Evaluation and Characterization of Trk Kinase Inhibitors for the Treatment of Pain: Reliable Binding Affinity Predictions from Theory and Computation
- Shunzhou Wan
- ,
- Agastya P. Bhati
- ,
- Sarah Skerratt
- ,
- Kiyoyuki Omoto
- ,
- Veerabahu Shanmugasundaram
- ,
- Sharan K. Bagal
- , and
- Peter V. Coveney
Abstract

Optimization of ligand binding affinity to the target protein of interest is a primary objective in small-molecule drug discovery. Until now, the prediction of binding affinities by computational methods has not been widely applied in the drug discovery process, mainly because of its lack of accuracy and reproducibility as well as the long turnaround times required to obtain results. Herein we report on a collaborative study that compares tropomyosin receptor kinase A (TrkA) binding affinity predictions using two recently formulated fast computational approaches, namely, Enhanced Sampling of Molecular dynamics with Approximation of Continuum Solvent (ESMACS) and Thermodynamic Integration with Enhanced Sampling (TIES), to experimentally derived TrkA binding affinities for a set of Pfizer pan-Trk compounds. ESMACS gives precise and reproducible results and is applicable to highly diverse sets of compounds. It also provides detailed chemical insight into the nature of ligand–protein binding. TIES can predict and thus optimize more subtle changes in binding affinities between compounds of similar structure. Individual binding affinities were calculated in a few hours, exhibiting good correlations with the experimental data of 0.79 and 0.88 from the ESMACS and TIES approaches, respectively. The speed, level of accuracy, and precision of the calculations are such that the affinity predictions can be used to rapidly explain the effects of compound modifications on TrkA binding affinity. The methods could therefore be used as tools to guide lead optimization efforts across multiple prospective structurally enabled programs in the drug discovery setting for a wide range of compounds and targets.
Introduction
Computational Methods


All of the ligands have the same net neutral charge. The experimental TrkA inhibitory values (IC50) and the binding free energies derived from them are shown. Experimental IC50 measurements were conducted independently in two separate laboratories using an identical protocol; (2) Pfizer, Sandwich (U.K.) IC50 values are shown in black; TCG Lifescience (India) IC50 values are shown in blue.



Results
Figure 1

Figure 1. Crystal structure of 1 bound to TrkA, viewed from the N-lobe to the C-lobe of the kinase. Hydrogen bonds are displayed by dashed lines. The protein is shown in cyan cartoon, and ligand atoms are colored by element: hydrogen in white, carbon in cyan, oxygen in red, and nitrogen in blue. For clarity, the N-lobe is not shown.
Comparison of Experimental TrkA Binding Free Energies with ESMACS Predictions
Figure 2

Figure 2. Comparisons of the TrkA experimental data and the calculated binding free energies from (a) 1-traj, (b) 2-traj, and (c) 3-traj ESMACS approaches. The equation in each panel indicates the calculations used in each case. The term Gcom/rec/ligcom/lig represents the free energy of the component (subscript: complex, receptor, or ligand) obtained from simulation of the same or different component (superscript: complex or free ligand in aqueous solution). The calculated binding free energies are associated with standard errors of ca. 0.6 kcal/mol in the 1-traj approach and ca. 3 kcal/mol in the 2- and 3-traj approaches (see Table S2), which are not shown in the figures for reasons of clarity. The experimental data from two sites (Pfizer, Sandwich and TCG Lifescience) are displayed in black and red, respectively. The correlation coefficients shown in the figure were calculated using the averages of the calculated binding free energies and the experimental data from Pfizer, Sandwich (black circles) and TCG Lifescience (red circles) where the former are not available. Large uncertainties are associated with the correlation coefficients because of the large error bars of the calculated and experimental binding affinities. Further analyses with bootstrapping resampling (see details in the Supporting Information) generate correlation coefficients of 0.39 ± 0.26, 0.60 ± 0.26, and 0.62 ± 0.27 for the 1-, 2-, and 3-traj approaches, respectively. It is evident that the 2- and 3-traj methods improve the ranking provided by the 1-traj method.
Improvement of Docking Predictions by the ESMACS Approach
Figure 3

Figure 3. Calculated binding free energies for 1, 3, 4, 6, 7, 8, 13, 16, and 22 (a) from single structures after docking, (b) from 25 structures for each compound after 11 000-step minimization with a sophisticated conjugate-gradient method, and (c) from ensemble averages from three-trajectory ESMACS studies. Compounds 1, 3, 4, 6, 7, 8, 13, 16, and 22 are highly structurally similar (see Table 1).
ESMACS Binding Affinities: Contributions from Energy Components
Figure 4

Figure 4. Correlations of free energy components and the experimental data from the 3-traj approach. Both (a) bonded and (b) nonbonded energy terms contribute to the ranking of binding affinities, with similar correlation coefficients between the calculations and experimental data. Their combination, the MMPBSA energy (c), exhibits better correlations with the experimental data than the components themselves.
Adaptation Energy: A Measure of Conformational Change upon Binding
Figure 5

Figure 5. Adaptation free energies of (a) the receptor and (b) the compounds showing the binding free energy changes between the 1- and 3-traj approaches. The terms Grec/ligcom/lig are the free energies of receptor or ligands (subscript) calculated from simulations performed for the complex or the ligands (superscript).
Comparison of Experimental TrkA Binding Free Energies with TIES Predictions
Figure 6

Figure 6. Correlation between TIES-predicted relative binding affinities and experimental data. The black line is the correlation line, while the dotted lines (x = 0 and y = 0) create four quadrants. Ten out of the 14 data points are in quadrants I (x > 0 and y > 0) and III (x < 0 and y < 0), meaning that the calculated binding free energy differences have the same sign as those from the experimental data.
Discussion
Hinge Binding Group Modifications: Compounds 1, 4, and 22

TrkA free energy activities derived from experimental IC50 values are denoted as ΔGexp. The relative binding free energies from experiment and ESMACS and TIES calculations are denoted as ΔΔGexp, ΔΔGESMACS, and ΔΔGTIES, respectively. Experimental data from Pfizer, Sandwich (U.K.) are shown in black, and those from TCG Lifescience (India) are shown in blue.
Figure 7

Figure 7. ESMACS simulations of (a) 1, (b) 22, and (c) 4 bound to TrkA. Final conformations of 4 ns production runs from all 25 replicas are overlapped and smoothed by averaging over 10 frames. Key residues E590 and M592 (for clarity, side-chain atoms are not shown) at the TrkA hinge region are highlighted. Hydrogen bonds are shown as dashed lines. The protein is shown in cyan cartoon, and ligand atoms are colored by element: hydrogen in white, carbon in cyan, oxygen in red, and nitrogen in blue. For reasons of clarity, the N-lobe of the protein has been removed.
Hinge Binding Group Modifications: Compounds 12, 17, 23, and 24

TrkA free energy activities derived from experimental IC50 values are denoted as ΔGexp. The relative binding free energies from experiment and ESMACS and TIES calculations are denoted as ΔΔGexp, ΔΔGESMACS, and ΔΔGTIES, respectively. Experimental data from Pfizer, Sandwich (U.K.) are shown in black, and those from TCG Lifescience (India) are shown in blue.
Figure 8

Figure 8. ESMACS simulations of (a) 12, (b) 17, (c) 23, and (d) 24 bound to TrkA. Representative conformations are displayed using the final conformations of 4 ns production runs of one replica from each 25-member ensemble. The protein is shown by the surface representation and the ligand by the ball-and-stick representation with hydrogen in white, carbon in cyan, oxygen in red, and nitrogen in blue. The surfaces of the R2-position groups are shown in the wireframe representation.
Linker Group Modifications: Compounds 1, 4, and 22

TrkA free energy activities derived from experimental IC50 values are denoted as ΔGexp. The relative binding free energies from experiment and ESMACS and TIES calculations are denoted as ΔΔGexp, ΔΔGESMACS, and ΔΔGTIES, respectively. Experimental data from Pfizer, Sandwich (U.K.) are shown in black, and those from TCG Lifescience (India) are shown in blue.
Figure 9

Figure 9. ESMACS simulations of (a) 1, (b) 3, and (c) 6 bound to TrkA. Final conformations of 4 ns production runs from all 25 replicas are overlapped and smoothed by averaging over 10 frames. The key TrkA protein residues D668, K544, and E560 are highlighted. The protein is shown in cyan cartoon, and ligand atoms are colored by element: hydrogen in white, carbon in cyan, oxygen in red, and nitrogen in blue. Hydrogen bonds between the linker group carbonyl oxygen of 1, 3, and 6 and the backbone −NH group of D668 are shown as dashed lines.
Linker Group Modifications: Compounds 7 and 8

TrkA free energy activities derived from experimental IC50 values are denoted as ΔGexp. The relative binding free energies from experiment and ESMACS and TIES calculations are denoted as ΔΔGexp, ΔΔGESMACS, and ΔΔGTIES, respectively. Experimental data were obtained from Pfizer, Sandwich (U.K.) for these two compounds.
Figure 10

Figure 10. ESMACS simulations of (a) 7 and (b) 8 bound to TrkA. Final conformations of 4 ns production runs from all 25 replicas are overlapped and smoothed by averaging over 10 frames. The key TrkA protein residues D668, E560, and L564 are highlighted. The protein is shown in cyan cartoon, and ligand atoms are colored by element: hydrogen in white, carbon in cyan, oxygen in red, and nitrogen in blue. The unfavorable electrostatic interactions between the methoxy group of 8 and the carbonyl group of D668 and between the methoxy group and the carboxylate group of E560 are highlighted by pink arrows.
Linker Group Modifications: Compounds 4 and 16

TrkA free energy activities derived from experimental IC50 values are denoted as ΔGexp. The relative binding free energies from experiment and ESMACS and TIES calculations are denoted as ΔΔGexp, ΔΔGESMACS, and ΔΔGTIES, respectively. Experimental data were obtained from Pfizer, Sandwich (U.K.) for these two compounds.
Figure 11

