Ensemble Simulations and Experimental Free Energy Distributions: Evaluation and Characterization of Isoxazole Amides as SMYD3 InhibitorsClick to copy article linkArticle link copied!
- Shunzhou WanShunzhou WanCentre for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, U.K.More by Shunzhou Wan
- Agastya P. BhatiAgastya P. BhatiCentre for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, U.K.More by Agastya P. Bhati
- David W. WrightDavid W. WrightCentre for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, U.K.More by David W. Wright
- Ian D. WallIan D. WallGlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K.More by Ian D. Wall
- Alan P. GravesAlan P. GravesGlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United StatesMore by Alan P. Graves
- Darren GreenDarren GreenGlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K.More by Darren Green
- Peter V. Coveney*Peter V. Coveney*Email: [email protected]Centre for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, U.K.Advanced Research Computing Centre, University College London, London WC1H 0AJ U.K.Institute for Informatics, Faculty of Science, University of Amsterdam, 1098XH Amsterdam, The NetherlandsMore by Peter V. Coveney
Abstract
Optimization of binding affinities for ligands to their target protein is a primary objective in rational drug discovery. Herein, we report on a collaborative study that evaluates various compounds designed to bind to the SET and MYND domain-containing protein 3 (SMYD3). SMYD3 is a histone methyltransferase and plays an important role in transcriptional regulation in cell proliferation, cell cycle, and human carcinogenesis. Experimental measurements using the scintillation proximity assay show that the distributions of binding free energies from a large number of independent measurements exhibit non-normal properties. We use ESMACS (enhanced sampling of molecular dynamics with approximation of continuum solvent) and TIES (thermodynamic integration with enhanced sampling) protocols to predict the binding free energies and to provide a detailed chemical insight into the nature of ligand–protein binding. Our results show that the 1-trajectory ESMACS protocol works well for the set of ligands studied here. Although one unexplained outlier exists, we obtain excellent statistical ranking across the set of compounds from the ESMACS protocol and good agreement between calculations and experiments for the relative binding free energies from the TIES protocol. ESMACS and TIES are again found to be powerful protocols for the accurate comparison of the binding free energies.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
1. Introduction
The binding free energy is estimated by ΔGexp = RT × ln (IC50) = – RT × ln (10) × pIC50, where R is the gas constant and T is temperature, which is set to 300 K in this study.
There was no activity at the highest concentration (250 μM) tested.
2. Material and Methods
2.1. Experiments
2.2. Computational Approach
2.2.1. Compounds
2.2.2. Model Preparation
2.2.3. ESMACS
2.2.4. Thermodynamic Integration with Enhanced Sampling
2.2.5. Simulations
3. Results
3.1. Reproducibility of the ESMACS Simulations
3.2. Correlations between ESMACS Calculations and Experimental Measurements
3.3. TIES Results
3.4. S08 Remains an Outlier
3.5. Non-normal Distributions of Free Energy Calculations and Measurement
compound | no. of test | average | SD | skewness | kurtosis |
---|---|---|---|---|---|
a | 264 | 5.29 | 0.10 | 0.88 | 1.47 |
b | 114 | 7.31 | 0.12 | –0.35 | 0.23 |
c | 124 | 7.47 | 0.19 | 2.04 | 7.56 |
d | 116 | 7.09 | 0.21 | 2.11 | 5.30 |
compound | (−∞, −2σ) | [−2σ, −σ) | [−σ, 0) | [0, σ) | [σ, 2σ) | [2σ, ∞) |
---|---|---|---|---|---|---|
a | 4.9 | 9.8 | 29.5 | 43.9 | 11.0 | 0.8 |
b | 1.8 | 15.8 | 32.5 | 36.8 | 9.6 | 3.5 |
c | 3.2 | 7.3 | 28.2 | 54.8 | 6.5 | 0 |
d | 5.2 | 4.3 | 28.4 | 55.2 | 6.9 | 0 |
normal | 2.3 | 13.6 | 34.1 | 34.1 | 13.6 | 2.3 |
The experimental binding affinities pIC50 have been converted into binding free energies using ΔG = RT ln (IC50). σ values are the standard deviations of the normal distributions which best fit the binding free energies for each of the compounds. The percentages from a normal distribution are listed for reference.
4. Conclusions
Acknowledgments
We are grateful for funding for the UK MRC Medical Bioinformatics project (grant no. MR/L016311/1), the EPSRC funded UK Consortium on Mesoscale Engineering Sciences (UKCOMES grant no. EP/L00030X/1), the European Commission for EU H2020 CompBioMed2 Centre of Excellence (grant no. 823712) and for EU H2020 EXDCI-2 project (grant no. 800957), and NSF award (https://www.nsf.gov/pubs/2017/nsf17542/nsf17542.htm, Award No. NSF 1713749). We made use of the Blue Waters supercomputer at the National Center for Supercomputing Applications of the University of Illinois at Urbana-Champaign, access to which was made available through the aforementioned NSF award. Additional calculations were conducted using an award of computer time on the Theta machine at Argonne Leadership Computing Facility provided by the US Department of Energy’s Innovative and Novel Computational Impact on Theory and Experiment (INCITE) program (through the INSPIRE project and a 2021 award “COMPBIO”). We are grateful to DNAnexus for both computational resources and technical support from Fiona Ford, Brett Hannigan and Chai Fungtammasan. We acknowledge the Leibniz Supercomputing Centre for providing access to SuperMUC (https://www.lrz.de/services/compute/) and the very able assistance of its scientific support staff.
References
This article references 39 other publications.
- 1Mazur, P. K.; Reynoird, N.; Khatri, P.; Jansen, P. W.; Wilkinson, A. W.; Liu, S.; Barbash, O.; Van Aller, G. S.; Huddleston, M.; Dhanak, D.; Tummino, P. J.; Kruger, R. G.; Garcia, B. A.; Butte, A. J.; Vermeulen, M.; Sage, J.; Gozani, O. SMYD3 Links Lysine Methylation of Map3k2 to Ras-Driven Cancer. Nature 2014, 510, 283– 287, DOI: 10.1038/nature13320Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXps1Wmtbc%253D&md5=009705b03f725a45664d3feb74f92f7aSMYD3 links lysine methylation of MAP3K2 to Ras-driven cancerMazur, Pawel K.; Reynoird, Nicolas; Khatri, Purvesh; Jansen, Pascal W. T. C.; Wilkinson, Alex W.; Liu, Shichong; Barbash, Olena; Van Aller, Glenn S.; Huddleston, Michael; Dhanak, Dashyant; Tummino, Peter J.; Kruger, Ryan G.; Garcia, Benjamin A.; Butte, Atul J.; Vermeulen, Michiel; Sage, Julien; Gozani, OrNature (London, United Kingdom) (2014), 510 (7504), 283-287CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Deregulation of lysine methylation signaling has emerged as a common etiol. factor in cancer pathogenesis, with inhibitors of several histone lysine methyltransferases (KMTs) being developed as chemotherapeutics. The largely cytoplasmic KMT SMYD3 (SET and MYND domain contg. protein 3) is overexpressed in numerous human tumors. However, the mol. mechanism by which SMYD3 regulates cancer pathways and its relationship to tumorigenesis in vivo are largely unknown. Here we show that methylation of MAP3K2 by SMYD3 increases MAP kinase signaling and promotes the formation of Ras-driven carcinomas. Using mouse models for pancreatic ductal adenocarcinoma and lung adenocarcinoma, we found that abrogating SMYD3 catalytic activity inhibits tumor development in response to oncogenic Ras. We used protein array technol. to identify the MAP3K2 kinase as a target of SMYD3. In cancer cell lines, SMYD3-mediated methylation of MAP3K2 at lysine 260 potentiates activation of the Ras/Raf/MEK/ERK signaling module and SMYD3 depletion synergizes with a MEK inhibitor to block Ras-driven tumorigenesis. Finally, the PP2A phosphatase complex, a key neg. regulator of the MAP kinase pathway, binds to MAP3K2 and this interaction is blocked by methylation. Together, our results elucidate a new role for lysine methylation in integrating cytoplasmic kinase-signaling cascades and establish a pivotal role for SMYD3 in the regulation of oncogenic Ras signaling.
