S. L.; Uings, I. J.; Toum, J.; Clement, C. A.; Boullay, A. B.; Grimley, R. L.; Blandel, F. M.; Prinjha, R. K.; Lee, K.; Kirilovsky, J.; Nicodeme, E. Discovery and characterization of small molecule inhibitors of the BET family bromodomains J. Med. Chem. 2011, 54 (11) 3827– 3838 DOI: 10.1021/jm200108tGoogle Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmtVWnurs%253D&md5=ebc0f6062321dece877259d775f1c19bDiscovery and characterization of small molecule inhibitors of the BET family bromodomainsChung, Chun-wa; Coste, Herve; White, Julia H.; Mirguet, Olivier; Wilde, Jonathan; Gosmini, Romain L.; Delves, Chris; Magny, Sylvie M.; Woodward, Robert; Hughes, Stephen A.; Boursier, Eric V.; Flynn, Helen; Bouillot, Anne M.; Bamborough, Paul; Brusq, Jean-Marie G.; Gellibert, Francoise J.; Jones, Emma J.; Riou, Alizon M.; Homes, Paul; Martin, Sandrine L.; Uings, Iain J.; Toum, Jerome; Clement, Catherine A.; Boullay, Anne-Benedicte; Grimley, Rachel L.; Blandel, Florence M.; Prinjha, Rab K.; Lee, Kevin; Kirilovsky, Jorge; Nicodeme, EdwigeJournal of Medicinal Chemistry (2011), 54 (11), 3827-3838CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Epigenetic mechanisms of gene regulation have a profound role in normal development and disease processes. An integral part of this mechanism occurs through lysine acetylation of histone tails which are recognized by bromodomains. While the biol. and structural characterization of many bromodomain contg. proteins has advanced considerably, the therapeutic tractability of this protein family is only now becoming understood. This paper describes the discovery and mol. characterization of potent (nM) small mol. inhibitors that disrupt the function of the BET family of bromodomains (Brd2, Brd3, and Brd4). By using a combination of phenotypic screening, chemoproteomics, and biophys. studies, we have discovered that the protein-protein interactions between bromodomains and acetylated histones can be antagonized by selective small mols. that bind at the acetylated lysine recognition pocket. X-ray crystal structures of compds. bound into bromodomains of Brd2 and Brd4 elucidate the mol. interactions of binding and explain the precisely defined stereochem. required for activity.
- 12Chung, C. W.; Dean, A. W.; Woolven, J. M.; Bamborough, P. Fragment-based discovery of bromodomain inhibitors part 1: inhibitor binding modes and implications for lead discovery J. Med. Chem. 2012, 55 (2) 576– 586 DOI: 10.1021/jm201320wGoogle Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsFKktrnN&md5=5b7d7239d0de57266d94beef1ea080aeFragment-Based Discovery of Bromodomain Inhibitors Part 1: Inhibitor Binding Modes and Implications for Lead DiscoveryChung, Chun-wa; Dean, Anthony W.; Woolven, James M.; Bamborough, PaulJournal of Medicinal Chemistry (2012), 55 (2), 576-586CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Bromodomain-contg. proteins are key epigenetic regulators of gene transcription and readers of the histone code. However, the therapeutic benefits of modulating this target class are largely unexplored due to the lack of suitable chem. probes. This article describes the generation of lead mols. for the BET bromodomains through screening a fragment set chosen using structural insights and computational approaches. Anal. of 40 BRD2/fragment x-ray complexes highlights both shared and disparate interaction features that may be exploited for affinity and selectivity. Six representative crystal structures are then exemplified in detail. Two of the fragments are completely new bromodomain chemotypes, and three have never before been crystd. in a bromodomain, so our results significantly extend the limited public knowledge-base of crystallog. small mol./bromodomain interactions. Certain fragments (including paracetamol) bind in a consistent mode to different bromodomains such as CREBBP, suggesting their potential to act as generic bromodomain templates. An important implication is that the bromodomains are not only a phylogenetic family but also a system in which chem. and structural knowledge of one bromodomain gives insights transferable to others.
- 13Sadiq, S. K.; Wright, D.; Watson, S. J.; Zasada, S. J.; Stoica, I.; Coveney, P. V. Automated molecular simulation based binding affinity calculator for ligand-bound HIV-1 proteases J. Chem. Inf. Model. 2008, 48 (9) 1909– 1919 DOI: 10.1021/ci8000937Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtVShtLjN&md5=c616ac9330dc1f4cb9808d68fabc7e4cAutomated Molecular Simulation Based Binding Affinity Calculator for Ligand-Bound HIV-1 ProteasesSadiq, S. Kashif; Wright, David; Watson, Simon J.; Zasada, Stefan J.; Stoica, Ileana; Coveney, Peter V.Journal of Chemical Information and Modeling (2008), 48 (9), 1909-1919CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)The successful application of high throughput mol. simulations to det. biochem. properties would be of great importance to the biomedical community if such simulations could be turned around in a clin. relevant timescale. An important example is the detn. of antiretroviral inhibitor efficacy against varying strains of HIV through calcn. of drug-protein binding affinities. We describe the Binding Affinity Calculator (BAC), a tool for the automated calcn. of HIV-1 protease-ligand binding affinities. The tool employs fully atomistic mol. simulations alongside the well established mol. mechanics Poisson-Boltzmann solvent accessible surface area (MMPBSA) free energy methodol. to enable the calcn. of the binding free energy of several ligand-protease complexes, including all nine FDA approved inhibitors of HIV-1 protease and seven of the natural substrates cleaved by the protease. This enables the efficacy of these inhibitors to be ranked across several mutant strains of the protease relative to the wildtype. BAC is a tool that utilizes the power provided by a computational grid to automate all of the stages required to compute free energies of binding: model prepn., equilibration, simulation, postprocessing, and data-marshaling around the generally widely distributed compute resources utilized. Such automation enables the mol. dynamics methodol. to be used in a high throughput manner not achievable by manual methods. This paper describes the architecture and workflow management of BAC and the function of each of its components. Given adequate compute resources, BAC can yield quant. information regarding drug resistance at the mol. level within 96 h. Such a timescale is of direct clin. relevance and can assist in decision support for the assessment of patient-specific optimal drug treatment and the subsequent response to therapy for any given genotype.