Figure 11. ESMACS simulations of (a) 4 and (b) 16 bound to TrkA. Final conformations of 4 ns production runs from all 25 replicas are overlapped and smoothed by averaging over 10 conformations. The key TrkA protein residue D668 is highlighted. The protein is shown in cyan cartoon, and ligand atoms are colored by element: hydrogen in white, carbon in cyan, oxygen in red, and nitrogen in blue. The hydrogen bond between the linker-group carbonyl oxygen of 4 and the backbone −NH group of D668 is shown as a dashed line. As can be seen in (b), switching of the N–H and C═O groups prevents the formation of a hydrogen bond between the carbonyl oxygen of 16 and the backbone −NH group of D668.
Conclusions
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jcim.6b00780.
Detailed description of the energy decomposition, the energy convergence, and the error analyses, experimental measurements of TrkA inhibitory activity, and the predicted binding free energies (PDF)
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.
Acknowledgment
The authors acknowledge the support from the EPSRC via the 2020 Science Programme (http://www.2020science.net/, EP/I017909/1), the EU H2020 Projects ComPat (http://www.compat-project.eu/, 671564) and CompBioMed (http://www.compbiomed.eu/, 675451), the Qatar National Research Fund (7-1083-1-191), the MRC Medical Bioinformatics Project (MR/L016311/1), and the UCL Provost. We are grateful to the Hartree Centre for access to the BlueWonder2 computer and for the help of its scientific support staff. We acknowledge the Leibniz Supercomputing Centre for providing access to SuperMUC and the assistance of its scientific support staff. We also made use of ARCHER, the U.K.’s National High Performance Computing Service, funded by the Office of Science and Technology through the EPSRC’s High-End Computing Programme. Access to ARCHER was provided through the 2020 Science Programme. This research was also partially supported by the PLGrid Infrastructure, through which access to Prometheus, the Polish supercomputer run by ACK Cyfronet AGH in Krakow, was provided. A.P.B. is supported by an Overseas Research Scholarship from UCL and an Inlaks Scholarship from the Inlaks Shivdasani Foundation.
References
This article references 33 other publications.
- 1Sherborne, B.; Shanmugasundaram, V.; Cheng, A. C.; Christ, C. D.; DesJarlais, R. L.; Duca, J. S.; Lewis, R. A.; Loughney, D. A.; Manas, E. S.; McGaughey, G. B.; Peishoff, C. E.; van Vlijmen, H. Collaborating to Improve the Use of Free-Energy and Other Quantitative Methods in Drug Discovery J. Comput.-Aided Mol. Des. 2016, 30, 1139– 1141 DOI: 10.1007/s10822-016-9996-yGoogle Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFClsLfN&md5=c00386f775bd826c6b845bbbef4a975dCollaborating to improve the use of free-energy and other quantitative methods in drug discoverySherborne, Bradley; Shanmugasundaram, Veerabahu; Cheng, Alan C.; Christ, Clara D.; DesJarlais, Renee L.; Duca, Jose S.; Lewis, Richard A.; Loughney, Deborah A.; Manas, Eric S.; McGaughey, Georgia B.; Peishoff, Catherine E.; van Vlijmen, HermanJournal of Computer-Aided Molecular Design (2016), 30 (12), 1139-1141CODEN: JCADEQ; ISSN:0920-654X. (Springer)In May and August, 2016, several pharmaceutical companies convened to discuss and compare experiences with Free Energy Perturbation (FEP). This unusual synchronization of interest was prompted by Schrodinger's FEP+ implementation and offered the opportunity to share fresh studies with FEP and enable broader discussions on the topic. This article summarizes key conclusions of the meetings, including a path forward of actions for this group to aid the accelerated evaluation, application and development of free energy and related quant., structure-based design methods.
- 2Skerratt, S. E.; Andrews, M.; Bagal, S. K.; Bilsland, J.; Brown, D.; Bungay, P. J.; Cole, S.; Gibson, K. R.; Jones, R.; Morao, I.; Nedderman, A.; Omoto, K.; Robinson, C.; Ryckmans, T.; Skinner, K.; Stupple, P.; Waldron, G. The Discovery of a Potent, Selective, and Peripherally Restricted Pan-Trk Inhibitor (PF-06273340) for the Treatment of Pain J. Med. Chem. 2016, 59, 10084– 10099 DOI: 10.1021/acs.jmedchem.6b00850Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslWjsbfL&md5=f8c904a92c1716062c217fefe2374ce6The Discovery of a Potent, Selective, and Peripherally Restricted Pan-Trk Inhibitor (PF-06273340) for the Treatment of PainSkerratt, Sarah E.; Andrews, Mark; Bagal, Sharan K.; Bilsland, James; Brown, David; Bungay, Peter J.; Cole, Susan; Gibson, Karl R.; Jones, Russell; Morao, Inaki; Nedderman, Angus; Omoto, Kiyoyuki; Robinson, Colin; Ryckmans, Thomas; Skinner, Kimberly; Stupple, Paul; Waldron, GarethJournal of Medicinal Chemistry (2016), 59 (22), 10084-10099CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The neurotrophin family of growth factors, comprised of nerve growth factor (NGF), brain derived neurotrophic factor (BDNF), neurotrophin 3 (NT3) and neurotrophin 4 (NT4), is implicated in the physiol. of chronic pain. Given the clin. efficacy of anti-NGF monoclonal antibody (mAb) therapies, there is significant interest in the development of small mol. modulators of neurotrophin activity. Neurotrophins signal through the tropomyosin related kinase (Trk) family of tyrosine kinase receptors, hence Trk kinase inhibition represents a potentially "druggable" point of intervention. To deliver the safety profile required for chronic, non-life threatening pain indications, highly kinase-selective Trk inhibitors with minimal brain availability are sought. Herein the authors describe how the use of SBDD, 2D QSAR models and Matched Mol. Pair data in compd. design enabled the delivery of the highly potent, kinase-selective and peripherally restricted clin. candidate PF-06273340.
- 3Sadiq, 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, 1909– 1919 DOI: 10.1021/ci8000937Google Scholar3https://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.
- 4Groen, 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 Scholar4https://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.
- 5Norman, B. H.; McDermott, J. S. Targeting the Nerve Growth Factor (NGF) Pathway in Drug Discovery. Potential Applications to New Therapies for Chronic Pain J. Med. Chem. 2017, 60, 66– 88 DOI: 10.1021/acs.jmedchem.6b00964Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslSqtLjF&md5=606097686779a102db9e6e54f8252a40Targeting the Nerve Growth Factor (NGF) Pathway in Drug Discovery. Potential Applications to New Therapies for Chronic PainNorman, Bryan H.; McDermott, Jeff S.Journal of Medicinal Chemistry (2017), 60 (1), 66-88CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. The neurotrophin nerve growth factor (NGF) has been implicated as a key mediator of chronic pain. NGF binds the tropomyosin receptor kinase A (TrkA) and p75, resulting in the activation of downstream signaling pathways that have been linked to pro-nociception. While anti-NGF antibodies have demonstrated analgesia both preclinically and in patients, the mechanism of action of these agents remains unclear. The authors describe ligands targeting NGF, its receptors and downstream/related targets. This perspective highlights large and small mol. approaches to targeting the NGF-TrkA pathway both extra- and intracellularly. In addn., the authors present a strategic framework for future drug discovery efforts in this pathway beyond the targeting of NGF or its receptors. While existing tools have greatly informed NGF-mediated signaling, ongoing and future pathway research may help focus new drug discovery efforts on key novel targets and mechanisms. This may result in highly differentiated therapeutics with greater efficacy and/or improved safety profiles.
- 6Hefti, F. F.; Rosenthal, A.; Walicke, P. A.; Wyatt, S.; Vergara, G.; Shelton, D. L.; Davies, A. M. Novel Class of Pain Drugs Based on Antagonism of NGF Trends Pharmacol. Sci. 2006, 27, 85– 91 DOI: 10.1016/j.tips.2005.12.001Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtV2lsLo%253D&md5=3cfe9e06069c3b6b9aa9d96b7411d199Novel class of pain drugs based on antagonism of NGFHefti, Franz F.; Rosenthal, Arnon; Walicke, Patricia A.; Wyatt, Sean; Vergara, German; Shelton, David L.; Davies, Alun M.Trends in Pharmacological Sciences (2006), 27 (2), 85-91CODEN: TPHSDY; ISSN:0165-6147. (Elsevier Ltd.)A review. Nerve growth factor (NGF) was identified originally as a survival factor for sensory and sympathetic neurons in the developing nervous system. In adults, NGF is not required for survival but it has a crucial role in the generation of pain and hyperalgesia in several acute and chronic pain states. The expression of NGF is high in injured and inflamed tissues, and activation of the NGF receptor tyrosine kinase trkA on nociceptive neurons triggers and potentiates pain signaling by multiple mechanisms. Inhibition of NGF function and signaling blocks pain sensation as effectively as cyclooxygenase inhibitors and opiates in rodent models of pain. Several pharmaceutical companies have active drug-discovery and development programs that are based on a variety of approaches to antagonize NGF, including NGF capture', blocking the binding of NGF to trkA and inhibiting trkA signaling. NGF antagonism is expected to be a highly effective therapeutic approach in many pain states, and to be free of the adverse effects of traditional analgesic drugs.
- 7Mantyh, P. W.; Koltzenburg, M.; Mendell, L. M.; Tive, L.; Shelton, D. L. Antagonism of Nerve Growth Factor-TrkA Signaling and the Relief of Pain Anesthesiology 2011, 115, 189– 204 DOI: 10.1097/ALN.0b013e31821b1ac5Google ScholarThere is no corresponding record for this reference.
- 8Wan, 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, 3346– 3356 DOI: 10.1021/acs.jctc.5b00179Google Scholar8https://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.
- 9Kollman, 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, 889– 897 DOI: 10.1021/ar000033jGoogle Scholar9https://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.
- 10Coveney, P. V.; Wan, S. On the Calculation of Equilibrium Thermodynamic Properties from Molecular Dynamics Phys. Chem. Chem. Phys. 2016, 18, 30236– 30240 DOI: 10.1039/C6CP02349EGoogle Scholar10https://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.
- 11Miller, B. R., 3rd; McGee, T. D., Jr.; Swails, J. M.; Homeyer, N.; Gohlke, H.; Roitberg, A. E. Mmpbsa.Py: An Efficient Program for End-State Free Energy Calculations J. Chem. Theory Comput. 2012, 8, 3314– 3321 DOI: 10.1021/ct300418hGoogle Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtV2gtrzP&md5=cc4148bd8f70c7cad94fd3ec6f580e52MMPBSA.py: An Efficient Program for End-State Free Energy CalculationsMiller, Bill R., III; McGee, T. Dwight, Jr.; Swails, Jason M.; Homeyer, Nadine; Gohlke, Holger; Roitberg, Adrian E.Journal of Chemical Theory and Computation (2012), 8 (9), 3314-3321CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)MM-PBSA is a post-processing end-state method to calc. free energies of mols. in soln. MMPBSA.py is a program written in Python for streamlining end-state free energy calcns. using ensembles derived from mol. dynamics (MD) or Monte Carlo (MC) simulations. Several implicit solvation models are available with MMPBSA.py, including the Poisson-Boltzmann Model, the Generalized Born Model, and the Ref. Interaction Site Model. Vibrational frequencies may be calcd. using normal mode or quasi-harmonic anal. to approx. the solute entropy. Specific interactions can also be dissected using free energy decompn. or alanine scanning. A parallel implementation significantly speeds up the calcn. by dividing frames evenly across available processors. MMPBSA.py is an efficient, user-friendly program with the flexibility to accommodate the needs of users performing end-state free energy calcns. The source code can be downloaded at http://ambermd.org/ with AmberTools, released under the GNU General Public License.