- 2Bottino, C.; Peserico, A.; Simone, C.; Caretti, G. SMYD3: An Oncogenic Driver Targeting Epigenetic Regulation and Signaling Pathways. Cancers 2020, 12, 142, DOI: 10.3390/cancers12010142Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtValsLbF&md5=67e9371b4268abd2785789cc9c500664SMYD3: an oncogenic driver targeting epigenetic regulation and signaling pathwaysBottino, Cinzia; Peserico, Alessia; Simone, Cristiano; Caretti, GiuseppinaCancers (2020), 12 (1), 142CODEN: CANCCT; ISSN:2072-6694. (MDPI AG)SMYD3 is a member of the SMYD lysine methylase family and plays an important role in the methylation of various histone and non-histone targets. Aberrant SMYD3 expression contributes to carcinogenesis and SMYD3 upregulation was proposed as a prognostic marker in various solid cancers. Here we summarize SMYD3-mediated regulatory mechanisms, which are implicated in the pathophysiol. of cancer, as drivers of distinct oncogenic pathways. We describe SMYD3-dependent mechanisms affecting cancer progression, highlighting SMYD3 interplay with proteins and RNAs involved in the regulation of cancer cell proliferation, migration and invasion. We also address the effectiveness and mechanisms of action for the currently available SMYD3 inhibitors. The findings analyzed herein demonstrate that a complex network of SMYD3-mediated cytoplasmic and nuclear interactions promote oncogenesis across different cancer types. These evidences depict SMYD3 as a modulator of the transcriptional response and of key signaling pathways, orchestrating multiple oncogenic inputs and ultimately, promoting transcriptional reprogramming and tumor transformation. Further insights into the oncogenic role of SMYD3 and its targeting of different synergistic oncogenic signals may be beneficial for effective cancer treatment.
- 3Van Aller, G. S.; Graves, A. P.; Elkins, P. A.; Bonnette, W. G.; McDevitt, P. J.; Zappacosta, F.; Annan, R. S.; Dean, T. W.; Su, D. S.; Carpenter, C. L.; Mohammad, H. P.; Kruger, R. G. Structure-Based Design of a Novel SMYD3 Inhibitor That Bridges the Sam-and Mekk2-Binding Pockets. Structure 2016, 24, 774– 781, DOI: 10.1016/j.str.2016.03.010Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xlslelsrs%253D&md5=205b7374f1bdd97c71d832da92a08d62Structure-Based Design of a Novel SMYD3 Inhibitor that Bridges the SAM-and MEKK2-Binding PocketsVan Aller, Glenn S.; Graves, Alan P.; Elkins, Patricia A.; Bonnette, William G.; McDevitt, Patrick J.; Zappacosta, Francesca; Annan, Roland S.; Dean, Tony W.; Su, Dai-Shi; Carpenter, Christopher L.; Mohammad, Helai P.; Kruger, Ryan G.Structure (Oxford, United Kingdom) (2016), 24 (5), 774-781CODEN: STRUE6; ISSN:0969-2126. (Elsevier Ltd.)SMYD3 is a lysine methyltransferase overexpressed in colorectal, breast, prostate, and hepatocellular tumors, and has been implicated as an oncogene in human malignancies. Methylation of MEKK2 by SMYD3 is important for regulation of the MEK/ERK pathway, suggesting the possibility of selectively targeting SMYD3 in RAS-driven cancers. Structural and kinetic characterization of SMYD3 was undertaken leading to a co-crystal structure of SMYD3 with a MEKK2-peptide substrate bound, and the observation that SMYD3 follows a partially processive mechanism. These insights allowed for the design of GSK2807, a potent and selective, SAM-competitive inhibitor of SMYD3 (Ki = 14 nM). A high-resoln. crystal structure reveals that GSK2807 bridges the gap between the SAM-binding pocket and the substrate lysine tunnel of SMYD3. Taken together, our data demonstrate that small-mol. inhibitors of SMYD3 can be designed to prevent methylation of MEKK2 and these could have potential use as anticancer therapeutics.
- 4Su, D. S.; Qu, J.; Schulz, M.; Blackledge, C. W.; Yu, H.; Zeng, J.; Burgess, J.; Reif, A.; Stern, M.; Nagarajan, R.; Pappalardi, M. B.; Wong, K.; Graves, A. P.; Bonnette, W.; Wang, L.; Elkins, P.; Knapp-Reed, B.; Carson, J. D.; McHugh, C.; Mohammad, H.; Kruger, R.; Luengo, J.; Heerding, D. A.; Creasy, C. L. Discovery of Isoxazole Amides as Potent and Selective SMYD3 Inhibitors. ACS Med. Chem. Lett. 2020, 11, 133– 140, DOI: 10.1021/acsmedchemlett.9b00493Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXosF2g&md5=f6bce1620816d9050f03d22ba7c42e22Discovery of Isoxazole Amides as Potent and Selective SMYD3 InhibitorsSu, Dai-Shi; Qu, Junya; Schulz, Mark; Blackledge, Chuck W.; Yu, Hongyi; Zeng, Jenny; Burgess, Joelle; Reif, Alexander; Stern, Melissa; Nagarajan, Raman; Pappalardi, Melissa Baker; Wong, Kristen; Graves, Alan P.; Bonnette, William; Wang, Liping; Elkins, Patricia; Knapp-Reed, Beth; Carson, Jeffrey D.; McHugh, Charles; Mohammad, Helai; Kruger, Ryan; Luengo, Juan; Heerding, Dirk A.; Creasy, Caretha L.ACS Medicinal Chemistry Letters (2020), 11 (2), 133-140CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)We report herein the discovery of isoxazole amides as potent and selective SET and MYND Domain-Contg. Protein 3 (SMYD3) inhibitors. Elucidation of the structure-activity relationship of the high-throughput screening (HTS) lead compd. 1 provided potent and selective SMYD3 inhibitors. The SAR optimization, cocrystal structures of small mols. with SMYD3, and mode of inhibition (MOI) characterization of compds. are described. The synthesis and biol. and pharmacokinetic profiles of compds. are also presented.