- 14Groen, D.; Bhati, A. P.; Suter, J.; Hetherington, J.; Zasada, S. J.; Coveney, P. V. FabSim: Facilitating computational research through automation on large-scale and distributed e-infrastructures Comput. Phys. Commun. 2016, 207, 375– 385 DOI: 10.1016/j.cpc.2016.05.020Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xptl2nu7s%253D&md5=4b7e50658ec8307f7fa5fcd16747cab6FabSim: Facilitating computational research through automation on large-scale and distributed e-infrastructuresGroen, Derek; Bhati, Agastya P.; Suter, James; Hetherington, James; Zasada, Stefan J.; Coveney, Peter V.Computer Physics Communications (2016), 207 (), 375-385CODEN: CPHCBZ; ISSN:0010-4655. (Elsevier B.V.)We present FabSim, a toolkit developed to simplify a range of computational tasks for researchers in diverse disciplines. FabSim is flexible, adaptable, and allows users to perform a wide range of tasks with ease. It also provides a systematic way to automate the use of resources, including HPC and distributed machines, and to make tasks easier to repeat by recording contextual information. To demonstrate this, we present three use cases where FabSim has enhanced our research productivity. These include simulating cerebrovascular bloodflow, modeling clay-polymer nanocomposites across multiple scales, and calcg. ligand-protein binding affinities.
- 15Wyce, A.; Ganji, G.; Smitheman, K. N.; Chung, C. W.; Korenchuk, S.; Bai, Y.; Barbash, O.; Le, B.; Craggs, P. D.; McCabe, M. T.; Kennedy-Wilson, K. M.; Sanchez, L. V.; Gosmini, R. L.; Parr, N.; McHugh, C. F.; Dhanak, D.; Prinjha, R. K.; Auger, K. R.; Tummino, P. J. BET inhibition silences expression of MYCN and BCL2 and induces cytotoxicity in neuroblastoma tumor models PLoS One 2013, 8 (8) e72967 DOI: 10.1371/journal.pone.0072967Google ScholarThere is no corresponding record for this reference.
- 16Gosmini, R.; Nguyen, V. L.; Toum, J.; Simon, C.; Brusq, J. M.; Krysa, G.; Mirguet, O.; Riou-Eymard, A. M.; Boursier, E. V.; Trottet, L.; Bamborough, P.; Clark, H.; Chung, C. W.; Cutler, L.; Demont, E. H.; Kaur, R.; Lewis, A. J.; Schilling, M. B.; Soden, P. E.; Taylor, S.; Walker, A. L.; Walker, M. D.; Prinjha, R. K.; Nicodeme, E. The discovery of I-BET726 (GSK1324726A), a potent tetrahydroquinoline ApoA1 up-regulator and selective BET bromodomain inhibitor J. Med. Chem. 2014, 57 (19) 8111– 8131 DOI: 10.1021/jm5010539Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1Wlur7J&md5=b1c9c9cae73c41252cf73908d561bd9bThe Discovery of I-BET726 (GSK1324726A), a Potent Tetrahydroquinoline ApoA1 Up-Regulator and Selective BET Bromodomain InhibitorGosmini, Romain; Nguyen, Van Loc; Toum, Jerome; Simon, Christophe; Brusq, Jean-Marie G.; Krysa, Gael; Mirguet, Olivier; Riou-Eymard, Alizon M.; Boursier, Eric V.; Trottet, Lionel; Bamborough, Paul; Clark, Hugh; Chung, Chun-wa; Cutler, Leanne; Demont, Emmanuel H.; Kaur, Rejbinder; Lewis, Antonia J.; Schilling, Mark B.; Soden, Peter E.; Taylor, Simon; Walker, Ann L.; Walker, Matthew D.; Prinjha, Rab K.; Nicodeme, EdwigeJournal of Medicinal Chemistry (2014), 57 (19), 8111-8131CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Through their function as epigenetic readers of the histone code, the BET family of bromodomain-contg. proteins regulate expression of multiple genes of therapeutic relevance, including those involved in tumor cell growth and inflammation. BET bromodomain inhibitors have profound antiproliferative and anti-inflammatory effects which translate into efficacy in oncol. and inflammation models, and the first compds. have now progressed into clin. trials. The exciting biol. of the BETs has led to great interest in the discovery of novel inhibitor classes. Here we describe the identification of a novel tetrahydroquinoline series through up-regulation of apolipoprotein A1 and the optimization into potent compds. active in murine models of septic shock and neuroblastoma. At the mol. level, these effects are produced by inhibition of BET bromodomains. X-ray crystallog. reveals the interactions explaining the structure-activity relationships of binding. The resulting lead mol., I-BET726, represents a new, potent, and selective class of tetrahydroquinoline-based BET inhibitors.