- 12Case, D. A.; Cheatham, T. E., III; 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, 1668– 1688 DOI: 10.1002/jcc.20290Google Scholar12https://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.
- 13Bhati, 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. 2017, 13, 210– 222 DOI: 10.1021/acs.jctc.6b00979Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitVGntrrJ&md5=510b70188112a3578030e291ce19b127Rapid, Accurate, Precise, and Reliable Relative Free Energy Prediction Using Ensemble Based Thermodynamic IntegrationBhati, Agastya P.; Wan, Shunzhou; Wright, David W.; Coveney, Peter V.Journal of Chemical Theory and Computation (2017), 13 (1), 210-222CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The accurate prediction of the binding affinities of ligands to proteins is a major goal in drug discovery and personalized medicine. The time taken to make such predictions is of similar importance to their accuracy, precision and reliability. In the last few years, an ensemble based mol. dynamics approach has been proposed that provides a route to reliable predictions of free energies based on the mol. mechanics Poisson-Boltzmann surface area method which meets the requirements of accuracy, precision and reliability. Here, we describe an equiv. methodol. based on thermodn. integration to substantially improve the accuracy, precision and reliability of calcd. relative binding free energies. We report the performance of the method when applied to a diverse set of protein targets and ligands. The results are in very good agreement with exptl. data (90% of calcns. agree to within 1 kcal/mol) while the method is reproducible by construction. Statistical uncertainties of the order of 0.5 kcal/mol or less are achieved. We present a systematic account of how the uncertainty in the predictions may be estd.
- 14Wright, 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, 1228– 1241 DOI: 10.1021/ct4007037Google Scholar14https://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.
- 15Swanson, 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 Scholar15https://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.
- 16Chodera, J. D.; Mobley, D. L.; Shirts, M. R.; Dixon, R. W.; Branson, K.; Pande, V. S. Alchemical Free Energy Methods for Drug Discovery: Progress and Challenges Curr. Opin. Struct. Biol. 2011, 21, 150– 160 DOI: 10.1016/j.sbi.2011.01.011Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXkt1Kisrk%253D&md5=fa17c0982d921cd1c32cd095fd34420eAlchemical free energy methods for drug discovery: Progress and challengesChodera, John D.; Mobley, David L.; Shirts, Michael R.; Dixon, Richard W.; Branson, Kim; Pande, Vijay S.Current Opinion in Structural Biology (2011), 21 (2), 150-160CODEN: COSBEF; ISSN:0959-440X. (Elsevier Ltd.)A review. Improved rational drug design methods are needed to lower the cost and increase the success rate of drug discovery and development. Alchem. binding free energy calcns., one potential tool for rational design, have progressed rapidly over the past decade, but still fall short of providing robust tools for pharmaceutical engineering. Recent studies, esp. on model receptor systems, have clarified many of the challenges that must be overcome for robust predictions of binding affinity to be useful in rational design. In this review, inspired by a recent joint academic/industry meeting organized by the authors, we discuss these challenges and suggest a no. of promising approaches for overcoming them.
- 17Wang, 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, 2695– 2703 DOI: 10.1021/ja512751qGoogle Scholar17https://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.
- 18Frisch, M. J.; Trucks, G. W.; Schlegel, H. B.; Scuseria, G. E.; Robb, M. A.; Cheeseman, J. R.; Montgomery, J. J.; 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.
- 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, 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.
- 20Lindorff-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., Bioinf. 2010, 78, 1950– 1958 DOI: 10.1002/prot.22711Google Scholar20https://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.
- 21Phillips, 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, 1781– 1802 DOI: 10.1002/jcc.20289Google Scholar21https://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.
- 22Highfield, R. Supercomputer bid to create the first truly personalised medicine. https://blog.sciencemuseum.org.uk/supercomputer-bid-to-create-the-first-truly-personalised-medicine/ (accessed Feb 22, 2017).Google ScholarThere is no corresponding record for this reference.
- 23Fiser, A.; Sali, A. Modloop: Automated Modeling of Loops in Protein Structures Bioinformatics 2003, 19, 2500– 2501 DOI: 10.1093/bioinformatics/btg362Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXpvVSisLk%253D&md5=4b0fdaca0a412715682496883ab2019cModLoop: automated modeling of loops in protein structuresFiser, Andras; Sali, AndrejBioinformatics (2003), 19 (18), 2500-2501CODEN: BOINFP; ISSN:1367-4803. (Oxford University Press)Summary: ModLoop is a web server for automated modeling of loops in protein structures. The input is the at. coordinates of the protein structure in the Protein Data Bank format, and the specification of the starting and ending residues of one or more segments to be modeled, contg. no more than 20 residues in total. The output is the coordinates of the non-hydrogen atoms in the modeled segments. A user provides the input to the server via a simple web interface, and receives the output by e-mail. The server relies on the loop modeling routine in MODELLER that predicts the loop conformations by satisfaction of spatial restraints, without relying on a database of known protein structures. For a rapid response, ModLoop runs on a cluster of Linux PC computers.
- 24Allen, W. J.; Balius, T. E.; Mukherjee, S.; Brozell, S. R.; Moustakas, D. T.; Lang, P. T.; Case, D. A.; Kuntz, I. D.; Rizzo, R. C. Dock 6: Impact of New Features and Current Docking Performance J. Comput. Chem. 2015, 36, 1132– 1156 DOI: 10.1002/jcc.23905Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXnt1Smtbs%253D&md5=8b5fc6cc7533f975e1e740acb3688807DOCK 6: Impact of new features and current docking performanceAllen, William J.; Balius, Trent E.; Mukherjee, Sudipto; Brozell, Scott R.; Moustakas, Demetri T.; Lang, P. Therese; Case, David A.; Kuntz, Irwin D.; Rizzo, Robert C.Journal of Computational Chemistry (2015), 36 (15), 1132-1156CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)This manuscript presents the latest algorithmic and methodol. developments to the structure-based design program DOCK 6.7 focused on an updated internal energy function, new anchor selection control, enhanced minimization options, a footprint similarity scoring function, a symmetry-cor. root-mean-square deviation algorithm, a database filter, and docking forensic tools. An important strategy during development involved use of three orthogonal metrics for assessment and validation: pose reprodn. over a large database of 1043 protein-ligand complexes (SB2012 test set), cross-docking to 24 drug-target protein families, and database enrichment using large active and decoy datasets (Directory of Useful Decoys [DUD]-E test set) for five important proteins including HIV protease and IGF-1R. Relative to earlier versions, a key outcome of the work is a significant increase in pose reprodn. success in going from DOCK 4.0.2 (51.4%) → 5.4 (65.2%) → 6.7 (73.3%) as a result of significant decreases in failure arising from both sampling 24.1% → 13.6% → 9.1% and scoring 24.4% → 21.1% → 17.5%. Companion cross-docking and enrichment studies with the new version highlight other strengths and remaining areas for improvement, esp. for systems contg. metal ions. The source code for DOCK 6.7 is available for download and free for academic users at. © 2015 Wiley Periodicals, Inc.
- 25Chodera, 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 Scholar25https://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.
- 26Bissantz, C.; Folkers, G.; Rognan, D. Protein-Based Virtual Screening of Chemical Databases. 1. Evaluation of Different Docking/Scoring Combinations J. Med. Chem. 2000, 43, 4759– 4767 DOI: 10.1021/jm001044lGoogle Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXotFymurY%253D&md5=ee74ac9a99e55759c2df27fd1db58010Protein-Based Virtual Screening of Chemical Databases. 1. Evaluation of Different Docking/Scoring CombinationsBissantz, Caterina; Folkers, Gerd; Rognan, DidierJournal of Medicinal Chemistry (2000), 43 (25), 4759-4767CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Three different database docking programs (Dock, FlexX, Gold) have been used in combination with seven scoring functions (Chemscore, Dock, FlexX, Fresno, Gold, Pmf, Score) to assess the accuracy of virtual screening methods against two protein targets (thymidine kinase, estrogen receptor) of known three-dimensional structure. For both targets, it was generally possible to discriminate about 7 out of 10 true hits from a random database of 990 ligands. The use of consensus lists common to two or three scoring functions clearly enhances hit rates among the top 5% scorers from 10% (single scoring) to 25-40% (double scoring) and up to 65-70% (triple scoring). However, in all tested cases, no clear relationships could be found between docking and ranking accuracies. Moreover, predicting the abs. binding free energy of true hits was not possible whatever docking accuracy was achieved and scoring function used. As the best docking/consensus scoring combination varies with the selected target and the physicochem. of target-ligand interactions, we propose a two-step protocol for screening large databases: (i) screening of a reduced dataset contg. a few known ligands for deriving the optimal docking/consensus scoring scheme, (ii) applying the latter parameters to the screening of the entire database.