- 5Schindler, C. E. M.; Baumann, H.; Blum, A.; Bose, D.; Buchstaller, H. P.; Burgdorf, L.; Cappel, D.; Chekler, E.; Czodrowski, P.; Dorsch, D.; Eguida, M. K. I.; Follows, B.; Fuchss, T.; Gradler, U.; Gunera, J.; Johnson, T.; Jorand Lebrun, C.; Karra, S.; Klein, M.; Knehans, T.; Koetzner, L.; Krier, M.; Leiendecker, M.; Leuthner, B.; Li, L.; Mochalkin, I.; Musil, D.; Neagu, C.; Rippmann, F.; Schiemann, K.; Schulz, R.; Steinbrecher, T.; Tanzer, E. M.; Unzue Lopez, A.; Viacava Follis, A.; Wegener, A.; Kuhn, D. Large-Scale Assessment of Binding Free Energy Calculations in Active Drug Discovery Projects. J. Chem. Inf. Model. 2020, 60, 5457– 5474, DOI: 10.1021/acs.jcim.0c00900Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1CktrbE&md5=f2372d56cd501717435ed8095dd0dbb0Large-Scale Assessment of Binding Free Energy Calculations in Active Drug Discovery ProjectsSchindler, Christina E. M.; Baumann, Hannah; Blum, Andreas; Boese, Dietrich; Buchstaller, Hans-Peter; Burgdorf, Lars; Cappel, Daniel; Chekler, Eugene; Czodrowski, Paul; Dorsch, Dieter; Eguida, Merveille K. I.; Follows, Bruce; Fuchss, Thomas; Graedler, Ulrich; Gunera, Jakub; Johnson, Theresa; Jorand Lebrun, Catherine; Karra, Srinivasa; Klein, Markus; Knehans, Tim; Koetzner, Lisa; Krier, Mireille; Leiendecker, Matthias; Leuthner, Birgitta; Li, Liwei; Mochalkin, Igor; Musil, Djordje; Neagu, Constantin; Rippmann, Friedrich; Schiemann, Kai; Schulz, Robert; Steinbrecher, Thomas; Tanzer, Eva-Maria; Unzue Lopez, Andrea; Viacava Follis, Ariele; Wegener, Ansgar; Kuhn, DanielJournal of Chemical Information and Modeling (2020), 60 (11), 5457-5474CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Accurate ranking of compds. with regards to their binding affinity to a protein using computational methods is of great interest to pharmaceutical research. Physics-based free energy calcns. are regarded as the most rigorous way to est. binding affinity. In recent years, many retrospective studies carried out both in academia and industry have demonstrated its potential. Here, we present the results of large-scale prospective application of the FEP+ method in active drug discovery projects in an industry setting at Merck KGaA, Darmstadt, Germany. We compare these prospective data to results obtained on a new diverse, public benchmark of eight pharmaceutically relevant targets. Our results offer insights into the challenges faced when using free energy calcns. in real-life drug discovery projects and identify limitations that could be tackled by future method development. The new public data set we provide to the community can support further method development and comparative benchmarking of free energy calcns.
- 6Ciordia, M.; Pérez-Benito, L.; Delgado, F.; Trabanco, A. A.; Tresadern, G. Application of Free Energy Perturbation for the Design of Bace1 Inhibitors. J. Chem. Inf. Model. 2016, 56, 1856– 1871, DOI: 10.1021/acs.jcim.6b00220Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xht12mu7%252FK&md5=b7577a89492867fc334cb1ec937a4eb6Application of Free Energy Perturbation for the Design of BACE1 InhibitorsCiordia, Myriam; Perez-Benito, Laura; Delgado, Francisca; Trabanco, Andres A.; Tresadern, GaryJournal of Chemical Information and Modeling (2016), 56 (9), 1856-1871CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Novel spiroaminodihydropyrroles probing for optimized interactions at the P3 pocket of β-secretase 1 (BACE1) were designed with the use of free energy perturbation (FEP) calcns. The resulting mols. showed pIC50 potencies in enzymic BACE1 inhibition assays ranging from approx. 5 to 7. Good correlation was obsd. between the predicted activity from the FEP calcns. and exptl. activity. Simulations run with a default 5 ns approach delivered a mean unsigned error (MUE) between prediction and expt. of 0.58 and 0.91 kcal/mol for retrospective and prospective applications, resp. With longer simulations of 10 and 20 ns, the MUE was in both cases 0.57 kcal/mol for the retrospective application, and 0.69 and 0.59 kcal/mol for the prospective application. Other considerations that impact the quality of the calcns. are discussed. This work provides an example of the value of FEP as a computational tool for drug discovery.
- 7Wang, 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 Scholar7https://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.
- 8Wan, S.; Bhati, A. P.; Zasada, S. J.; Coveney, P. V. Rapid, Accurate, Precise and Reproducible Ligand-Protein Binding Free Energy Prediction. Interface Focus 2020, 10, 20200007 DOI: 10.1098/rsfs.2020.0007Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3s3js1Wiuw%253D%253D&md5=9626db15b14559e08c35bfabc0716e4cRapid, accurate, precise and reproducible ligand-protein binding free energy predictionWan Shunzhou; Bhati Agastya P; Zasada Stefan J; Coveney Peter V; Coveney Peter VInterface focus (2020), 10 (6), 20200007 ISSN:2042-8898.A central quantity of interest in molecular biology and medicine is the free energy of binding of a molecule to a target biomacromolecule. Until recently, the accurate prediction of binding affinity had been widely regarded as out of reach of theoretical methods owing to the lack of reproducibility of the available methods, not to mention their complexity, computational cost and time-consuming procedures. The lack of reproducibility stems primarily from the chaotic nature of classical molecular dynamics (MD) and the associated extreme sensitivity of trajectories to their initial conditions. Here, we review computational approaches for both relative and absolute binding free energy calculations, and illustrate their application to a diverse set of ligands bound to a range of proteins with immediate relevance in a number of medical domains. We focus on ensemble-based methods which are essential in order to compute statistically robust results, including two we have recently developed, namely thermodynamic integration with enhanced sampling and enhanced sampling of MD with an approximation of continuum solvent. Together, these form a set of rapid, accurate, precise and reproducible free energy methods. They can be used in real-world problems such as hit-to-lead and lead optimization stages in drug discovery, and in personalized medicine. These applications show that individual binding affinities equipped with uncertainty quantification may be computed in a few hours on a massive scale given access to suitable high-end computing resources and workflow automation. A high level of accuracy can be achieved using these approaches.
- 9Wan, S.; Tresadern, G.; Pérez-Benito, L.; Vlijmen, H.; Coveney, P. V. Accuracy and Precision of Alchemical Relative Free Energy Predictions with and without Replica-Exchange. Adv. Theory Simul. 2019, 3, 1900195 DOI: 10.1002/adts.201900195Google ScholarThere is no corresponding record for this reference.
- 10Wan, 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 Scholar10https://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.
- 11Bhati, 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 Scholar11https://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.
- 12Kollman, 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., 3rd 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 Scholar12https://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.
- 13Coveney, 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 Scholar13https://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.
- 14Vassaux, M.; Wan, S.; Edeling, W.; Coveney, P. V. Ensembles Are Required to Handle Aleatoric and Parametric Uncertainty in Molecular Dynamics Simulation. J. Chem. Theory Comput. 2021, 17, 5187– 5197, DOI: 10.1021/acs.jctc.1c00526Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsFKktbjI&md5=12376b0c3d728bb7b54be27e450867f8Aleatoric and parametric uncertainty in molecular dynamics simulationVassaux, Maxime; Wan, Shunzhou; Edeling, Wouter; Coveney, Peter V.Journal of Chemical Theory and Computation (2021), 17 (8), 5187-5197CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Classical mol. dynamics is a computer simulation technique that is in widespread use across many areas of science, from physics and chem. to materials, biol. and medicine. The method continues to attract criticism due its oft-reported lack of reproducibility which is in part due to a failure to submit it to reliable uncertainty quantification (UQ). Here we show that the uncertainty arises from a combination of (i) the input parameters and (ii) the intrinsic stochasticity of the method controlled by the random seeds. To illustrate the situation, we make a systematic UQ anal. of a widely used mol. dynamics code (NAMD), applied to est. binding free energy of a ligand-bound to a protein. In particular, we replace the usually fixed input parameters with random variables, systematically distributed about their mean values, and study the resulting distribution of the simulation output. We also perform a sensitivity anal., which reveals that, out of a total of 175 parameters, just six dominate the variance in the code output. Furthermore, we show that binding energy calcn. damps the input uncertainty, in the sense that the variation around the mean output free energy is less than the variation around the mean of the assumed input distributions, if the output is ensemble-averaged over the random seeds. Without such ensemble averaging, the predicted free energy is five times more uncertain. The distribution of the predicted properties is thus strongly dependent upon the random seed. Owing to this substantial uncertainty, robust statistical measures of uncertainty in mol. dynamics simulation require the use of ensembles in all contexts.