- 17Aldeghi, M.; Heifetz, A.; Bodkin, M. J.; Knapp, S.; Biggin, P. C. Accurate calculation of the absolute free energy of binding for drug molecules Chem. Sci. 2016, 7 (1) 207– 218 DOI: 10.1039/C5SC02678DGoogle Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFGku7%252FI&md5=d00cac3b87f1bd11837d6f292bd8c6e5Accurate calculation of the absolute free energy of binding for drug moleculesAldeghi, Matteo; Heifetz, Alexander; Bodkin, Michael J.; Knapp, Stefan; Biggin, Philip C.Chemical Science (2016), 7 (1), 207-218CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Accurate prediction of binding affinities has been a central goal of computational chem. for decades, yet remains elusive. Despite good progress, the required accuracy for use in a drug-discovery context has not been consistently achieved for drug-like mols. Here, we perform abs. free energy calcns. based on a thermodn. cycle for a set of diverse inhibitors binding to bromodomain-contg. protein 4 (BRD4) and demonstrate that a mean abs. error of 0.6 kcal mol-1 can be achieved. We also show a similar level of accuracy (1.0 kcal mol-1) can be achieved in pseudo prospective approach. Bromodomains are epigenetic mark readers that recognize acetylation motifs and regulate gene transcription, and are currently being investigated as therapeutic targets for cancer and inflammation. The unprecedented accuracy offers the exciting prospect that the binding free energy of drug-like compds. can be predicted for pharmacol. relevant targets.
- 18Lindorff-Larsen, K.; Piana, S.; Palmo, K.; Maragakis, P.; Klepeis, J. L.; Dror, R. O.; Shaw, D. E. Improved side-chain torsion potentials for the Amber ff99SB protein force field Proteins: Struct., Funct., Genet. 2010, 78 (8) 1950– 1958 DOI: 10.1002/prot.22711Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXkvFegtLo%253D&md5=447a9004026e2b93f0f7beff165daa09Improved side-chain torsion potentials for the Amber ff99SB protein force fieldLindorff-Larsen, Kresten; Piana, Stefano; Palmo, Kim; Maragakis, Paul; Klepeis, John L.; Dror, Ron O.; Shaw, David E.Proteins: Structure, Function, and Bioinformatics (2010), 78 (8), 1950-1958CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)Recent advances in hardware and software have enabled increasingly long mol. dynamics (MD) simulations of biomols., exposing certain limitations in the accuracy of the force fields used for such simulations and spurring efforts to refine these force fields. Recent modifications to the Amber and CHARMM protein force fields, for example, have improved the backbone torsion potentials, remedying deficiencies in earlier versions. Here, the authors further advance simulation accuracy by improving the amino acid side-chain torsion potentials of the Amber ff99SB force field. First, the authors used simulations of model alpha-helical systems to identify the four residue types whose rotamer distribution differed the most from expectations based on Protein Data Bank statistics. Second, the authors optimized the side-chain torsion potentials of these residues to match new, high-level quantum-mech. calcns. Finally, the authors used microsecond-timescale MD simulations in explicit solvent to validate the resulting force field against a large set of exptl. NMR measurements that directly probe side-chain conformations. The new force field, which the authors have termed Amber ff99SB-ILDN, exhibits considerably better agreement with the NMR data. Proteins 2010. © 2010 Wiley-Liss, Inc.
- 19Wang, J.; Wolf, R. M.; Caldwell, J. W.; Kollman, P. A.; Case, D. A. Development and testing of a general amber force field J. Comput. Chem. 2004, 25 (9) 1157– 1174 DOI: 10.1002/jcc.20035Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXksFakurc%253D&md5=2992017a8cf51f89290ae2562403b115Development and testing of a general Amber force fieldWang, Junmei; Wolf, Romain M.; Caldwell, James W.; Kollman, Peter A.; Case, David A.Journal of Computational Chemistry (2004), 25 (9), 1157-1174CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)We describe here a general Amber force field (GAFF) for org. mols. GAFF is designed to be compatible with existing Amber force fields for proteins and nucleic acids, and has parameters for most org. and pharmaceutical mols. that are composed of H, C, N, O, S, P, and halogens. It uses a simple functional form and a limited no. of atom types, but incorporates both empirical and heuristic models to est. force consts. and partial at. charges. The performance of GAFF in test cases is encouraging. In test I, 74 crystallog. structures were compared to GAFF minimized structures, with a root-mean-square displacement of 0.26 Å, which is comparable to that of the Tripos 5.2 force field (0.25 Å) and better than those of MMFF 94 and CHARMm (0.47 and 0.44 Å, resp.). In test II, gas phase minimizations were performed on 22 nucleic acid base pairs, and the minimized structures and intermol. energies were compared to MP2/6-31G* results. The RMS of displacements and relative energies were 0.25 Å and 1.2 kcal/mol, resp. These data are comparable to results from Parm99/RESP (0.16 Å and 1.18 kcal/mol, resp.), which were parameterized to these base pairs. Test III looked at the relative energies of 71 conformational pairs that were used in development of the Parm99 force field. The RMS error in relative energies (compared to expt.) is about 0.5 kcal/mol. GAFF can be applied to wide range of mols. in an automatic fashion, making it suitable for rational drug design and database searching.
- 20Frisch, M. J.; Trucks, G. W.; Schlegel, H. B.; Scuseria, G. E.; Robb, M. A.; Cheeseman, J. R.; Montgomery, J. A., Jr.; Vreven, T.; Kudin, K. N.; Burant, J. C.; Millam, J. M.; Iyengar, S. S.; Tomasi, J.; Barone, V.; Mennucci, B.; Cossi, M.; Scalmani, G.; Rega, N.; Petersson, G. A.; Nakatsuji, H.; Hada, M.; Ehara, M.; Toyota, K.; Fukuda, R.; Hasegawa, J.; Ishida, M.; Nakajima, T.; Honda, Y.; Kitao, O.; Nakai, H.; Klene, M.; Li, X.; Knox, J. E.; Hratchian, H. P.; Cross, J. B.; Bakken, V.; Adamo, C.; Jaramillo, J.; Gomperts, R.; Stratmann, R. E.; Yazyev, O.; Austin, A. J.; Cammi, R.; Pomelli, C.; Ochterski, J. W.; Ayala, P. Y.; Morokuma, K.; Voth, G. A.; Salvador, P.; Dannenberg, J. J.; Zakrzewski, V. G.; Dapprich, S.; Daniels, A. D.; Strain, M. C.; Farkas, O.; Malick, D. K.; Rabuck, A. D.; Raghavachari, K.; Foresman, J. B.; Ortiz, J. V.; Cui, Q.; Baboul, A. G.; Clifford, S.; Cioslowski, J.; Stefanov, B. B.; Liu, G.; Liashenko, A.; Piskorz, P.; Komaromi, I.; Martin, R. L.; Fox, D. J.; Keith, T.; Al-Laham, M. A.; Peng, C. Y.; Nanayakkara, A.; Challacombe, M.; Gill, P. M. W.; Johnson, B.; Chen, W.; Wong, M. W.; Gonzalez, C.; Pople, J. A., Gaussian 03; Gaussian, Inc.: Wallingford, CT, 2004.Google ScholarThere is no corresponding record for this reference.