- 27Gohlke, H.; Kiel, C.; Case, D. A. Insights into Protein-Protein Binding by Binding Free Energy Calculation and Free Energy Decomposition for the Ras-Raf and Ras-RaiGDS Complexes J. Mol. Biol. 2003, 330, 891– 913 DOI: 10.1016/S0022-2836(03)00610-7Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXlt1aqtbs%253D&md5=0a8acf22e053534f34365f264ed4192aInsights into protein-protein binding by binding free energy calculation and free energy decomposition for the Ras-Raf and Ras-RalGDS complexesGohlke, Holger; Kiel, Christina; Case, David A.Journal of Molecular Biology (2003), 330 (4), 891-913CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Science Ltd.)Abs. binding free energy calcns. and free energy decompns. are presented for the protein-protein complexes H-Ras/C-Raf1 and H-Ras/RalGDS. Ras is a central switch in the regulation of cell proliferation and differentiation. In our study, we investigate the capability of the mol. mechanics (MM)-generalized Born surface area (GBSA) approach to est. abs. binding free energies for the protein-protein complexes. Averaging gas-phase energies, solvation free energies, and entropic contributions over snapshots extd. from trajectories of the unbound proteins and the complexes, calcd. binding free energies (Ras-Raf: -15.0(±6.3) kcal mol-1; Ras-RalGDS: -19.5(±5.9) kcal mol-1) are in fair agreement with exptl. detd. values (-9.6 kcal mol-1; -8.4 kcal mol-1), if appropriate ionic strength is taken into account. Structural determinants of the binding affinity of Ras-Raf and Ras-RalGDS are identified by means of free energy decompn. For the first time, computationally inexpensive generalized Born (GB) calcns. are applied in this context to partition solvation free energies along with gas-phase energies between residues of both binding partners. For selected residues, in addn., entropic contributions are estd. by classical statistical mechanics. Comparison of the decompn. results with exptl. detd. binding free energy differences for alanine mutants of interface residues yielded correlations with r2=0.55 and 0.46 for Ras-Raf and Ras-RalGDS, resp. Extension of the decompn. reveals residues as far apart as 25 A from the binding epitope that can contribute significantly to binding free energy. These "hotspots" are found to show large at. fluctuations in the unbound proteins, indicating that they reside in structurally less stable regions. Furthermore, hotspot residues experience a significantly larger-than-av. decrease in local fluctuations upon complex formation. Finally, by calcg. a pair-wise decompn. of interactions, interaction pathways originating in the binding epitope of Raf are found that protrude through the protein structure towards the loop L1. This explains the finding of a conformational change in this region upon complex formation with Ras, and it may trigger a larger structural change in Raf, which is considered to be necessary for activation of the effector by Ras.
- 28Bunney, 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, 194– 204 DOI: 10.1016/j.ebiom.2015.02.009Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2srotlKmtA%253D%253D&md5=8414f3530d24bbba0ea8cc98cb784b96The Effect of Mutations on Drug Sensitivity and Kinase Activity of Fibroblast Growth Factor Receptors: A Combined Experimental and Theoretical StudyBunney Tom D; Thiyagarajan Nethaji; Ashford Paul; Katan Matilda; Wan Shunzhou; Coveney Peter V; Sutto Ludovico; Gervasio Francesco L; Williams Sarah V; Knowles Margaret A; Koss HansEBioMedicine (2015), 2 (3), 194-204 ISSN:.Fibroblast growth factor receptors (FGFRs) are recognized therapeutic targets in cancer. We here describe insights underpinning the impact of mutations on FGFR1 and FGFR3 kinase activity and drug efficacy, using a combination of computational calculations and experimental approaches including cellular studies, X-ray crystallography and biophysical and biochemical measurements. Our findings reveal that some of the tested compounds, in particular TKI258, could provide therapeutic opportunity not only for patients with primary alterations in FGFR but also for acquired resistance due to the gatekeeper mutation. The accuracy of the computational methodologies applied here shows a potential for their wider application in studies of drug binding and in assessments of functional and mechanistic impacts of mutations, thus assisting efforts in precision medicine.
- 29Andrews, M. D.; Bagal, S. K.; Gibson, K. R.; Omoto, K.; Ryckmans, T.; Skerratt, S. E.; Stupple, P. A. Pyrrolo[2,3-d]pyrimidine Derivatives as Inhibitors of Tropomyosin-Related Kinases and Their Preparation and Use in the Treatment of Pain. WO2012137089A1, 2012.Google ScholarThere is no corresponding record for this reference.
- 30Xing, L.; Klug-Mcleod, J.; Rai, B.; Lunney, E. A. Kinase Hinge Binding Scaffolds and Their Hydrogen Bond Patterns Bioorg. Med. Chem. 2015, 23, 6520– 6527 DOI: 10.1016/j.bmc.2015.08.006Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVentr3L&md5=048b4b99628c6e1ae273b00a47ab2377Kinase hinge binding scaffolds and their hydrogen bond patternsXing, Li; Klug-Mcleod, Jacquelyn; Rai, Brajesh; Lunney, Elizabeth A.Bioorganic & Medicinal Chemistry (2015), 23 (19), 6520-6527CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Protein kinases constitute a major class of intracellular signaling mols., and describe some of the most prominent drug targets. Kinase inhibitors commonly employ small chem. scaffolds that form hydrogen bonds with the kinase hinge residues connecting the N- and C-terminal lobes of the catalytic domain. In general the satisfied hydrogen bonds are required for potent inhibition, therefore constituting a conserved feature in the majority of inhibitor-kinase interactions. From systematically analyzing the kinase scaffolds extd. from Pfizer crystal structure database (CSDb) the authors recognize that large no. of kinase inhibitors of diverse chem. structures are derived from a relatively small no. of common scaffolds. Depending on specific substitution patterns, scaffolds may demonstrate versatile binding capacities to interact with kinase hinge. Afforded by thousands of ligand-protein binary complexes, the hinge hydrogen bond patterns were analyzed with a focus on their three-dimensional configurations. Most of the compds. engage H6 NH for hinge recognition. Dual hydrogen bonds are commonly obsd. with addnl. recruitment of H4 CO upstream and/or H6 CO downstream. Triple hydrogen bonds accounts for small no. of binary complexes. An unusual hydrogen bond with a non-canonical H5 conformation is obsd., requiring a peptide bond flip by a glycine residue at the H6 position. Addnl. hydrogen bonds to kinase hinge do not necessarily correlate with an increase in potency; conversely they appear to compromise kinase selectivity. Such learnings could enhance the prospect of successful therapy design.
- 31Morphy, R. Selectively Nonselective Kinase Inhibition: Striking the Right Balance J. Med. Chem. 2010, 53, 1413– 1437 DOI: 10.1021/jm901132vGoogle Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtlWmtbfF&md5=3c915b5debf9d005993c8ad31c68bc2eSelectively Nonselective Kinase Inhibition: Striking the Right BalanceMorphy, RichardJournal of Medicinal Chemistry (2010), 53 (4), 1413-1437CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Marketed kinase inhibitors (MKIs) can deliver superior efficacy compared to inhibitors with high specificity for a single kinase, and the recent introduction of several MKIs to the market opens the door to a new era of safer and effective anticancer therapy. The key to combining high efficacy with acceptable safety is to inhibit multiple targets in a selectively nonselective fashion. Strategies for intentionally designing MKIs are emerging, but the field is still in its infancy and we are as medicinal chemists currently on the steepest part of the learning curve. MTDD can be time-consuming and expensive, and we need to become more proficient first at identifying disease-relevant target combinations and second at discovering MKIs that combine optimal physicochem. and biol. properties. Bold and innovative medicinal chem. strategies are required to tackle "difficult combinations" where the disease rationale is compelling but where it is a struggle to combine all the desired attributes of an oral MKI drug into a single mol. At present it is unclear to what extent MKIs with highly tuned selectivity profiles can be rationally designed, particularly for targets that are unrelated by sequence. In addn. to the well-known selectivity challenge, the physicochem. property profiles of AT P-competitive MKIs can be inherently challenging and limited scope for patentability can also be a serious hindrance. On the plus side, the amt. of kinase-specific structural information is growing very rapidly, and ultimately this may reveal distinct features and design rules that enable a medicinal chemist to rationally modify and refine the profile of MKIs. In addn., increasing SAR knowledge is emerging from large scale panel screening with the binding profiles starting to reveal to medicinal chemists how chem. structure affects cross-reactivity across large parts of the kinome. The merit of MKIs compared with single kinase inhibitors is a subject of controversy in drug discovery that is unlikely to be resolved in the near future. At the start of a new MTDD project, a rigorous debate needs to take place as to whether it makes more sense to seek a combination of highly selective agents or a DML. Many factors need to be taken into account in this decision such as the no., similarity, and promiscuity of the targets in the profile and the disease area. Conformational plasticity and the occurrence of multiple binding modes complicate the in silico prediction of kinase polypharmacol. based solely upon protein structure. The use of ligand-based similarity to assess the feasibility of a given combination can add real value. Currently, serendipity plays a significant role in MKI discovery and many, if not most, MKIs have been discovered by chance during the search for selective inhibitors. Medicinal chemists need to be alert to the possibilities when a surprising combination is found by chance. To exploit such serendipity, you need a good appreciation of when you have a sufficiently high quality starting compd. and then you need to be able to make and test sufficient analogs to explore your new disease-based hypothesis. MKIs are costly to develop and are consequently priced at a premium level, so they will need to show clear improvements in order to get reimbursement. There have already been problems with reimbursement for some MKIs in some markets due to concerns from funding bodies over insufficient efficacy. The true value of MKIs relative to other anticancer drugs still has to be established, and the results from recent clin. trials have been mixed. Despite the broad activity profile of many MKIs, the patient response can be inconsistent and unpredictable. The identification of predictive biomarkers of response or resistance is a crit. step to ascertain which specific combination of targets produces a significant clin. benefit with respect to specific tumor types. More clin. feedback is needed to facilitate the design of the next generation of inhibitors with more precisely defined profiles. Although it might seem immeasurably distant at the present time, the ultimate goal should be to derive the prerequisite knowledge and tools so that MTDD becomes a rational endeavor rather than a black box approach that relies upon serendipity. This will help banish claims that MKIs are merely dirty, nonspecific drugs with insufficient specificity for treating a wider range of human diseases.
- 32Ou-Yang, S. S.; Lu, J. Y.; Kong, X. Q.; Liang, Z. J.; Luo, C.; Jiang, H. Computational Drug Discovery Acta Pharmacol. Sin. 2012, 33, 1131– 1140 DOI: 10.1038/aps.2012.109Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht12lt7vL&md5=2a0bff666d5881d1109124049480b1a2Computational drug discoveryOu-Yang, Si-sheng; Lu, Jun-yan; Kong, Xiang-qian; Liang, Zhong-jie; Luo, Cheng; Jiang, HualiangActa Pharmacologica Sinica (2012), 33 (9), 1131-1140CODEN: APSCG5; ISSN:1671-4083. (Nature Publishing Group)A review. Computational drug discovery is an effective strategy for accelerating and economizing drug discovery and development process. Because of the dramatic increase in the availability of biol. macromol. and small mol. information, the applicability of computational drug discovery has been extended and broadly applied to nearly every stage in the drug discovery and development workflow, including target identification and validation, lead discovery and optimization and preclin. tests. Over the past decades, computational drug discovery methods such as mol. docking, pharmacophore modeling and mapping, de novo design, mol. similarity calcn. and sequence-based virtual screening have been greatly improved. In this review, we present an overview of these important computational methods, platforms and successful applications in this field.