- 15Wade, A.; Bhati, A. P.; Wan, S.; Coveney, P. V. Alchemical Free Energy Estimators and Molecular Dynamics Engines: Accuracy Precision and Reproducibility. ChemRxiv 2021, DOI: 10.26434/chemrxiv-2021-nqp8r .Google ScholarThere is no corresponding record for this reference.
- 16Bhati, A. P.; Coveney, P. V. Large Scale Study of Ligand-Protein Relative Binding Free Energy Calculations: Actionable Predictions from Statistically Robust Protocols. J. Chem. Theory Comput. 2022, 18, 2687– 2702, DOI: 10.1021/acs.jctc.1c01288Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XntVaqtL8%253D&md5=2b4f884275deced8f66f6b4a6b067b36Large Scale Study of Ligand-Protein Relative Binding Free Energy Calculations: Actionable Predictions from Statistically Robust ProtocolsBhati, Agastya P.; Coveney, Peter V.Journal of Chemical Theory and Computation (2022), 18 (4), 2687-2702CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The accurate and reliable prediction of protein-ligand binding affinities can play a central role in the drug discovery process as well as in personalized medicine. Of considerable importance during lead optimization are the alchem. free energy methods that furnish an estn. of relative binding free energies (RBFE) of similar mols. Recent advances in these methods have increased their speed, accuracy, and precision. This is evident from the increasing no. of retrospective as well as prospective studies employing them. However, such methods still have limited applicability in real-world scenarios due to a no. of important yet unresolved issues. Here, we report the findings from a large data set comprising over 500 ligand transformations spanning over 300 ligands binding to a diverse set of 14 different protein targets which furnish statistically robust results on the accuracy, precision, and reproducibility of RBFE calcns. We use ensemble-based methods which are the only way to provide reliable uncertainty quantification given that the underlying mol. dynamics is chaotic. These are implemented using TIES (Thermodn. Integration with Enhanced Sampling). Results achieve chem. accuracy in all cases. Ensemble simulations also furnish information on the statistical distributions of the free energy calcns. which exhibit non-normal behavior. We find that the "enhanced sampling" method known as replica exchange with solute tempering degrades RBFE predictions. We also report definitively on numerous assocd. alchem. factors including the choice of ligand charge method, flexibility in ligand structure, and the size of the alchem. region including the no. of atoms involved in transforming one ligand into another. Our findings provide a key set of recommendations that should be adopted for the reliable application of RBFE methods.
- 17Knapp, B.; Ospina, L.; Deane, C. M. Avoiding False Positive Conclusions in Molecular Simulation: The Importance of Replicas. J. Chem. Theory Comput. 2018, 14, 6127– 6138, DOI: 10.1021/acs.jctc.8b00391Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFygsb%252FN&md5=6ed83fe1ea4a74d6739c2cc521fcfc70Avoiding False Positive Conclusions in Molecular Simulation: The Importance of ReplicasKnapp, Bernhard; Ospina, Luis; Deane, Charlotte M.Journal of Chemical Theory and Computation (2018), 14 (12), 6127-6138CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Mol. simulations are a computational technique used to investigate the dynamics of proteins and other mols. The free energy landscape of these simulations is often rugged, and minor differences in the initial velocities, floating-point precision, or underlying hardware can cause identical simulations (replicas) to take different paths in the landscape. In this study the authors investigated the magnitude of these effects based on 310 000 ns of simulation time. The authors performed 100 identically parametrized replicas of 3000 ns each for a small 10 amino acid system as well as 100 identically parametrized replicas of 100 ns each for an 827 residue T-cell receptor/MHC system. Comparing randomly chosen subgroups within these replica sets, the authors estd. the reproducibility and reliability that can be achieved by a given no. of replicas at a given simulation time. These results demonstrate that conclusions drawn from single simulations are often not reproducible and that conclusions drawn from multiple shorter replicas are more reliable than those from a single longer simulation. The actual no. of replicas needed will always depend on the question asked and the level of reliability sought. On the basis of the data, it appears that a good rule of thumb is to perform a min. of five to 10 replicas.
- 18Wan, S.; Sinclair, R. C.; Coveney, P. V. Uncertainty Quantification in Classical Molecular Dynamics. Philos. Trans. R. Soc., A 2021, 379, 20200082 DOI: 10.1098/rsta.2020.0082Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXptFentrY%253D&md5=d2b00cf950ff1e1b69ab4d468dfa5cfeUncertainty quantification in classical molecular dynamicsWan, Shunzhou; Sinclair, Robert C.; Coveney, Peter V.Philosophical Transactions of the Royal Society, A: Mathematical, Physical & Engineering Sciences (2021), 379 (2197), 20200082CODEN: PTRMAD; ISSN:1364-503X. (Royal Society)Mol. dynamics simulation is now a widespread approach for understanding complex systems on the atomistic scale. It finds applications from physics and chem. to engineering, life and medical science. In the last decade, the approach has begun to advance from being a computer-based means of rationalizing exptl. observations to producing apparently credible predictions for a no. of real-world applications within industrial sectors such as advanced materials and drug discovery. However, key aspects concerning the reproducibility of the method have not kept pace with the speed of its uptake in the scientific community. Here, we present a discussion of uncertainty quantification for mol. dynamics simulation designed to endow the method with better error ests. that will enable it to be used to report actionable results. The approach adopted is a std. one in the field of uncertainty quantification, namely using ensemble methods, in which a sufficiently large no. of replicas are run concurrently, from which reliable statistics can be extd. Indeed, because mol. dynamics is intrinsically chaotic, the need to use ensemble methods is fundamental and holds regardless of the duration of the simulations performed. We discuss the approach and illustrate it in a range of applications from materials science to ligand-protein binding free energy estn.
- 19Bieniek, M. K.; Bhati, A. P.; Wan, S.; Coveney, P. V. TIES 20: Relative Binding Free Energy with a Flexible Superimposition Algorithm and Partial Ring Morphing. J. Chem. Theory Comput. 2021, 17, 1250– 1265, DOI: 10.1021/acs.jctc.0c01179Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVWks7w%253D&md5=fdeea48a835674ad89813380561ad2bfTIES 20: Relative Binding Free Energy with a Flexible Superimposition Algorithm and Partial Ring MorphingBieniek, Matuesz K.; Bhati, Agastya P.; Wan, Shunzhou; Coveney, Peter V.Journal of Chemical Theory and Computation (2021), 17 (2), 1250-1265CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The TIES (Thermodn. Integration with Enhanced Sampling) protocol is a formally exact alchem. approach in computational chem. to the calcn. of relative binding free energies. The validity of TIES relies on the correctness of matching atoms across compared pairs of ligands, laying the foundation for the transformation along an alchem. pathway. We implement a flexible topol. superimposition algorithm which uses an exhaustive joint-traversal for computing the largest common component(s). The algorithm is employed to enable matching and morphing of partial rings in the TIES protocol along with a validation study using 55 transformations and five different proteins from our previous work. We find that TIES 20 with the RESP charge system, using the new superimposition algorithm, reproduces the previous results with mean unsigned error of 0.75 kcal/mol with respect to the exptl. data. Enabling the morphing of partial rings decreases the size of the alchem. region in the dual-topol. transformations resulting in a significant improvement in the prediction precision. We find that increasing the ensemble size from 5 to 20 replicas per λ window only has a minimal impact on the accuracy. However, the non-normal nature of the relative free energy distributions underscores the importance of ensemble simulation. We further compare the results with the AM1-BCC charge system and show that it improves agreement with the exptl. data by slightly over 10%. This improvement is partly due to AM1-BCC affecting only the charges of the atoms local to the mutation, which translates to even fewer morphed atoms, consequently reducing issues with sampling and therefore ensemble averaging. TIES 20, in conjunction with the enablement of ring morphing, reduces the size of the alchem. region and significantly improves the precision of the predicted free energies.