- 21Case, D. A.; Cheatham, T. E., 3rd; Darden, T.; Gohlke, H.; Luo, R.; Merz, K. M., Jr.; Onufriev, A.; Simmerling, C.; Wang, B.; Woods, R. J. The Amber biomolecular simulation programs J. Comput. Chem. 2005, 26 (16) 1668– 1688 DOI: 10.1002/jcc.20290Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1SlsbbM&md5=93be29ff894bab96c783d24e9886c7d0The amber biomolecular simulation programsCase, David A.; Cheatham, Thomas E., III; Darden, Tom; Gohlke, Holger; Luo, Ray; Merz, Kenneth M., Jr.; Onufriev, Alexey; Simmerling, Carlos; Wang, Bing; Woods, Robert J.Journal of Computational Chemistry (2005), 26 (16), 1668-1688CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)The authors describe the development, current features, and some directions for future development of the Amber package of computer programs. This package evolved from a program that was constructed in the late 1970s to do Assisted Model Building with Energy Refinement, and now contains a group of programs embodying a no. of powerful tools of modern computational chem., focused on mol. dynamics and free energy calcns. of proteins, nucleic acids, and carbohydrates.
- 22Laio, A.; Gervasio, F. L. Metadynamics: a method to simulate rare events and reconstruct the free energy in biophysics, chemistry and material science Rep. Prog. Phys. 2008, 71 (12) 126601 DOI: 10.1088/0034-4885/71/12/126601Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFyntrk%253D&md5=cd84cfc103f97c7d7ccf09fc434e2478Metadynamics: a method to stimulate rare events and reconstruct the free energy in biophysics, chemistry and material scienceLaio, Alessandro; Gervasio, Francesco L.Reports on Progress in Physics (2008), 71 (12), 126601/1-126601/22CODEN: RPPHAG; ISSN:0034-4885. (Institute of Physics Publishing)A review. Metadynamics is a powerful algorithm that can be used both for reconstructing the free energy and for accelerating rare events in systems described by complex Hamiltonians, at the classical or at the quantum level. In the algorithm the normal evolution of the system is biased by a history-dependent potential constructed as a sum of Gaussians centered along the trajectory followed by a suitably chosen set of collective variables. The sum of Gaussians is exploited for reconstructing iteratively an estimator of the free energy and forcing the system to escape from local min. This review is intended to provide a comprehensive description of the algorithm, with a focus on the practical aspects that need to be addressed when one attempts to apply metadynamics to a new system: (i) the choice of the appropriate set of collective variables; (ii) the optimal choice of the metadynamics parameters and (iii) how to control the error and ensure convergence of the algorithm.
- 23Lin, Y. L.; Aleksandrov, A.; Simonson, T.; Roux, B. An overview of electrostatic free energy computations for solutions and proteins J. Chem. Theory Comput. 2014, 10 (7) 2690– 2709 DOI: 10.1021/ct500195pGoogle Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXnslaksLc%253D&md5=6aa50d77dc3326ae518f682336b9f4a8An Overview of Electrostatic Free Energy Computations for Solutions and ProteinsLin, Yen-Lin; Aleksandrov, Alexey; Simonson, Thomas; Roux, BenoitJournal of Chemical Theory and Computation (2014), 10 (7), 2690-2709CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)A review. Free energy simulations for electrostatic and charging processes in complex mol. systems encounter specific difficulties owing to the long-range, 1/r Coulomb interaction. To calc. the solvation free energy of a simple ion, it is essential to take into account the polarization of nearby solvent but also the electrostatic potential drop across the liq.-gas boundary, however distant. The latter does not exist in a simulation model based on periodic boundary conditions because there is no phys. boundary to the system. An important consequence is that the ref. value of the electrostatic potential is not an ion in a vacuum. Also, in an infinite system, the electrostatic potential felt by a perturbing charge is conditionally convergent and dependent on the choice of computational conventions. Furthermore, with Ewald lattice summation and tinfoil conducting boundary conditions, the charges experience a spurious shift in the potential that depends on the details of the simulation system such as the vol. fraction occupied by the solvent. All these issues can be handled with established computational protocols, as reviewed here and illustrated for several small ions and three solvated proteins.
- 24Wan, S.; Knapp, B.; Wright, D. W.; Deane, C. M.; Coveney, P. V. Rapid, precise, and reproducible prediction of peptide-MHC binding affinities from molecular dynamics that correlate well with experiment J. Chem. Theory Comput. 2015, 11 (7) 3346– 3356 DOI: 10.1021/acs.jctc.5b00179Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVaksLjN&md5=860aa35a6e8e4013e2b0c7f0ae99170fRapid, precise, and reproducible prediction of peptide-MHC binding affinities from molecular dynamics that correlate well with experimentWan, Shunzhou; Knapp, Bernhard; Wright, David W.; Deane, Charlotte M.; Coveney, Peter V.Journal of Chemical Theory and Computation (2015), 11 (7), 3346-3356CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The presentation of potentially pathogenic peptides by major histocompatibility complex (MHC) mols. is one of the most important processes in adaptive immune defense. Prediction of peptide-MHC (pMHC) binding affinities is therefore a principal objective of theor. immunol. Machine learning techniques achieve good results if substantial exptl. training data are available. Approaches based on structural information become necessary if sufficiently similar training data are unavailable for a specific MHC allele, although they have often been deemed to lack accuracy. In this study, we use a free energy method to rank the binding affinities of 12 diverse peptides bound by a class I MHC mol. HLA-A*02:01. The method is based on enhanced sampling of mol. dynamics calcns. in combination with a continuum solvent approxn. and includes ests. of the configurational entropy based on either a one or a three trajectory protocol. It produces precise and reproducible free energy ests. which correlate well with exptl. measurements. If the results are combined with an amino acid hydrophobicity scale, then an extremely good ranking of peptide binding affinities emerges. Our approach is rapid, robust, and applicable to a wide range of ligand-receptor interactions without further adjustment.