- 33Wan, S.; Bhati, A. P.; Zasada, S. J.; Wall, I.; Green, D.; Bamborough, P.; Coveney, P. V. Rapid and Reliable Binding Affinity Prediction of Bromodomain Inhibitors: A Computational Study J. Chem. Theory Comput. 2017, 13, 784– 795 DOI: 10.1021/acs.jctc.6b00794Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFektLnF&md5=713436084662420482684fb50db0832eRapid and Reliable Binding Affinity Prediction of Bromodomain Inhibitors: A Computational StudyWan, Shunzhou; Bhati, Agastya P.; Zasada, Stefan J.; Wall, Ian; Green, Darren; Bamborough, Paul; Coveney, Peter V.Journal of Chemical Theory and Computation (2017), 13 (2), 784-795CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Binding free energies of bromodomain inhibitors are calcd. with recently formulated approaches, namely ESMACS (enhanced sampling of mol. dynamics with approxn. of continuum solvent) and TIES (thermodn. integration with enhanced sampling). A set of compds. is provided by GlaxoSmithKline, which represents a range of chem. functionality and binding affinities. The predicted binding free energies exhibit a good Spearman correlation of 0.78 with the exptl. 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, the authors 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 kcal/mol and 1.71 kcal/mol for the 1- and 3-trajectory ESMACS approaches.
Cited By
This article is cited by 29 publications.
- 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
- 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
- Sharan K. Bagal, Mark Andrews, Bruce M. Bechle, Jianwei Bian, James Bilsland, David C. Blakemore, John F. Braganza, Peter J. Bungay, Matthew S. Corbett, Ciaran N. Cronin, Jingrong Jean Cui, Rebecca Dias, Neil J. Flanagan, Samantha E. Greasley, Rachel Grimley, Kim James, Eric Johnson, Linda Kitching, Michelle L. Kraus, Indrawan McAlpine, Asako Nagata, Sacha Ninkovic, Kiyoyuki Omoto, Stephanie Scales, Sarah E. Skerratt, Jianmin Sun, Michelle Tran-Dubé, Gareth J. Waldron, Fen Wang, Joseph S. Warmus. Discovery of Potent, Selective, and Peripherally Restricted Pan-Trk Kinase Inhibitors for the Treatment of Pain. Journal of Medicinal Chemistry 2018, 61
(15)
, 6779-6800. https://doi.org/10.1021/acs.jmedchem.8b00633
- 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
- José Jiménez, Miha Škalič, Gerard Martínez-Rosell, and Gianni De Fabritiis . KDEEP: Protein–Ligand Absolute Binding Affinity Prediction via 3D-Convolutional Neural Networks. Journal of Chemical Information and Modeling 2018, 58
(2)
, 287-296. https://doi.org/10.1021/acs.jcim.7b00650
- 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
- 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
- Siyabonga Melamane, Tavonga T. Mandava, Arthur Manda, Nonhlanhla Luphade, Sandile M.M. Khamanga, Pedzisai A. Makoni, Patrick H. Demana, Scott K. Matafwali, Bwalya A. Witika. Artificial neural network–based inference of drug–target interactions. 2023, 35-62. https://doi.org/10.1016/B978-0-323-91763-6.00015-1
- 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
- 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
- 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
- Francis Giraud, Elisabeth Pereira, Fabrice Anizon, Pascale Moreau. Recent Advances in Pain Management: Relevant Protein Kinases and Their Inhibitors. Molecules 2021, 26
(9)
, 2696. https://doi.org/10.3390/molecules26092696
- 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
- Anita de Ruiter, Chris Oostenbrink. Advances in the calculation of binding free energies. Current Opinion in Structural Biology 2020, 61 , 207-212. https://doi.org/10.1016/j.sbi.2020.01.016
- 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
- José Jiménez-Luna, Laura Pérez-Benito, Gerard Martínez-Rosell, Simone Sciabola, Rubben Torella, Gary Tresadern, Gianni De Fabritiis. DeltaDelta neural networks for lead optimization of small molecule potency. Chemical Science 2019, 10
(47)
, 10911-10918. https://doi.org/10.1039/C9SC04606B
- David W. Wright, Shunzhou Wan, Christophe Meyer, Herman van Vlijmen, Gary Tresadern, Peter V. Coveney. Application of ESMACS binding free energy protocols to diverse datasets: Bromodomain-containing protein 4. Scientific Reports 2019, 9
(1)
https://doi.org/10.1038/s41598-019-41758-1
- Robert Roskoski. Targeting ERK1/2 protein-serine/threonine kinases in human cancers. Pharmacological Research 2019, 142 , 151-168. https://doi.org/10.1016/j.phrs.2019.01.039
- 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
- Govindan Subramanian, Gennady Poda. In silico ligand‐based modeling of
h
BACE
‐1 inhibitors. Chemical Biology & Drug Design 2018, 91
(3)
, 817-827. https://doi.org/10.1111/cbdd.13147
- 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. Crystal structure of 1 bound to TrkA, viewed from the N-lobe to the C-lobe of the kinase. Hydrogen bonds are displayed by dashed lines. The protein is shown in cyan cartoon, and ligand atoms are colored by element: hydrogen in white, carbon in cyan, oxygen in red, and nitrogen in blue. For clarity, the N-lobe is not shown.
Figure 2
Figure 2. Comparisons of the TrkA experimental data and the calculated binding free energies from (a) 1-traj, (b) 2-traj, and (c) 3-traj ESMACS approaches. The equation in each panel indicates the calculations used in each case. The term Gcom/rec/ligcom/lig represents the free energy of the component (subscript: complex, receptor, or ligand) obtained from simulation of the same or different component (superscript: complex or free ligand in aqueous solution). The calculated binding free energies are associated with standard errors of ca. 0.6 kcal/mol in the 1-traj approach and ca. 3 kcal/mol in the 2- and 3-traj approaches (see Table S2), which are not shown in the figures for reasons of clarity. The experimental data from two sites (Pfizer, Sandwich and TCG Lifescience) are displayed in black and red, respectively. The correlation coefficients shown in the figure were calculated using the averages of the calculated binding free energies and the experimental data from Pfizer, Sandwich (black circles) and TCG Lifescience (red circles) where the former are not available. Large uncertainties are associated with the correlation coefficients because of the large error bars of the calculated and experimental binding affinities. Further analyses with bootstrapping resampling (see details in the Supporting Information) generate correlation coefficients of 0.39 ± 0.26, 0.60 ± 0.26, and 0.62 ± 0.27 for the 1-, 2-, and 3-traj approaches, respectively. It is evident that the 2- and 3-traj methods improve the ranking provided by the 1-traj method.
Figure 3
Figure 3. Calculated binding free energies for 1, 3, 4, 6, 7, 8, 13, 16, and 22 (a) from single structures after docking, (b) from 25 structures for each compound after 11 000-step minimization with a sophisticated conjugate-gradient method, and (c) from ensemble averages from three-trajectory ESMACS studies. Compounds 1, 3, 4, 6, 7, 8, 13, 16, and 22 are highly structurally similar (see Table 1).
Figure 4
Figure 4. Correlations of free energy components and the experimental data from the 3-traj approach. Both (a) bonded and (b) nonbonded energy terms contribute to the ranking of binding affinities, with similar correlation coefficients between the calculations and experimental data. Their combination, the MMPBSA energy (c), exhibits better correlations with the experimental data than the components themselves.
Figure 5
Figure 5. Adaptation free energies of (a) the receptor and (b) the compounds showing the binding free energy changes between the 1- and 3-traj approaches. The terms Grec/ligcom/lig are the free energies of receptor or ligands (subscript) calculated from simulations performed for the complex or the ligands (superscript).
Figure 6
Figure 6. Correlation between TIES-predicted relative binding affinities and experimental data. The black line is the correlation line, while the dotted lines (x = 0 and y = 0) create four quadrants. Ten out of the 14 data points are in quadrants I (x > 0 and y > 0) and III (x < 0 and y < 0), meaning that the calculated binding free energy differences have the same sign as those from the experimental data.
Figure 7
Figure 7. ESMACS simulations of (a) 1, (b) 22, and (c) 4 bound to TrkA. Final conformations of 4 ns production runs from all 25 replicas are overlapped and smoothed by averaging over 10 frames. Key residues E590 and M592 (for clarity, side-chain atoms are not shown) at the TrkA hinge region are highlighted. Hydrogen bonds are shown as dashed lines. The protein is shown in cyan cartoon, and ligand atoms are colored by element: hydrogen in white, carbon in cyan, oxygen in red, and nitrogen in blue. For reasons of clarity, the N-lobe of the protein has been removed.
Figure 8
Figure 8. ESMACS simulations of (a) 12, (b) 17, (c) 23, and (d) 24 bound to TrkA. Representative conformations are displayed using the final conformations of 4 ns production runs of one replica from each 25-member ensemble. The protein is shown by the surface representation and the ligand by the ball-and-stick representation with hydrogen in white, carbon in cyan, oxygen in red, and nitrogen in blue. The surfaces of the R2-position groups are shown in the wireframe representation.
Figure 9
Figure 9. ESMACS simulations of (a) 1, (b) 3, and (c) 6 bound to TrkA. Final conformations of 4 ns production runs from all 25 replicas are overlapped and smoothed by averaging over 10 frames. The key TrkA protein residues D668, K544, and E560 are highlighted. The protein is shown in cyan cartoon, and ligand atoms are colored by element: hydrogen in white, carbon in cyan, oxygen in red, and nitrogen in blue. Hydrogen bonds between the linker group carbonyl oxygen of 1, 3, and 6 and the backbone −NH group of D668 are shown as dashed lines.
Figure 10
Figure 10. ESMACS simulations of (a) 7 and (b) 8 bound to TrkA. Final conformations of 4 ns production runs from all 25 replicas are overlapped and smoothed by averaging over 10 frames. The key TrkA protein residues D668, E560, and L564 are highlighted. The protein is shown in cyan cartoon, and ligand atoms are colored by element: hydrogen in white, carbon in cyan, oxygen in red, and nitrogen in blue. The unfavorable electrostatic interactions between the methoxy group of 8 and the carbonyl group of D668 and between the methoxy group and the carboxylate group of E560 are highlighted by pink arrows.
Figure 11
Figure 11. ESMACS simulations of (a) 4 and (b) 16 bound to TrkA. Final conformations of 4 ns production runs from all 25 replicas are overlapped and smoothed by averaging over 10 conformations. The key TrkA protein residue D668 is highlighted. The protein is shown in cyan cartoon, and ligand atoms are colored by element: hydrogen in white, carbon in cyan, oxygen in red, and nitrogen in blue. The hydrogen bond between the linker-group carbonyl oxygen of 4 and the backbone −NH group of D668 is shown as a dashed line. As can be seen in (b), switching of the N–H and C═O groups prevents the formation of a hydrogen bond between the carbonyl oxygen of 16 and the backbone −NH group of D668.