- 20Sadiq, 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 Scholar20https://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.
- 21Suruzhon, M.; Bodnarchuk, M. S.; Ciancetta, A.; Viner, R.; Wall, I. D.; Essex, J. W. Sensitivity of Binding Free Energy Calculations to Initial Protein Crystal Structure. J. Chem. Theory Comput. 2021, 17, 1806– 1821, DOI: 10.1021/acs.jctc.0c00972Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXislGjsr8%253D&md5=7e1754a44e1387726c524896f071247fSensitivity of binding free energy calculations to initial protein crystal structureSuruzhon, Miroslav; Bodnarchuk, Michael S.; Ciancetta, Antonella; Viner, Russell; Wall, Ian D.; Essex, Jonathan W.Journal of Chemical Theory and Computation (2021), 17 (3), 1806-1821CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Binding free energy calcns. using alchem. free energy (AFE) methods are widely considered to be the most rigorous tool in the computational drug discovery arsenal. Despite this, the calcns. suffer from accuracy, precision, and reproducibility issues. In this publication, the authors perform a high-throughput study of more than a thousand AFE calcns., utilizing over 220μs of total sampling time, on three different protein systems to investigate the impact of the initial crystal structure on the resulting binding free energy values. The authors also consider the influence of equilibration time and discover that the initial crystal structure can have a significant effect on free energy values obtained at short timescales that can manifest itself as a free energy difference of more than 1 kcal/mol. At longer timescales, these differences are largely overtaken by important rare events, such as torsional ligand motions, typically resulting in a much higher uncertainty in the obtained values. This work emphasizes the importance of rare event sampling and long-timescale dynamics in free energy calcns. even for routinely performed alchem. perturbations. The authors conclude that an optimal protocol should not only conc. computational resources on achieving convergence in the alchem. coupling parameter (λ) space but also on longer simulations and multiple repeats.
- 22Pérez-Benito, L.; Keränen, H.; van Vlijmen, H.; Tresadern, G. Predicting Binding Free Energies of PDE2 Inhibitors. The Difficulties of Protein Conformation. Sci. Rep. 2018, 8, 4883, DOI: 10.1038/s41598-018-23039-5Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MnjtFKlsA%253D%253D&md5=44c26c5bd2fe3e35bba344abb8262eebPredicting Binding Free Energies of PDE2 Inhibitors. The Difficulties of Protein ConformationPerez-Benito Laura; Keranen Henrik; van Vlijmen Herman; Tresadern Gary; Keranen HenrikScientific reports (2018), 8 (1), 4883 ISSN:.A congeneric series of 21 phosphodiesterase 2 (PDE2) inhibitors are reported. Crystal structures show how the molecules can occupy a 'top-pocket' of the active site. Molecules with small substituents do not enter the pocket, a critical leucine (Leu770) is closed and water molecules are present. Large substituents enter the pocket, opening the Leu770 conformation and displacing the waters. We also report an X-ray structure revealing a new conformation of the PDE2 active site domain. The relative binding affinities of these compounds were studied with free energy perturbation (FEP) methods and it represents an attractive real-world test case. In general, the calculations could predict the energy of small-to-small, or large-to-large molecule perturbations. However, accurately capturing the transition from small-to-large proved challenging. Only when using alternative protein conformations did results improve. The new X-ray structure, along with a modelled dimer, conferred stability to the catalytic domain during the FEP molecular dynamics (MD) simulations, increasing the convergence and thereby improving the prediction of ΔΔG of binding for some small-to-large transitions. In summary, we found the most significant improvement in results when using different protein structures, and this data set is useful for future free energy validation studies.
- 23Friesner, R. A.; Murphy, R. B.; Repasky, M. P.; Frye, L. L.; Greenwood, J. R.; Halgren, T. A.; Sanschagrin, P. C.; Mainz, D. T. Extra Precision Glide: Docking and Scoring Incorporating a Model of Hydrophobic Enclosure for Protein-Ligand Complexes. J. Med. Chem. 2006, 49, 6177– 6196, DOI: 10.1021/jm051256oGoogle Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XpvVGmurg%253D&md5=ea428c82ead0d8c27f8c1a7b694a1edfExtra Precision Glide: Docking and Scoring Incorporating a Model of Hydrophobic Enclosure for Protein-Ligand ComplexesFriesner, Richard A.; Murphy, Robert B.; Repasky, Matthew P.; Frye, Leah L.; Greenwood, Jeremy R.; Halgren, Thomas A.; Sanschagrin, Paul C.; Mainz, Daniel T.Journal of Medicinal Chemistry (2006), 49 (21), 6177-6196CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A novel scoring function to est. protein-ligand binding affinities has been developed and implemented as the Glide 4.0 XP scoring function and docking protocol. In addn. to unique water desolvation energy terms, protein-ligand structural motifs leading to enhanced binding affinity are included:(1) hydrophobic enclosure where groups of lipophilic ligand atoms are enclosed on opposite faces by lipophilic protein atoms, (2) neutral-neutral single or correlated hydrogen bonds in a hydrophobically enclosed environment, and (3) five categories of charged-charged hydrogen bonds. The XP scoring function and docking protocol have been developed to reproduce exptl. binding affinities for a set of 198 complexes (RMSDs of 2.26 and 1.73 kcal/mol over all and well-docked ligands, resp.) and to yield quality enrichments for a set of fifteen screens of pharmaceutical importance. Enrichment results demonstrate the importance of the novel XP mol. recognition and water scoring in sepg. active and inactive ligands and avoiding false positives.
- 24Case, D. A.; Cheatham, T. E., 3rd; Darden, T.; Gohlke, H.; Luo, R.; Merz, K. M., Jr.; Onufriev, A.; Simmerling, C.; Wang, B.; Woods, R. J. The Amber Biomolecular Simulation Programs. J. Comput. Chem. 2005, 26, 1668– 1688, DOI: 10.1002/jcc.20290Google Scholar24https://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.
- 25Zhang, X.; Perez-Sanchez, H.; Lightstone, F. C. A Comprehensive Docking and MM/GBSA Rescoring Study of Ligand Recognition Upon Binding Antithrombin. Curr. Top. Med. Chem. 2017, 17, 1631– 1639, DOI: 10.2174/1568026616666161117112604Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXmt1eht7s%253D&md5=c1baa4455076543f619a194bc5559bffA Comprehensive Docking and MM/GBSA Rescoring Study of Ligand Recognition upon Binding AntithrombinZhang, Xiaohua; Perez-Sanchez, Horacio; Lightstone, Felice C.Current Topics in Medicinal Chemistry (Sharjah, United Arab Emirates) (2017), 17 (14), 1631-1639CODEN: CTMCCL; ISSN:1568-0266. (Bentham Science Publishers Ltd.)Background: A high-throughput virtual screening pipeline has been extended from single energetically minimized structure Mol. Mechanics/Generalized Born Surface Area (MM/GBSA) rescoring to ensemble-av. MM/GBSA rescoring. The correlation coeff. (R2) of calcd. and exptl. binding free energies for a series of antithrombin ligands has been improved from 0.36 to 0.69 when switching from the single-structure MM/GBSA rescoring to ensemble-av. one. The electrostatic interactions in both solute and solvent are identified to play an important role in detg. the binding free energy after the decompn. of the calcd. binding free energy. The increasing neg. charge of the compds. provides a more favorable electrostatic energy change but creates a higher penalty for the solvation free energy. Such a penalty is compensated by the electrostatic energy change, which results in a better binding affinity. A highly hydrophobic ligand is detd. by the docking program to be a non-specific binder. Results: Our results have demonstrated that it is very important to keep a few top poses for rescoring, if the binding is non-specific or the binding mode is not well detd. by the docking calcn.