- 25Bhati, A. P.; Wan, S.; Wright, D. W.; Coveney, P. V. Rapid, accurate, precise and reliable relative free energy prediction using ensemble based thermodynamic integration J. Chem. Theory Comput. 2016, DOI: 10.1021/acs.jctc.6b00979Google ScholarThere is no corresponding record for this reference.
- 26Kollman, P. A.; Massova, I.; Reyes, C.; Kuhn, B.; Huo, S.; Chong, L.; Lee, M.; Lee, T.; Duan, Y.; Wang, W.; Donini, O.; Cieplak, P.; Srinivasan, J.; Case, D. A.; Cheatham, T. E., III Calculating structures and free energies of complex molecules: combining molecular mechanics and continuum models Acc. Chem. Res. 2000, 33 (12) 889– 897 DOI: 10.1021/ar000033jGoogle Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXmvFGiu7g%253D&md5=8436ee610ae145894428db1a1deff73cCalculating Structures and Free Energies of Complex Molecules: Combining Molecular Mechanics and Continuum ModelsKollman, Peter A.; Massova, Irina; Reyes, Carolina; Kuhn, Bernd; Huo, Shuanghong; Chong, Lillian; Lee, Matthew; Lee, Taisung; Duan, Yong; Wang, Wei; Donini, Oreola; Cieplak, Piotr; Srinivasan, Jaysharee; Case, David A.; Cheatham, Thomas E., IIIAccounts of Chemical Research (2000), 33 (12), 889-897CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review, with 63 refs. A historical perspective on the application of mol. dynamics (MD) to biol. macromols. is presented. Recent developments combining state-of-the-art force fields with continuum solvation calcns. have allowed us to reach the fourth era of MD applications in which one can often derive both accurate structure and accurate relative free energies from mol. dynamics trajectories. We illustrate such applications on nucleic acid duplexes, RNA hairpins, protein folding trajectories, and protein-ligand, protein-protein, and protein-nucleic acid interactions.
- 27Wright, D. W.; Hall, B. A.; Kenway, O. A.; Jha, S.; Coveney, P. V. Computing Clinically Relevant Binding Free Energies of HIV-1 Protease Inhibitors J. Chem. Theory Comput. 2014, 10 (3) 1228– 1241 DOI: 10.1021/ct4007037Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXht12ltrk%253D&md5=56bc7e7c8f6bbdd694bfc76c20dcec63Computing Clinically Relevant Binding Free Energies of HIV-1 Protease InhibitorsWright, David W.; Hall, Benjamin A.; Kenway, Owain A.; Jha, Shantenu; Coveney, Peter V.Journal of Chemical Theory and Computation (2014), 10 (3), 1228-1241CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The use of mol. simulation to est. the strength of macromol. binding free energies is becoming increasingly widespread, with goals ranging from lead optimization and enrichment in drug discovery to personalizing or stratifying treatment regimes. To realize the potential of such approaches to predict new results, not merely to explain previous exptl. findings, it is necessary that the methods used are reliable and accurate, and that their limitations are thoroughly understood. However, the computational cost of atomistic simulation techniques such as mol. dynamics (MD) has meant that until recently little work has focused on validating and verifying the available free energy methodologies, with the consequence that many of the results published in the literature are not reproducible. Here, we present a detailed anal. of two of the most popular approx. methods for calcg. binding free energies from mol. simulations, mol. mechanics Poisson-Boltzmann surface area (MMPBSA) and mol. mechanics generalized Born surface area (MMGBSA), applied to the nine FDA-approved HIV-1 protease inhibitors. Our results show that the values obtained from replica simulations of the same protease-drug complex, differing only in initially assigned atom velocities, can vary by as much as 10 kcal mol-1, which is greater than the difference between the best and worst binding inhibitors under investigation. Despite this, anal. of ensembles of simulations producing 50 trajectories of 4 ns duration leads to well converged free energy ests. For seven inhibitors, we find that with correctly converged normal mode ests. of the configurational entropy, we can correctly distinguish inhibitors in agreement with exptl. data for both the MMPBSA and MMGBSA methods and thus have the ability to rank the efficacy of binding of this selection of drugs to the protease (no account is made for free energy penalties assocd. with protein distortion leading to the over estn. of the binding strength of the two largest inhibitors ritonavir and atazanavir). We obtain improved rankings and ests. of the relative binding strengths of the drugs by using a novel combination of MMPBSA/MMGBSA with normal mode entropy ests. and the free energy of assocn. calcd. directly from simulation trajectories. Our work provides a thorough assessment of what is required to produce converged and hence reliable free energies for protein-ligand binding.