References
ARTICLE SECTIONSThis article references 33 other publications.
- 1Sherborne, B.; Shanmugasundaram, V.; Cheng, A. C.; Christ, C. D.; DesJarlais, R. L.; Duca, J. S.; Lewis, R. A.; Loughney, D. A.; Manas, E. S.; McGaughey, G. B.; Peishoff, C. E.; van Vlijmen, H. Collaborating to Improve the Use of Free-Energy and Other Quantitative Methods in Drug Discovery J. Comput.-Aided Mol. Des. 2016, 30, 1139– 1141 DOI: 10.1007/s10822-016-9996-yGoogle Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFClsLfN&md5=c00386f775bd826c6b845bbbef4a975dCollaborating to improve the use of free-energy and other quantitative methods in drug discoverySherborne, Bradley; Shanmugasundaram, Veerabahu; Cheng, Alan C.; Christ, Clara D.; DesJarlais, Renee L.; Duca, Jose S.; Lewis, Richard A.; Loughney, Deborah A.; Manas, Eric S.; McGaughey, Georgia B.; Peishoff, Catherine E.; van Vlijmen, HermanJournal of Computer-Aided Molecular Design (2016), 30 (12), 1139-1141CODEN: JCADEQ; ISSN:0920-654X. (Springer)In May and August, 2016, several pharmaceutical companies convened to discuss and compare experiences with Free Energy Perturbation (FEP). This unusual synchronization of interest was prompted by Schrodinger's FEP+ implementation and offered the opportunity to share fresh studies with FEP and enable broader discussions on the topic. This article summarizes key conclusions of the meetings, including a path forward of actions for this group to aid the accelerated evaluation, application and development of free energy and related quant., structure-based design methods.
- 2Skerratt, S. E.; Andrews, M.; Bagal, S. K.; Bilsland, J.; Brown, D.; Bungay, P. J.; Cole, S.; Gibson, K. R.; Jones, R.; Morao, I.; Nedderman, A.; Omoto, K.; Robinson, C.; Ryckmans, T.; Skinner, K.; Stupple, P.; Waldron, G. The Discovery of a Potent, Selective, and Peripherally Restricted Pan-Trk Inhibitor (PF-06273340) for the Treatment of Pain J. Med. Chem. 2016, 59, 10084– 10099 DOI: 10.1021/acs.jmedchem.6b00850Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslWjsbfL&md5=f8c904a92c1716062c217fefe2374ce6The Discovery of a Potent, Selective, and Peripherally Restricted Pan-Trk Inhibitor (PF-06273340) for the Treatment of PainSkerratt, Sarah E.; Andrews, Mark; Bagal, Sharan K.; Bilsland, James; Brown, David; Bungay, Peter J.; Cole, Susan; Gibson, Karl R.; Jones, Russell; Morao, Inaki; Nedderman, Angus; Omoto, Kiyoyuki; Robinson, Colin; Ryckmans, Thomas; Skinner, Kimberly; Stupple, Paul; Waldron, GarethJournal of Medicinal Chemistry (2016), 59 (22), 10084-10099CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The neurotrophin family of growth factors, comprised of nerve growth factor (NGF), brain derived neurotrophic factor (BDNF), neurotrophin 3 (NT3) and neurotrophin 4 (NT4), is implicated in the physiol. of chronic pain. Given the clin. efficacy of anti-NGF monoclonal antibody (mAb) therapies, there is significant interest in the development of small mol. modulators of neurotrophin activity. Neurotrophins signal through the tropomyosin related kinase (Trk) family of tyrosine kinase receptors, hence Trk kinase inhibition represents a potentially "druggable" point of intervention. To deliver the safety profile required for chronic, non-life threatening pain indications, highly kinase-selective Trk inhibitors with minimal brain availability are sought. Herein the authors describe how the use of SBDD, 2D QSAR models and Matched Mol. Pair data in compd. design enabled the delivery of the highly potent, kinase-selective and peripherally restricted clin. candidate PF-06273340.
- 3Sadiq, 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, 1909– 1919 DOI: 10.1021/ci8000937Google Scholar3https://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.
- 4Groen, 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 Scholar4https://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.
- 5Norman, B. H.; McDermott, J. S. Targeting the Nerve Growth Factor (NGF) Pathway in Drug Discovery. Potential Applications to New Therapies for Chronic Pain J. Med. Chem. 2017, 60, 66– 88 DOI: 10.1021/acs.jmedchem.6b00964Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslSqtLjF&md5=606097686779a102db9e6e54f8252a40Targeting the Nerve Growth Factor (NGF) Pathway in Drug Discovery. Potential Applications to New Therapies for Chronic PainNorman, Bryan H.; McDermott, Jeff S.Journal of Medicinal Chemistry (2017), 60 (1), 66-88CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. The neurotrophin nerve growth factor (NGF) has been implicated as a key mediator of chronic pain. NGF binds the tropomyosin receptor kinase A (TrkA) and p75, resulting in the activation of downstream signaling pathways that have been linked to pro-nociception. While anti-NGF antibodies have demonstrated analgesia both preclinically and in patients, the mechanism of action of these agents remains unclear. The authors describe ligands targeting NGF, its receptors and downstream/related targets. This perspective highlights large and small mol. approaches to targeting the NGF-TrkA pathway both extra- and intracellularly. In addn., the authors present a strategic framework for future drug discovery efforts in this pathway beyond the targeting of NGF or its receptors. While existing tools have greatly informed NGF-mediated signaling, ongoing and future pathway research may help focus new drug discovery efforts on key novel targets and mechanisms. This may result in highly differentiated therapeutics with greater efficacy and/or improved safety profiles.
- 6Hefti, F. F.; Rosenthal, A.; Walicke, P. A.; Wyatt, S.; Vergara, G.; Shelton, D. L.; Davies, A. M. Novel Class of Pain Drugs Based on Antagonism of NGF Trends Pharmacol. Sci. 2006, 27, 85– 91 DOI: 10.1016/j.tips.2005.12.001Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtV2lsLo%253D&md5=3cfe9e06069c3b6b9aa9d96b7411d199Novel class of pain drugs based on antagonism of NGFHefti, Franz F.; Rosenthal, Arnon; Walicke, Patricia A.; Wyatt, Sean; Vergara, German; Shelton, David L.; Davies, Alun M.Trends in Pharmacological Sciences (2006), 27 (2), 85-91CODEN: TPHSDY; ISSN:0165-6147. (Elsevier Ltd.)A review. Nerve growth factor (NGF) was identified originally as a survival factor for sensory and sympathetic neurons in the developing nervous system. In adults, NGF is not required for survival but it has a crucial role in the generation of pain and hyperalgesia in several acute and chronic pain states. The expression of NGF is high in injured and inflamed tissues, and activation of the NGF receptor tyrosine kinase trkA on nociceptive neurons triggers and potentiates pain signaling by multiple mechanisms. Inhibition of NGF function and signaling blocks pain sensation as effectively as cyclooxygenase inhibitors and opiates in rodent models of pain. Several pharmaceutical companies have active drug-discovery and development programs that are based on a variety of approaches to antagonize NGF, including NGF capture', blocking the binding of NGF to trkA and inhibiting trkA signaling. NGF antagonism is expected to be a highly effective therapeutic approach in many pain states, and to be free of the adverse effects of traditional analgesic drugs.
- 7Mantyh, P. W.; Koltzenburg, M.; Mendell, L. M.; Tive, L.; Shelton, D. L. Antagonism of Nerve Growth Factor-TrkA Signaling and the Relief of Pain Anesthesiology 2011, 115, 189– 204 DOI: 10.1097/ALN.0b013e31821b1ac5Google ScholarThere is no corresponding record for this reference.
- 8Wan, 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, 3346– 3356 DOI: 10.1021/acs.jctc.5b00179Google Scholar8https://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.
- 9Kollman, 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, 889– 897 DOI: 10.1021/ar000033jGoogle Scholar9https://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.
- 10Coveney, P. V.; Wan, S. On the Calculation of Equilibrium Thermodynamic Properties from Molecular Dynamics Phys. Chem. Chem. Phys. 2016, 18, 30236– 30240 DOI: 10.1039/C6CP02349EGoogle Scholar10https://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.
- 11Miller, B. R., 3rd; McGee, T. D., Jr.; Swails, J. M.; Homeyer, N.; Gohlke, H.; Roitberg, A. E. Mmpbsa.Py: An Efficient Program for End-State Free Energy Calculations J. Chem. Theory Comput. 2012, 8, 3314– 3321 DOI: 10.1021/ct300418hGoogle Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtV2gtrzP&md5=cc4148bd8f70c7cad94fd3ec6f580e52MMPBSA.py: An Efficient Program for End-State Free Energy CalculationsMiller, Bill R., III; McGee, T. Dwight, Jr.; Swails, Jason M.; Homeyer, Nadine; Gohlke, Holger; Roitberg, Adrian E.Journal of Chemical Theory and Computation (2012), 8 (9), 3314-3321CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)MM-PBSA is a post-processing end-state method to calc. free energies of mols. in soln. MMPBSA.py is a program written in Python for streamlining end-state free energy calcns. using ensembles derived from mol. dynamics (MD) or Monte Carlo (MC) simulations. Several implicit solvation models are available with MMPBSA.py, including the Poisson-Boltzmann Model, the Generalized Born Model, and the Ref. Interaction Site Model. Vibrational frequencies may be calcd. using normal mode or quasi-harmonic anal. to approx. the solute entropy. Specific interactions can also be dissected using free energy decompn. or alanine scanning. A parallel implementation significantly speeds up the calcn. by dividing frames evenly across available processors. MMPBSA.py is an efficient, user-friendly program with the flexibility to accommodate the needs of users performing end-state free energy calcns. The source code can be downloaded at http://ambermd.org/ with AmberTools, released under the GNU General Public License.
- 12Case, D. A.; Cheatham, T. E., III; 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, 1668– 1688 DOI: 10.1002/jcc.20290Google Scholar12https://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.
- 13Bhati, 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. 2017, 13, 210– 222 DOI: 10.1021/acs.jctc.6b00979Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitVGntrrJ&md5=510b70188112a3578030e291ce19b127Rapid, Accurate, Precise, and Reliable Relative Free Energy Prediction Using Ensemble Based Thermodynamic IntegrationBhati, Agastya P.; Wan, Shunzhou; Wright, David W.; Coveney, Peter V.Journal of Chemical Theory and Computation (2017), 13 (1), 210-222CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The accurate prediction of the binding affinities of ligands to proteins is a major goal in drug discovery and personalized medicine. The time taken to make such predictions is of similar importance to their accuracy, precision and reliability. In the last few years, an ensemble based mol. dynamics approach has been proposed that provides a route to reliable predictions of free energies based on the mol. mechanics Poisson-Boltzmann surface area method which meets the requirements of accuracy, precision and reliability. Here, we describe an equiv. methodol. based on thermodn. integration to substantially improve the accuracy, precision and reliability of calcd. relative binding free energies. We report the performance of the method when applied to a diverse set of protein targets and ligands. The results are in very good agreement with exptl. data (90% of calcns. agree to within 1 kcal/mol) while the method is reproducible by construction. Statistical uncertainties of the order of 0.5 kcal/mol or less are achieved. We present a systematic account of how the uncertainty in the predictions may be estd.