- 26Wright, D. W.; Husseini, F.; Wan, S.; Meyer, C.; van Vlijmen, H.; Tresadern, G.; Coveney, P. V. Application of the ESMACS Binding Free Energy Protocol to a Multi-Binding Site Lactate Dehydogenase a Ligand Dataset. Adv. Theory Simul. 2019, 3, 1900194 DOI: 10.1002/adts.201900194Google ScholarThere is no corresponding record for this reference.
- 27Wright, D. W.; Wan, S.; Meyer, C.; van Vlijmen, H.; Tresadern, G.; Coveney, P. V. Application of ESMACS Binding Free Energy Protocols to Diverse Datasets: Bromodomain-Containing Protein 4. Sci. Rep. 2019, 9, 6017, DOI: 10.1038/s41598-019-41758-1Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M%252FkvFaqsQ%253D%253D&md5=b62c5a234b09797af7f8e21846c7815fApplication of ESMACS binding free energy protocols to diverse datasets: Bromodomain-containing protein 4Wright David W; Wan Shunzhou; Coveney Peter V; Meyer Christophe; van Vlijmen Herman; Tresadern GaryScientific reports (2019), 9 (1), 6017 ISSN:.As the application of computational methods in drug discovery pipelines becomes more widespread it is increasingly important to understand how reproducible their results are and how sensitive they are to choices made in simulation setup and analysis. Here we use ensemble simulation protocols, termed ESMACS (enhanced sampling of molecular dynamics with approximation of continuum solvent), to investigate the sensitivity of the popular molecular mechanics Poisson-Boltzmann surface area (MMPBSA) methodology. Using the bromodomain-containing protein 4 (BRD4) system bound to a diverse set of ligands as our target, we show that robust rankings can be produced only through combining ensemble sampling with multiple trajectories and enhanced solvation via an explicit ligand hydration shell.
- 28Wan, S.; Potterton, A.; Husseini, F. S.; Wright, D. W.; Heifetz, A.; Malawski, M.; Townsend-Nicholson, A.; Coveney, P. V. Hit-to-Lead and Lead Optimization Binding Free Energy Calculations for G Protein-Coupled Receptors. Interface Focus 2020, 10, 20190128 DOI: 10.1098/rsfs.2019.0128Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3s3js1Witw%253D%253D&md5=cd117abc40d9d90d9878a35d49729109Hit-to-lead and lead optimization binding free energy calculations for G protein-coupled receptorsWan Shunzhou; Husseini Fouad S; Wright David W; Coveney Peter V; Potterton Andrew; Heifetz Alexander; Townsend-Nicholson Andrea; Heifetz Alexander; Malawski Maciej; Coveney Peter VInterface focus (2020), 10 (6), 20190128 ISSN:2042-8898.We apply the hit-to-lead ESMACS (enhanced sampling of molecular dynamics with approximation of continuum solvent) and lead-optimization TIES (thermodynamic integration with enhanced sampling) methods to compute the binding free energies of a series of ligands at the A1 and A2A adenosine receptors, members of a subclass of the GPCR (G protein-coupled receptor) superfamily. Our predicted binding free energies, calculated using ESMACS, show a good correlation with previously reported experimental values of the ligands studied. Relative binding free energies, calculated using TIES, accurately predict experimentally determined values within a mean absolute error of approximately 1 kcal mol(-1). Our methodology may be applied widely within the GPCR superfamily and to other small molecule-receptor protein systems.
- 29Wright, 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 Scholar29https://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.
- 30Phillips, 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 Scholar30https://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.
- 31Beveridge, D. L.; Dicapua, F. M. Free-Energy Via Molecular Simulation - Applications to Chemical and Biomolecular Systems. Annu. Rev. Biophys. Bioeng. 1989, 18, 431– 492, DOI: 10.1146/annurev.bb.18.060189.002243Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXlvVajsLs%253D&md5=396c2dbf6a5c78a579badf764f070c0dFree energy via molecular simulation: applications to chemical and biomolecular systemsBeveridge, D. L.; DiCapua, F. M.Annual Review of Biophysics and Biophysical Chemistry (1989), 18 (), 431-92CODEN: ARBCEY; ISSN:0883-9182.A review with over 250 refs. General approaches to free energy detns. by mol. simulation are formulated theor. and the practical uses are discussed. In general, the agreement between calcd. and exptl. data is good when some exptl. data are used in parametrization of the intermol. potentials. The applications of the methods are also discussed.
- 32Genheden, S.; Ryde, U. How to Obtain Statistically Converged MM/GBSA Results. J. Comput. Chem. 2010, 31, 837– 846, DOI: 10.1002/jcc.21366Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtlentbY%253D&md5=173f37e54d4f0c80a8ec62df78f95127How to obtain statistically converged MM/GBSA resultsGenheden, Samuel; Ryde, UlfJournal of Computational Chemistry (2010), 31 (4), 837-846CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)The mol. mechanics/generalized Born surface area (MM/GBSA) method has been investigated with the aim of achieving a statistical precision of 1 kJ/mol for the results. The authors studied the binding of seven biotin analogs to avidin, taking advantage of the fact that the protein is a tetramer with four independent binding sites, which should give the same estd. binding affinities. The authors show that it is not enough to use a single long simulation (10 ns), because the std. error of such a calcn. underestimates the difference between the four binding sites. Instead, it is better to run several independent simulations and av. the results. With such an approach, the authors obtain the same results for the four binding sites, and any desired precision can be obtained by running a proper no. of simulations. The authors discuss how the simulations should be performed to optimize the use of computer time. The correlation time between the MM/GBSA energies is ∼5 ps and an equilibration time of 100 ps is needed. For MM/GBSA, the authors recommend a sampling time of 20-200 ps for each sep. simulation, depending on the protein. With 200 ps prodn. time, 5-50 sep. simulations are required to reach a statistical precision of 1 kJ/mol (800-8000 energy calcns. or 1.5-15 ns total simulation time per ligand) for the seven avidin ligands. This is an order of magnitude more than what is normally used, but such a no. of simulations is needed to obtain statistically valid results for the MM/GBSA method. © 2009 Wiley Periodicals, Inc. J Comput Chem 2010.