- 28Genheden, S.; Ryde, U. The MM/PBSA and MM/GBSA methods to estimate ligand-binding affinities Expert Opin. Drug Discovery 2015, 10 (5) 449– 461 DOI: 10.1517/17460441.2015.1032936Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXntFGktr8%253D&md5=b123b88809f275564f95a2271ebd159fThe MM/PBSA and MM/GBSA methods to estimate ligand-binding affinitiesGenheden, Samuel; Ryde, UlfExpert Opinion on Drug Discovery (2015), 10 (5), 449-461CODEN: EODDBX; ISSN:1746-0441. (Informa Healthcare)Introduction: The mol. mechanics energies combined with the Poisson-Boltzmann or generalized Born and surface area continuum solvation (MM/PBSA and MM/GBSA) methods are popular approaches to est. the free energy of the binding of small ligands to biol. macromols. They are typically based on mol. dynamics simulations of the receptor-ligand complex and are therefore intermediate in both accuracy and computational effort between empirical scoring and strict alchem. perturbation methods. They have been applied to a large no. of systems with varying success. Areas covered: The authors review the use of MM/PBSA and MM/GBSA methods to calc. ligand-binding affinities, with an emphasis on calibration, testing and validation, as well as attempts to improve the methods, rather than on specific applications. Expert opinion: MM/PBSA and MM/GBSA are attractive approaches owing to their modular nature and that they do not require calcns. on a training set. They have been used successfully to reproduce and rationalize exptl. findings and to improve the results of virtual screening and docking. However, they contain several crude and questionable approxns., for example, the lack of conformational entropy and information about the no. and free energy of water mols. in the binding site. Moreover, there are many variants of the method and their performance varies strongly with the tested system. Likewise, most attempts to ameliorate the methods with more accurate approaches, for example, quantum-mech. calcns., polarizable force fields or improved solvation have deteriorated the results.
- 29Swanson, J. M.; Henchman, R. H.; McCammon, J. A. Revisiting free energy calculations: a theoretical connection to MM/PBSA and direct calculation of the association free energy Biophys. J. 2004, 86, 67– 74 DOI: 10.1016/S0006-3495(04)74084-9Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXlsV2hug%253D%253D&md5=720b2ec6ef0462edcda34f7be768bc44Revisiting free energy calculations: A theoretical connection to MM/PBSA and direct calculation of the association free energySwanson, Jessica M. J.; Henchman, Richard H.; McCammon, J. AndrewBiophysical Journal (2004), 86 (1, Pt. 1), 67-74CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)The prediction of abs. ligand-receptor binding affinities is essential in a wide range of biophys. queries, from the study of protein-protein interactions to structure-based drug design. End-point free energy methods, such as the Mol. Mechanics Poisson-Boltzmann Surface Area (MM/PBSA) model, have received much attention and widespread application in recent literature. These methods benefit from computational efficiency as only the initial and final states of the system are evaluated, yet there remains a need for strengthening their theor. foundation. Here a clear connection between statistical thermodn. and end-point free energy models is presented. The importance of the assocn. free energy, arising from one mol.'s loss of translational and rotational freedom from the std. state concn., is addressed. A novel method for calcg. this quantity directly from a mol. dynamics simulation is described. The challenges of accounting for changes in the protein conformation and its fluctuations from sep. simulations are discussed. A simple first-order approxn. of the configuration integral is presented to lay the groundwork for future efforts. This model has been applied to FKBP12, a small immunophilin that has been widely studied in the drug industry for its potential immunosuppressive and neuroregenerative effects.
- 30Genheden, S.; Ryde, U. How to obtain statistically converged MM/GBSA results J. Comput. Chem. 2010, 31 (4) 837– 846 DOI: 10.1002/jcc.21366Google ScholarThere is no corresponding record for this reference.
- 31Luo, R.; David, L.; Gilson, M. K. Accelerated Poisson-Boltzmann calculations for static and dynamic systems J. Comput. Chem. 2002, 23 (13) 1244– 1253 DOI: 10.1002/jcc.10120Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XmsVKgsLo%253D&md5=7ce67345061bdd6fcfae91be1650a578Accelerated Poisson-Boltzmann calculations for static and dynamic systemsLuo, Ray; David, Laurent; Gilson, Michael K.Journal of Computational Chemistry (2002), 23 (13), 1244-1253CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)We report an efficient implementation of the finite-difference Poisson-Boltzmann solvent model based on the Modified Incomplete Cholsky Conjugate Gradient algorithm, which gives rather impressive performance for both static and dynamic systems. This is achieved by implementing the algorithm with Eisenstat's two optimizations, utilizing the electrostatic update in simulations, and applying prudent approxns., including: relaxing the convergence criterion, not updating Poisson-Boltzmann-related forces every step, and using electrostatic focusing. It is also possible to markedly accelerate the supporting routines that are used to set up the calcns. and to obtain energies and forces. The resulting finite difference Poisson-Boltzmann method delivers efficiency comparable to the distance-dependent dielec. model for a system tested, HIV Protease, making it a strong candidate for soln.-phase mol. dynamics simulations. Further, the finite difference method includes all intra-solute electrostatic interactions, whereas the distance dependent dielec. calcns. use a 15-Å cutoff. The speed of our numerical finite difference method is comparable to that of the pair-wise Generalized Born approxn. to the Poisson-Boltzmann method.
- 32Beveridge, D. L.; Dicapua, F. M. Free-energy via molecular simulation - applications to chemical and biomolecular systems Annu. Rev. Biophys. Biophys. Chem. 1989, 18, 431– 492 DOI: 10.1146/annurev.bb.18.060189.002243Google ScholarThere is no corresponding record for this reference.
- 33Bunney, T. D.; Wan, S.; Thiyagarajan, N.; Sutto, L.; Williams, S. V.; Ashford, P.; Koss, H.; Knowles, M. A.; Gervasio, F. L.; Coveney, P. V.; Katan, M. The effect of mutations on drug sensitivity and kinase activity of fibroblast growth factor receptors: A combined experimental and theoretical study EBioMedicine 2015, 2 (3) 194– 204 DOI: 10.1016/j.ebiom.2015.02.009Google ScholarThere is no corresponding record for this reference.