- 14Wright, 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, 1228– 1241 DOI: 10.1021/ct4007037Google Scholar14https://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.
- 15Swanson, 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 Scholar15https://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.
- 16Chodera, J. D.; Mobley, D. L.; Shirts, M. R.; Dixon, R. W.; Branson, K.; Pande, V. S. Alchemical Free Energy Methods for Drug Discovery: Progress and Challenges Curr. Opin. Struct. Biol. 2011, 21, 150– 160 DOI: 10.1016/j.sbi.2011.01.011Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXkt1Kisrk%253D&md5=fa17c0982d921cd1c32cd095fd34420eAlchemical free energy methods for drug discovery: Progress and challengesChodera, John D.; Mobley, David L.; Shirts, Michael R.; Dixon, Richard W.; Branson, Kim; Pande, Vijay S.Current Opinion in Structural Biology (2011), 21 (2), 150-160CODEN: COSBEF; ISSN:0959-440X. (Elsevier Ltd.)A review. Improved rational drug design methods are needed to lower the cost and increase the success rate of drug discovery and development. Alchem. binding free energy calcns., one potential tool for rational design, have progressed rapidly over the past decade, but still fall short of providing robust tools for pharmaceutical engineering. Recent studies, esp. on model receptor systems, have clarified many of the challenges that must be overcome for robust predictions of binding affinity to be useful in rational design. In this review, inspired by a recent joint academic/industry meeting organized by the authors, we discuss these challenges and suggest a no. of promising approaches for overcoming them.
- 17Wang, 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, 2695– 2703 DOI: 10.1021/ja512751qGoogle Scholar17https://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.
- 18Frisch, M. J.; Trucks, G. W.; Schlegel, H. B.; Scuseria, G. E.; Robb, M. A.; Cheeseman, J. R.; Montgomery, J. J.; 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.
- 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, 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.
- 20Lindorff-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., Bioinf. 2010, 78, 1950– 1958 DOI: 10.1002/prot.22711Google Scholar20https://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.
- 21Phillips, 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, 1781– 1802 DOI: 10.1002/jcc.20289Google Scholar21https://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.
- 22Highfield, R. Supercomputer bid to create the first truly personalised medicine. https://blog.sciencemuseum.org.uk/supercomputer-bid-to-create-the-first-truly-personalised-medicine/ (accessed Feb 22, 2017).Google ScholarThere is no corresponding record for this reference.
- 23Fiser, A.; Sali, A. Modloop: Automated Modeling of Loops in Protein Structures Bioinformatics 2003, 19, 2500– 2501 DOI: 10.1093/bioinformatics/btg362Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXpvVSisLk%253D&md5=4b0fdaca0a412715682496883ab2019cModLoop: automated modeling of loops in protein structuresFiser, Andras; Sali, AndrejBioinformatics (2003), 19 (18), 2500-2501CODEN: BOINFP; ISSN:1367-4803. (Oxford University Press)Summary: ModLoop is a web server for automated modeling of loops in protein structures. The input is the at. coordinates of the protein structure in the Protein Data Bank format, and the specification of the starting and ending residues of one or more segments to be modeled, contg. no more than 20 residues in total. The output is the coordinates of the non-hydrogen atoms in the modeled segments. A user provides the input to the server via a simple web interface, and receives the output by e-mail. The server relies on the loop modeling routine in MODELLER that predicts the loop conformations by satisfaction of spatial restraints, without relying on a database of known protein structures. For a rapid response, ModLoop runs on a cluster of Linux PC computers.
- 24Allen, W. J.; Balius, T. E.; Mukherjee, S.; Brozell, S. R.; Moustakas, D. T.; Lang, P. T.; Case, D. A.; Kuntz, I. D.; Rizzo, R. C. Dock 6: Impact of New Features and Current Docking Performance J. Comput. Chem. 2015, 36, 1132– 1156 DOI: 10.1002/jcc.23905Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXnt1Smtbs%253D&md5=8b5fc6cc7533f975e1e740acb3688807DOCK 6: Impact of new features and current docking performanceAllen, William J.; Balius, Trent E.; Mukherjee, Sudipto; Brozell, Scott R.; Moustakas, Demetri T.; Lang, P. Therese; Case, David A.; Kuntz, Irwin D.; Rizzo, Robert C.Journal of Computational Chemistry (2015), 36 (15), 1132-1156CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)This manuscript presents the latest algorithmic and methodol. developments to the structure-based design program DOCK 6.7 focused on an updated internal energy function, new anchor selection control, enhanced minimization options, a footprint similarity scoring function, a symmetry-cor. root-mean-square deviation algorithm, a database filter, and docking forensic tools. An important strategy during development involved use of three orthogonal metrics for assessment and validation: pose reprodn. over a large database of 1043 protein-ligand complexes (SB2012 test set), cross-docking to 24 drug-target protein families, and database enrichment using large active and decoy datasets (Directory of Useful Decoys [DUD]-E test set) for five important proteins including HIV protease and IGF-1R. Relative to earlier versions, a key outcome of the work is a significant increase in pose reprodn. success in going from DOCK 4.0.2 (51.4%) → 5.4 (65.2%) → 6.7 (73.3%) as a result of significant decreases in failure arising from both sampling 24.1% → 13.6% → 9.1% and scoring 24.4% → 21.1% → 17.5%. Companion cross-docking and enrichment studies with the new version highlight other strengths and remaining areas for improvement, esp. for systems contg. metal ions. The source code for DOCK 6.7 is available for download and free for academic users at. © 2015 Wiley Periodicals, Inc.
- 25Chodera, 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 Scholar25https://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.
- 26Bissantz, C.; Folkers, G.; Rognan, D. Protein-Based Virtual Screening of Chemical Databases. 1. Evaluation of Different Docking/Scoring Combinations J. Med. Chem. 2000, 43, 4759– 4767 DOI: 10.1021/jm001044lGoogle Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXotFymurY%253D&md5=ee74ac9a99e55759c2df27fd1db58010Protein-Based Virtual Screening of Chemical Databases. 1. Evaluation of Different Docking/Scoring CombinationsBissantz, Caterina; Folkers, Gerd; Rognan, DidierJournal of Medicinal Chemistry (2000), 43 (25), 4759-4767CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Three different database docking programs (Dock, FlexX, Gold) have been used in combination with seven scoring functions (Chemscore, Dock, FlexX, Fresno, Gold, Pmf, Score) to assess the accuracy of virtual screening methods against two protein targets (thymidine kinase, estrogen receptor) of known three-dimensional structure. For both targets, it was generally possible to discriminate about 7 out of 10 true hits from a random database of 990 ligands. The use of consensus lists common to two or three scoring functions clearly enhances hit rates among the top 5% scorers from 10% (single scoring) to 25-40% (double scoring) and up to 65-70% (triple scoring). However, in all tested cases, no clear relationships could be found between docking and ranking accuracies. Moreover, predicting the abs. binding free energy of true hits was not possible whatever docking accuracy was achieved and scoring function used. As the best docking/consensus scoring combination varies with the selected target and the physicochem. of target-ligand interactions, we propose a two-step protocol for screening large databases: (i) screening of a reduced dataset contg. a few known ligands for deriving the optimal docking/consensus scoring scheme, (ii) applying the latter parameters to the screening of the entire database.
- 27Gohlke, H.; Kiel, C.; Case, D. A. Insights into Protein-Protein Binding by Binding Free Energy Calculation and Free Energy Decomposition for the Ras-Raf and Ras-RaiGDS Complexes J. Mol. Biol. 2003, 330, 891– 913 DOI: 10.1016/S0022-2836(03)00610-7Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXlt1aqtbs%253D&md5=0a8acf22e053534f34365f264ed4192aInsights into protein-protein binding by binding free energy calculation and free energy decomposition for the Ras-Raf and Ras-RalGDS complexesGohlke, Holger; Kiel, Christina; Case, David A.Journal of Molecular Biology (2003), 330 (4), 891-913CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Science Ltd.)Abs. binding free energy calcns. and free energy decompns. are presented for the protein-protein complexes H-Ras/C-Raf1 and H-Ras/RalGDS. Ras is a central switch in the regulation of cell proliferation and differentiation. In our study, we investigate the capability of the mol. mechanics (MM)-generalized Born surface area (GBSA) approach to est. abs. binding free energies for the protein-protein complexes. Averaging gas-phase energies, solvation free energies, and entropic contributions over snapshots extd. from trajectories of the unbound proteins and the complexes, calcd. binding free energies (Ras-Raf: -15.0(±6.3) kcal mol-1; Ras-RalGDS: -19.5(±5.9) kcal mol-1) are in fair agreement with exptl. detd. values (-9.6 kcal mol-1; -8.4 kcal mol-1), if appropriate ionic strength is taken into account. Structural determinants of the binding affinity of Ras-Raf and Ras-RalGDS are identified by means of free energy decompn. For the first time, computationally inexpensive generalized Born (GB) calcns. are applied in this context to partition solvation free energies along with gas-phase energies between residues of both binding partners. For selected residues, in addn., entropic contributions are estd. by classical statistical mechanics. Comparison of the decompn. results with exptl. detd. binding free energy differences for alanine mutants of interface residues yielded correlations with r2=0.55 and 0.46 for Ras-Raf and Ras-RalGDS, resp. Extension of the decompn. reveals residues as far apart as 25 A from the binding epitope that can contribute significantly to binding free energy. These "hotspots" are found to show large at. fluctuations in the unbound proteins, indicating that they reside in structurally less stable regions. Furthermore, hotspot residues experience a significantly larger-than-av. decrease in local fluctuations upon complex formation. Finally, by calcg. a pair-wise decompn. of interactions, interaction pathways originating in the binding epitope of Raf are found that protrude through the protein structure towards the loop L1. This explains the finding of a conformational change in this region upon complex formation with Ras, and it may trigger a larger structural change in Raf, which is considered to be necessary for activation of the effector by Ras.