- 33Sadiq, S. K.; Wright, D. W.; Kenway, O. A.; Coveney, P. V. Accurate Ensemble Molecular Dynamics Binding Free Energy Ranking of Multidrug-Resistant HIV-1 Proteases. J. Chem. Inf. Model. 2010, 50, 890– 905, DOI: 10.1021/ci100007wGoogle Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksFeqtb4%253D&md5=4a9c06d974aad014a9a5e027a08a76acAccurate Ensemble Molecular Dynamics Binding Free Energy Ranking of Multidrug-Resistant HIV-1 ProteasesSadiq, S. Kashif; Wright, David W.; Kenway, Owain A.; Coveney, Peter V.Journal of Chemical Information and Modeling (2010), 50 (5), 890-905CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Accurate calcn. of important thermodn. properties, such as macromol. binding free energies, is one of the principal goals of mol. dynamics simulations. However, single long simulation frequently produces incorrectly converged quant. results due to inadequate sampling of conformational space in a feasible wall-clock time. Multiple short (ensemble) simulations have been shown to explore conformational space more effectively than single long simulations, but the two methods have not yet been thermodynamically compared. Here we show that, for end-state binding free energy detn. methods, ensemble simulations exhibit significantly enhanced thermodn. sampling over single long simulations and result in accurate and converged relative binding free energies that are reproducible to within 0.5 kcal/mol. Completely correct ranking is obtained for six HIV-1 protease variants bound to lopinavir with a correlation coeff. of 0.89 and a mean relative deviation from expt. of 0.9 kcal/mol. Multidrug resistance to lopinavir is enthalpically driven and increases through a decrease in the protein-ligand van der Waals interaction, principally due to the V82A/I84V mutation, and an increase in net electrostatic repulsion due to water-mediated disruption of protein-ligand interactions in the catalytic region. Furthermore, we correctly rank, to within 1 kcal/mol of expt., the substantially increased chem. potency of lopinavir binding to the wild-type protease compared to saquinavir and show that lopinavir takes advantage of a decreased net electrostatic repulsion to confer enhanced binding. Our approach is dependent on the combined use of petascale computing resources and on an automated simulation workflow to attain the required level of sampling and turn around time to obtain the results, which can be as little as three days. This level of performance promotes integration of such methodol. with clin. decision support systems for the optimization of patient-specific therapy.
- 34Wan, S.; Bhati, A. P.; Skerratt, S.; Omoto, K.; Shanmugasundaram, V.; Bagal, S. K.; Coveney, P. V. Evaluation and Characterization of Trk Kinase Inhibitors for the Treatment of Pain: Reliable Binding Affinity Predictions from Theory and Computation. J. Chem. Inf. Model. 2017, 57, 897– 909, DOI: 10.1021/acs.jcim.6b00780Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXksVyrurc%253D&md5=2e366f56c1ee8c2f2b2760b9098e2030Evaluation and Characterization of Trk Kinase Inhibitors for the Treatment of Pain: Reliable Binding Affinity Predictions from Theory and ComputationWan, Shunzhou; Bhati, Agastya P.; Skerratt, Sarah; Omoto, Kiyoyuki; Shanmugasundaram, Veerabahu; Bagal, Sharan K.; Coveney, Peter V.Journal of Chemical Information and Modeling (2017), 57 (4), 897-909CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Optimization of ligand binding affinity to the target protein of interest is a primary objective in small-mol. drug discovery. Until now, the prediction of binding affinities by computational methods has not been widely applied in the drug discovery process, mainly due to its lack of accuracy and reproducibility, as well as the long turnaround times required to obtain results. Herein, the authors report on a collaborative study that compares tropomyosin receptor kinase A (TrkA) binding affinity predictions using two recently formulated fast computational approaches - namely ESMACS (Enhanced Sampling of Mol. dynamics with Approxn. of Continuum Solvent) and TIES (Thermodn. Integration with Enhanced Sampling) - to exptl. derived TrkA binding affinities for a set of Pfizer pan-Trk compds. ESMACS gives precise and reproducible results and is applicable to highly diverse sets of compds. It also provides detailed chem. insight into the nature of ligand-protein binding. TIES can predict and thus optimize more subtle changes in binding affinities between compds. of similar structure. Individual binding affinities were calcd. in a few hours, exhibiting good correlations with the exptl. data of 0.79 and 0.88 from ESMACS and TIES approaches resp. The speed, level of accuracy and precision of the calcns. are such that the affinity predictions can be used to rapidly explain the effects of compd. 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 compds. and targets.
- 35Wan, 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 Scholar35https://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.
- 36Scheen, J.; Wu, W.; Mey, A.; Tosco, P.; Mackey, M.; Michel, J. Hybrid Alchemical Free Energy/Machine-Learning Methodology for the Computation of Hydration Free Energies. J. Chem. Inf. Model. 2020, 60, 5331– 5339, DOI: 10.1021/acs.jcim.0c00600Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlGnsbvF&md5=5168eabd0e51fb6ddb40f0d782171458Hybrid Alchemical Free Energy/Machine-Learning Methodology for the Computation of Hydration Free EnergiesScheen, Jenke; Wu, Wilson; Mey, Antonia S. J. S.; Tosco, Paolo; Mackey, Mark; Michel, JulienJournal of Chemical Information and Modeling (2020), 60 (11), 5331-5339CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)A methodol. that combines alchem. free energy calcns. (FEP) with machine learning (ML) has been developed to compute accurate abs. hydration free energies. The hybrid FEP/ML methodol. was trained on a subset of the FreeSolv database, and retrospectively shown to outperform most submissions from the SAMPL4 competition. Compared to pure machine-learning approaches, FEP/ML yields more precise ests. of free energies of hydration, and requires a fraction of the training set size to outperform standalone FEP calcns. The ML-derived correction terms are further shown to be transferable to a range of related FEP simulation protocols. The approach may be used to inexpensively improve the accuracy of FEP calcns., and to flag mols. which will benefit the most from bespoke forcefield parameterization efforts.
- 37Frost, C.; Thompson, S. G. Correcting for Regression Dilution Bias: Comparison of Methods for a Single Predictor Variable. J. R. Stat. Soc. 2000, 163, 173– 189, DOI: 10.1111/1467-985X.00164Google ScholarThere is no corresponding record for this reference.
- 38Ruff, T. W.; Neelin, J. D. Long Tails in Regional Surface Temperature Probability Distributions with Implications for Extremes under Global Warming. Geophys. Res. Lett. 2012, 39, L04704, DOI: 10.1029/2011GL050610Google ScholarThere is no corresponding record for this reference.
- 39Weitzman, M. L. Fat-Tailed Uncertainty in the Economics of Catastrophic Climate Change. Rev. Environ. Econ. Policy 2011, 5, 275– 292, DOI: 10.1093/reep/rer006Google ScholarThere is no corresponding record for this reference.
Cited By
This article is cited by 5 publications.
- Hannes H. Loeffler, Shunzhou Wan, Marco Klähn, Agastya P. Bhati, Peter V. Coveney. Optimal Molecular Design: Generative Active Learning Combining REINVENT with Precise Binding Free Energy Ranking Simulations. Journal of Chemical Theory and Computation 2024, 20
(18)
, 8308-8328. https://doi.org/10.1021/acs.jctc.4c00576
- Shunzhou Wan, Agastya P. Bhati, Alexander D. Wade, Peter V. Coveney. Ensemble-Based Approaches Ensure Reliability and Reproducibility. Journal of Chemical Information and Modeling 2023, 63
(22)
, 6959-6963. https://doi.org/10.1021/acs.jcim.3c01654
- Shunzhou Wan, Agastya P. Bhati, Peter V. Coveney. Comparison of Equilibrium and Nonequilibrium Approaches for Relative Binding Free Energy Predictions. Journal of Chemical Theory and Computation 2023, 19
(21)
, 7846-7860. https://doi.org/10.1021/acs.jctc.3c00842
- 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, 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
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 39 other publications.