- 34Phillips, J. C.; Braun, R.; Wang, W.; Gumbart, J.; Tajkhorshid, E.; Villa, E.; Chipot, C.; Skeel, R. D.; Kale, L.; Schulten, K. Scalable molecular dynamics with NAMD J. Comput. Chem. 2005, 26 (16) 1781– 1802 DOI: 10.1002/jcc.20289Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1SlsbbJ&md5=189051128443b547f4300a1b8fb0e034Scalable molecular dynamics with NAMDPhillips, James C.; Braun, Rosemary; Wang, Wei; Gumbart, James; Tajkhorshid, Emad; Villa, Elizabeth; Chipot, Christophe; Skeel, Robert D.; Kale, Laxmikant; Schulten, KlausJournal of Computational Chemistry (2005), 26 (16), 1781-1802CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)NAMD is a parallel mol. dynamics code designed for high-performance simulation of large biomol. systems. NAMD scales to hundreds of processors on high-end parallel platforms, as well as tens of processors on low-cost commodity clusters, and also runs on individual desktop and laptop computers. NAMD works with AMBER and CHARMM potential functions, parameters, and file formats. This article, directed to novices as well as experts, first introduces concepts and methods used in the NAMD program, describing the classical mol. dynamics force field, equations of motion, and integration methods along with the efficient electrostatics evaluation algorithms employed and temp. and pressure controls used. Features for steering the simulation across barriers and for calcg. both alchem. and conformational free energy differences are presented. The motivations for and a roadmap to the internal design of NAMD, implemented in C++ and based on Charm++ parallel objects, are outlined. The factors affecting the serial and parallel performance of a simulation are discussed. Finally, typical NAMD use is illustrated with representative applications to a small, a medium, and a large biomol. system, highlighting particular features of NAMD, for example, the Tcl scripting language. The article also provides a list of the key features of NAMD and discusses the benefits of combining NAMD with the mol. graphics/sequence anal. software VMD and the grid computing/collab. software BioCoRE. NAMD is distributed free of charge with source code at www.ks.uiuc.edu.
- 35Sadiq, S. K.; Wright, D. W.; Kenway, O. A.; Coveney, P. V. Accurate ensemble molecular dynamics binding free energy ranking of multidrug-resistant HIV-1 proteases J. Chem. Inf. Model. 2010, 50 (5) 890– 905 DOI: 10.1021/ci100007wGoogle Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksFeqtb4%253D&md5=4a9c06d974aad014a9a5e027a08a76acAccurate Ensemble Molecular Dynamics Binding Free Energy Ranking of Multidrug-Resistant HIV-1 ProteasesSadiq, S. Kashif; Wright, David W.; Kenway, Owain A.; Coveney, Peter V.Journal of Chemical Information and Modeling (2010), 50 (5), 890-905CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Accurate calcn. of important thermodn. properties, such as macromol. binding free energies, is one of the principal goals of mol. dynamics simulations. However, single long simulation frequently produces incorrectly converged quant. results due to inadequate sampling of conformational space in a feasible wall-clock time. Multiple short (ensemble) simulations have been shown to explore conformational space more effectively than single long simulations, but the two methods have not yet been thermodynamically compared. Here we show that, for end-state binding free energy detn. methods, ensemble simulations exhibit significantly enhanced thermodn. sampling over single long simulations and result in accurate and converged relative binding free energies that are reproducible to within 0.5 kcal/mol. Completely correct ranking is obtained for six HIV-1 protease variants bound to lopinavir with a correlation coeff. of 0.89 and a mean relative deviation from expt. of 0.9 kcal/mol. Multidrug resistance to lopinavir is enthalpically driven and increases through a decrease in the protein-ligand van der Waals interaction, principally due to the V82A/I84V mutation, and an increase in net electrostatic repulsion due to water-mediated disruption of protein-ligand interactions in the catalytic region. Furthermore, we correctly rank, to within 1 kcal/mol of expt., the substantially increased chem. potency of lopinavir binding to the wild-type protease compared to saquinavir and show that lopinavir takes advantage of a decreased net electrostatic repulsion to confer enhanced binding. Our approach is dependent on the combined use of petascale computing resources and on an automated simulation workflow to attain the required level of sampling and turn around time to obtain the results, which can be as little as three days. This level of performance promotes integration of such methodol. with clin. decision support systems for the optimization of patient-specific therapy.
- 36Norman, G. E.; Stegailov, V. V. Stochastic theory of the classical molecular dynamics method Math. Models Comput. Simul. 2013, 5 (4) 305– 333 DOI: 10.1134/S2070048213040108Google ScholarThere is no corresponding record for this reference.
- 37Chodera, J. D.; Mobley, D. L. Entropy-enthalpy compensation: role and ramifications in biomolecular ligand recognition and design Annu. Rev. Biophys. 2013, 42, 121– 142 DOI: 10.1146/annurev-biophys-083012-130318Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFGrs7bP&md5=52a30a1d0f4f9ae49128e87d2a23e3faEntropy-enthalpy compensation: role and ramifications in biomolecular ligand recognition and designChodera, John D.; Mobley, David L.Annual Review of Biophysics (2013), 42 (), 121-142CODEN: ARBNCV; ISSN:1936-122X. (Annual Reviews)A review. Recent calorimetric studies of interactions between small mols. and biomol. targets have generated renewed interest in the phenomenon of entropy-enthalpy compensation. In these studies, entropic and enthalpic contributions to binding are obsd. to vary substantially and in an opposing manner as the ligand or protein is modified, whereas the binding free energy varies little. In severe examples, engineered enthalpic gains can lead to completely compensating entropic penalties, frustrating ligand design. Here, we examine the evidence for compensation, as well as its potential origins, prevalence, severity, and ramifications for ligand engineering. We find the evidence for severe compensation to be weak in light of the large magnitude of and correlation between errors in exptl. measurements of entropic and enthalpic contributions to binding, though a limited form of compensation may be common. Given the difficulty of predicting or measuring entropic and enthalpic changes to useful precision, or using this information in design, we recommend ligand engineering efforts instead focus on computational and exptl. methodologies to directly assess changes in binding free energy.