- 28Bunney, 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, 194– 204 DOI: 10.1016/j.ebiom.2015.02.009Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2srotlKmtA%253D%253D&md5=8414f3530d24bbba0ea8cc98cb784b96The Effect of Mutations on Drug Sensitivity and Kinase Activity of Fibroblast Growth Factor Receptors: A Combined Experimental and Theoretical StudyBunney Tom D; Thiyagarajan Nethaji; Ashford Paul; Katan Matilda; Wan Shunzhou; Coveney Peter V; Sutto Ludovico; Gervasio Francesco L; Williams Sarah V; Knowles Margaret A; Koss HansEBioMedicine (2015), 2 (3), 194-204 ISSN:.Fibroblast growth factor receptors (FGFRs) are recognized therapeutic targets in cancer. We here describe insights underpinning the impact of mutations on FGFR1 and FGFR3 kinase activity and drug efficacy, using a combination of computational calculations and experimental approaches including cellular studies, X-ray crystallography and biophysical and biochemical measurements. Our findings reveal that some of the tested compounds, in particular TKI258, could provide therapeutic opportunity not only for patients with primary alterations in FGFR but also for acquired resistance due to the gatekeeper mutation. The accuracy of the computational methodologies applied here shows a potential for their wider application in studies of drug binding and in assessments of functional and mechanistic impacts of mutations, thus assisting efforts in precision medicine.
- 29Andrews, M. D.; Bagal, S. K.; Gibson, K. R.; Omoto, K.; Ryckmans, T.; Skerratt, S. E.; Stupple, P. A. Pyrrolo[2,3-d]pyrimidine Derivatives as Inhibitors of Tropomyosin-Related Kinases and Their Preparation and Use in the Treatment of Pain. WO2012137089A1, 2012.Google ScholarThere is no corresponding record for this reference.
- 30Xing, L.; Klug-Mcleod, J.; Rai, B.; Lunney, E. A. Kinase Hinge Binding Scaffolds and Their Hydrogen Bond Patterns Bioorg. Med. Chem. 2015, 23, 6520– 6527 DOI: 10.1016/j.bmc.2015.08.006Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVentr3L&md5=048b4b99628c6e1ae273b00a47ab2377Kinase hinge binding scaffolds and their hydrogen bond patternsXing, Li; Klug-Mcleod, Jacquelyn; Rai, Brajesh; Lunney, Elizabeth A.Bioorganic & Medicinal Chemistry (2015), 23 (19), 6520-6527CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Protein kinases constitute a major class of intracellular signaling mols., and describe some of the most prominent drug targets. Kinase inhibitors commonly employ small chem. scaffolds that form hydrogen bonds with the kinase hinge residues connecting the N- and C-terminal lobes of the catalytic domain. In general the satisfied hydrogen bonds are required for potent inhibition, therefore constituting a conserved feature in the majority of inhibitor-kinase interactions. From systematically analyzing the kinase scaffolds extd. from Pfizer crystal structure database (CSDb) the authors recognize that large no. of kinase inhibitors of diverse chem. structures are derived from a relatively small no. of common scaffolds. Depending on specific substitution patterns, scaffolds may demonstrate versatile binding capacities to interact with kinase hinge. Afforded by thousands of ligand-protein binary complexes, the hinge hydrogen bond patterns were analyzed with a focus on their three-dimensional configurations. Most of the compds. engage H6 NH for hinge recognition. Dual hydrogen bonds are commonly obsd. with addnl. recruitment of H4 CO upstream and/or H6 CO downstream. Triple hydrogen bonds accounts for small no. of binary complexes. An unusual hydrogen bond with a non-canonical H5 conformation is obsd., requiring a peptide bond flip by a glycine residue at the H6 position. Addnl. hydrogen bonds to kinase hinge do not necessarily correlate with an increase in potency; conversely they appear to compromise kinase selectivity. Such learnings could enhance the prospect of successful therapy design.
- 31Morphy, R. Selectively Nonselective Kinase Inhibition: Striking the Right Balance J. Med. Chem. 2010, 53, 1413– 1437 DOI: 10.1021/jm901132vGoogle Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtlWmtbfF&md5=3c915b5debf9d005993c8ad31c68bc2eSelectively Nonselective Kinase Inhibition: Striking the Right BalanceMorphy, RichardJournal of Medicinal Chemistry (2010), 53 (4), 1413-1437CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Marketed kinase inhibitors (MKIs) can deliver superior efficacy compared to inhibitors with high specificity for a single kinase, and the recent introduction of several MKIs to the market opens the door to a new era of safer and effective anticancer therapy. The key to combining high efficacy with acceptable safety is to inhibit multiple targets in a selectively nonselective fashion. Strategies for intentionally designing MKIs are emerging, but the field is still in its infancy and we are as medicinal chemists currently on the steepest part of the learning curve. MTDD can be time-consuming and expensive, and we need to become more proficient first at identifying disease-relevant target combinations and second at discovering MKIs that combine optimal physicochem. and biol. properties. Bold and innovative medicinal chem. strategies are required to tackle "difficult combinations" where the disease rationale is compelling but where it is a struggle to combine all the desired attributes of an oral MKI drug into a single mol. At present it is unclear to what extent MKIs with highly tuned selectivity profiles can be rationally designed, particularly for targets that are unrelated by sequence. In addn. to the well-known selectivity challenge, the physicochem. property profiles of AT P-competitive MKIs can be inherently challenging and limited scope for patentability can also be a serious hindrance. On the plus side, the amt. of kinase-specific structural information is growing very rapidly, and ultimately this may reveal distinct features and design rules that enable a medicinal chemist to rationally modify and refine the profile of MKIs. In addn., increasing SAR knowledge is emerging from large scale panel screening with the binding profiles starting to reveal to medicinal chemists how chem. structure affects cross-reactivity across large parts of the kinome. The merit of MKIs compared with single kinase inhibitors is a subject of controversy in drug discovery that is unlikely to be resolved in the near future. At the start of a new MTDD project, a rigorous debate needs to take place as to whether it makes more sense to seek a combination of highly selective agents or a DML. Many factors need to be taken into account in this decision such as the no., similarity, and promiscuity of the targets in the profile and the disease area. Conformational plasticity and the occurrence of multiple binding modes complicate the in silico prediction of kinase polypharmacol. based solely upon protein structure. The use of ligand-based similarity to assess the feasibility of a given combination can add real value. Currently, serendipity plays a significant role in MKI discovery and many, if not most, MKIs have been discovered by chance during the search for selective inhibitors. Medicinal chemists need to be alert to the possibilities when a surprising combination is found by chance. To exploit such serendipity, you need a good appreciation of when you have a sufficiently high quality starting compd. and then you need to be able to make and test sufficient analogs to explore your new disease-based hypothesis. MKIs are costly to develop and are consequently priced at a premium level, so they will need to show clear improvements in order to get reimbursement. There have already been problems with reimbursement for some MKIs in some markets due to concerns from funding bodies over insufficient efficacy. The true value of MKIs relative to other anticancer drugs still has to be established, and the results from recent clin. trials have been mixed. Despite the broad activity profile of many MKIs, the patient response can be inconsistent and unpredictable. The identification of predictive biomarkers of response or resistance is a crit. step to ascertain which specific combination of targets produces a significant clin. benefit with respect to specific tumor types. More clin. feedback is needed to facilitate the design of the next generation of inhibitors with more precisely defined profiles. Although it might seem immeasurably distant at the present time, the ultimate goal should be to derive the prerequisite knowledge and tools so that MTDD becomes a rational endeavor rather than a black box approach that relies upon serendipity. This will help banish claims that MKIs are merely dirty, nonspecific drugs with insufficient specificity for treating a wider range of human diseases.
- 32Ou-Yang, S. S.; Lu, J. Y.; Kong, X. Q.; Liang, Z. J.; Luo, C.; Jiang, H. Computational Drug Discovery Acta Pharmacol. Sin. 2012, 33, 1131– 1140 DOI: 10.1038/aps.2012.109Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht12lt7vL&md5=2a0bff666d5881d1109124049480b1a2Computational drug discoveryOu-Yang, Si-sheng; Lu, Jun-yan; Kong, Xiang-qian; Liang, Zhong-jie; Luo, Cheng; Jiang, HualiangActa Pharmacologica Sinica (2012), 33 (9), 1131-1140CODEN: APSCG5; ISSN:1671-4083. (Nature Publishing Group)A review. Computational drug discovery is an effective strategy for accelerating and economizing drug discovery and development process. Because of the dramatic increase in the availability of biol. macromol. and small mol. information, the applicability of computational drug discovery has been extended and broadly applied to nearly every stage in the drug discovery and development workflow, including target identification and validation, lead discovery and optimization and preclin. tests. Over the past decades, computational drug discovery methods such as mol. docking, pharmacophore modeling and mapping, de novo design, mol. similarity calcn. and sequence-based virtual screening have been greatly improved. In this review, we present an overview of these important computational methods, platforms and successful applications in this field.
- 33Wan, S.; Bhati, A. P.; Zasada, S. J.; Wall, I.; Green, D.; Bamborough, P.; Coveney, P. V. Rapid and Reliable Binding Affinity Prediction of Bromodomain Inhibitors: A Computational Study J. Chem. Theory Comput. 2017, 13, 784– 795 DOI: 10.1021/acs.jctc.6b00794Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFektLnF&md5=713436084662420482684fb50db0832eRapid and Reliable Binding Affinity Prediction of Bromodomain Inhibitors: A Computational StudyWan, Shunzhou; Bhati, Agastya P.; Zasada, Stefan J.; Wall, Ian; Green, Darren; Bamborough, Paul; Coveney, Peter V.Journal of Chemical Theory and Computation (2017), 13 (2), 784-795CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Binding free energies of bromodomain inhibitors are calcd. with recently formulated approaches, namely ESMACS (enhanced sampling of mol. dynamics with approxn. of continuum solvent) and TIES (thermodn. integration with enhanced sampling). A set of compds. is provided by GlaxoSmithKline, which represents a range of chem. functionality and binding affinities. The predicted binding free energies exhibit a good Spearman correlation of 0.78 with the exptl. 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, the authors 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 kcal/mol and 1.71 kcal/mol for the 1- and 3-trajectory ESMACS approaches.
Supporting Information
Supporting Information
ARTICLE SECTIONSThe Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jcim.6b00780.
Detailed description of the energy decomposition, the energy convergence, and the error analyses, experimental measurements of TrkA inhibitory activity, and the predicted binding free energies (PDF)
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.