- 38Zhu, Y. L.; Beroza, P.; Artis, D. R. Including explicit water molecules as part of the protein structure in MM/PBSA calculations J. Chem. Inf. Model. 2014, 54 (2) 462– 469 DOI: 10.1021/ci4001794Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXitVWnsb7J&md5=285d8818f326df9f889912aaf0313e7dIncluding Explicit Water Molecules as Part of the Protein Structure in MM/PBSA CalculationsZhu, Yong-Liang; Beroza, Paul; Artis, Dean R.Journal of Chemical Information and Modeling (2014), 54 (2), 462-469CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Water is the natural medium of mols. in the cell and plays an important role in protein structure, function and interaction with small mol. ligands. However, the widely used mol. mechanics Poisson-Boltzmann surface area (MM/PBSA) method for binding energy calcn. does not explicitly take account of water mols. that mediate key protein-ligand interactions. We have developed a protocol to include water mols. that mediate ligand-protein interactions as part of the protein structure in calcn. of MM/PBSA binding energies (a method we refer to as water-MM/PBSA) for a series of JNK3 kinase inhibitors. Improved correlation between water-MM/PBSA binding energies and exptl. IC50 values was obtained compared to that obtained from classical MM/PBSA binding energy. This improved correlation was further validated using sets of neuraminidase and avidin inhibitors. The obsd. improvement, however, appears to be limited to systems in which there are water-mediated ligand-protein hydrogen bond interactions. We conclude that the water-MM/PBSA method performs better than classical MM/PBSA in predicting binding affinities when water mols. play a direct role in mediating ligand-protein hydrogen bond interactions.
- 39Rastelli, G.; Del Rio, A.; Degliesposti, G.; Sgobba, M. Fast and accurate predictions of binding free energies using MM-PBSA and MM-GBSA J. Comput. Chem. 2010, 31 (4) 797– 810 DOI: 10.1002/jcc.21372Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtlenuro%253D&md5=786af541a9e19b7d8c61c210c3aa96b5Fast and accurate predictions of binding free energies using MM-PBSA and MM-GBSARastelli, Giulio; Del Rio, Alberto; Degliesposti, Gianluca; Sgobba, MiriamJournal of Computational Chemistry (2010), 31 (4), 797-810CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)In the drug discovery process, accurate methods of computing the affinity of small mols. with a biol. target are strongly needed. This is particularly true for mol. docking and virtual screening methods, which use approximated scoring functions and struggle in estg. binding energies in correlation with exptl. values. Among the various methods, MM-PBSA (mol. mechanics Poisson-Boltzmann surface area) and MM-GBSA (mol. mechanics-generalized Born surface area) are emerging as useful and effective approaches. Although these methods are typically applied to large collections of equilibrated structures of protein-ligand complexes sampled during mol. dynamics in water, the possibility to reliably est. ligand affinity using a single energy-minimized structure and implicit solvation models has not been explored in sufficient detail. Herein, the authors thoroughly investigate this hypothesis by comparing different methods for the generation of protein-ligand complexes and diverse methods for free energy prediction for their ability to correlate with exptl. values. The methods were tested on a series of structurally diverse inhibitors of Plasmodium falciparum DHFR (dihydrofolate reductase) with known binding mode and measured affinities. The results showed that correlations between MM-PBSA or MM-GBSA binding free energies with exptl. affinities were in most cases excellent. Importantly, the authors found that correlations obtained with the use of a single protein-ligand minimized structure and with implicit solvation models were similar to those obtained after averaging over multiple MD snapshots with explicit water mols., with consequent save of computing time without loss of accuracy. When applied to a virtual screening expt., such an approach proved to discriminate between true binders and decoy mols. and yielded significantly better enrichment curves. © 2009 Wiley Periodicals, Inc. J Comput Chem, 31: 797-810, 2010.
- 40Reynolds, C. H.; Holloway, M. K. Thermodynamics of ligand binding and efficiency ACS Med. Chem. Lett. 2011, 2 (6) 433– 437 DOI: 10.1021/ml200010kGoogle Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXjs1Churk%253D&md5=e44c416392ab548a53d96802d030e12bThermodynamics of Ligand Binding and EfficiencyReynolds, Charles H.; Holloway, M. KatharineACS Medicinal Chemistry Letters (2011), 2 (6), 433-437CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Anal. of the exptl. binding thermodn. for approx. 100 protein-ligand complexes provides important insights into the factors governing ligand affinity and efficiency. The commonly accepted correlation between enthalpy and -TΔS is clearly obsd. for this relatively diverse data set. It is also clear that affinity (i.e., ΔG) is not generally correlated to either enthalpy or -TΔS. This is a worrisome trend since the vast majority of computational structure-based design is carried out using interaction energies for one, or at most a few, ligand poses. As such, these energies are most closely comparable to enthalpies not free energies. Closer inspection of the data shows that in a few cases the enthalpy (or -TΔS) is correlated with free energy. It is tempting to speculate that this could be an important consideration as to why some targets are readily amenable to modeling and others are not. Addnl., anal. of the enthalpy and -TΔS efficiencies shows that the trends obsd. for ligand efficiencies with respect to mol. size are primarily a consequence of enthalpic, not entropic, effects.
Supporting Information
Supporting Information
ARTICLE SECTIONSThe Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jctc.6b00794.
Detailed description of the methods used, additional energetic analyses of the metadynamics, free energy calculation with inclusion of explicit water molecules, alongside the atomic coordinates of the compound-protein complexes and experimental data on compound binding (PDF)
Structures of the studied compounds (ZIP)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.