- 1Mazur, P. K.; Reynoird, N.; Khatri, P.; Jansen, P. W.; Wilkinson, A. W.; Liu, S.; Barbash, O.; Van Aller, G. S.; Huddleston, M.; Dhanak, D.; Tummino, P. J.; Kruger, R. G.; Garcia, B. A.; Butte, A. J.; Vermeulen, M.; Sage, J.; Gozani, O. SMYD3 Links Lysine Methylation of Map3k2 to Ras-Driven Cancer. Nature 2014, 510, 283– 287, DOI: 10.1038/nature133201https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXps1Wmtbc%253D&md5=009705b03f725a45664d3feb74f92f7aSMYD3 links lysine methylation of MAP3K2 to Ras-driven cancerMazur, Pawel K.; Reynoird, Nicolas; Khatri, Purvesh; Jansen, Pascal W. T. C.; Wilkinson, Alex W.; Liu, Shichong; Barbash, Olena; Van Aller, Glenn S.; Huddleston, Michael; Dhanak, Dashyant; Tummino, Peter J.; Kruger, Ryan G.; Garcia, Benjamin A.; Butte, Atul J.; Vermeulen, Michiel; Sage, Julien; Gozani, OrNature (London, United Kingdom) (2014), 510 (7504), 283-287CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Deregulation of lysine methylation signaling has emerged as a common etiol. factor in cancer pathogenesis, with inhibitors of several histone lysine methyltransferases (KMTs) being developed as chemotherapeutics. The largely cytoplasmic KMT SMYD3 (SET and MYND domain contg. protein 3) is overexpressed in numerous human tumors. However, the mol. mechanism by which SMYD3 regulates cancer pathways and its relationship to tumorigenesis in vivo are largely unknown. Here we show that methylation of MAP3K2 by SMYD3 increases MAP kinase signaling and promotes the formation of Ras-driven carcinomas. Using mouse models for pancreatic ductal adenocarcinoma and lung adenocarcinoma, we found that abrogating SMYD3 catalytic activity inhibits tumor development in response to oncogenic Ras. We used protein array technol. to identify the MAP3K2 kinase as a target of SMYD3. In cancer cell lines, SMYD3-mediated methylation of MAP3K2 at lysine 260 potentiates activation of the Ras/Raf/MEK/ERK signaling module and SMYD3 depletion synergizes with a MEK inhibitor to block Ras-driven tumorigenesis. Finally, the PP2A phosphatase complex, a key neg. regulator of the MAP kinase pathway, binds to MAP3K2 and this interaction is blocked by methylation. Together, our results elucidate a new role for lysine methylation in integrating cytoplasmic kinase-signaling cascades and establish a pivotal role for SMYD3 in the regulation of oncogenic Ras signaling.
- 2Bottino, C.; Peserico, A.; Simone, C.; Caretti, G. SMYD3: An Oncogenic Driver Targeting Epigenetic Regulation and Signaling Pathways. Cancers 2020, 12, 142, DOI: 10.3390/cancers120101422https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtValsLbF&md5=67e9371b4268abd2785789cc9c500664SMYD3: an oncogenic driver targeting epigenetic regulation and signaling pathwaysBottino, Cinzia; Peserico, Alessia; Simone, Cristiano; Caretti, GiuseppinaCancers (2020), 12 (1), 142CODEN: CANCCT; ISSN:2072-6694. (MDPI AG)SMYD3 is a member of the SMYD lysine methylase family and plays an important role in the methylation of various histone and non-histone targets. Aberrant SMYD3 expression contributes to carcinogenesis and SMYD3 upregulation was proposed as a prognostic marker in various solid cancers. Here we summarize SMYD3-mediated regulatory mechanisms, which are implicated in the pathophysiol. of cancer, as drivers of distinct oncogenic pathways. We describe SMYD3-dependent mechanisms affecting cancer progression, highlighting SMYD3 interplay with proteins and RNAs involved in the regulation of cancer cell proliferation, migration and invasion. We also address the effectiveness and mechanisms of action for the currently available SMYD3 inhibitors. The findings analyzed herein demonstrate that a complex network of SMYD3-mediated cytoplasmic and nuclear interactions promote oncogenesis across different cancer types. These evidences depict SMYD3 as a modulator of the transcriptional response and of key signaling pathways, orchestrating multiple oncogenic inputs and ultimately, promoting transcriptional reprogramming and tumor transformation. Further insights into the oncogenic role of SMYD3 and its targeting of different synergistic oncogenic signals may be beneficial for effective cancer treatment.
- 3Van Aller, G. S.; Graves, A. P.; Elkins, P. A.; Bonnette, W. G.; McDevitt, P. J.; Zappacosta, F.; Annan, R. S.; Dean, T. W.; Su, D. S.; Carpenter, C. L.; Mohammad, H. P.; Kruger, R. G. Structure-Based Design of a Novel SMYD3 Inhibitor That Bridges the Sam-and Mekk2-Binding Pockets. Structure 2016, 24, 774– 781, DOI: 10.1016/j.str.2016.03.0103https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xlslelsrs%253D&md5=205b7374f1bdd97c71d832da92a08d62Structure-Based Design of a Novel SMYD3 Inhibitor that Bridges the SAM-and MEKK2-Binding PocketsVan Aller, Glenn S.; Graves, Alan P.; Elkins, Patricia A.; Bonnette, William G.; McDevitt, Patrick J.; Zappacosta, Francesca; Annan, Roland S.; Dean, Tony W.; Su, Dai-Shi; Carpenter, Christopher L.; Mohammad, Helai P.; Kruger, Ryan G.Structure (Oxford, United Kingdom) (2016), 24 (5), 774-781CODEN: STRUE6; ISSN:0969-2126. (Elsevier Ltd.)SMYD3 is a lysine methyltransferase overexpressed in colorectal, breast, prostate, and hepatocellular tumors, and has been implicated as an oncogene in human malignancies. Methylation of MEKK2 by SMYD3 is important for regulation of the MEK/ERK pathway, suggesting the possibility of selectively targeting SMYD3 in RAS-driven cancers. Structural and kinetic characterization of SMYD3 was undertaken leading to a co-crystal structure of SMYD3 with a MEKK2-peptide substrate bound, and the observation that SMYD3 follows a partially processive mechanism. These insights allowed for the design of GSK2807, a potent and selective, SAM-competitive inhibitor of SMYD3 (Ki = 14 nM). A high-resoln. crystal structure reveals that GSK2807 bridges the gap between the SAM-binding pocket and the substrate lysine tunnel of SMYD3. Taken together, our data demonstrate that small-mol. inhibitors of SMYD3 can be designed to prevent methylation of MEKK2 and these could have potential use as anticancer therapeutics.
- 4Su, D. S.; Qu, J.; Schulz, M.; Blackledge, C. W.; Yu, H.; Zeng, J.; Burgess, J.; Reif, A.; Stern, M.; Nagarajan, R.; Pappalardi, M. B.; Wong, K.; Graves, A. P.; Bonnette, W.; Wang, L.; Elkins, P.; Knapp-Reed, B.; Carson, J. D.; McHugh, C.; Mohammad, H.; Kruger, R.; Luengo, J.; Heerding, D. A.; Creasy, C. L. Discovery of Isoxazole Amides as Potent and Selective SMYD3 Inhibitors. ACS Med. Chem. Lett. 2020, 11, 133– 140, DOI: 10.1021/acsmedchemlett.9b004934https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXosF2g&md5=f6bce1620816d9050f03d22ba7c42e22Discovery of Isoxazole Amides as Potent and Selective SMYD3 InhibitorsSu, Dai-Shi; Qu, Junya; Schulz, Mark; Blackledge, Chuck W.; Yu, Hongyi; Zeng, Jenny; Burgess, Joelle; Reif, Alexander; Stern, Melissa; Nagarajan, Raman; Pappalardi, Melissa Baker; Wong, Kristen; Graves, Alan P.; Bonnette, William; Wang, Liping; Elkins, Patricia; Knapp-Reed, Beth; Carson, Jeffrey D.; McHugh, Charles; Mohammad, Helai; Kruger, Ryan; Luengo, Juan; Heerding, Dirk A.; Creasy, Caretha L.ACS Medicinal Chemistry Letters (2020), 11 (2), 133-140CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)We report herein the discovery of isoxazole amides as potent and selective SET and MYND Domain-Contg. Protein 3 (SMYD3) inhibitors. Elucidation of the structure-activity relationship of the high-throughput screening (HTS) lead compd. 1 provided potent and selective SMYD3 inhibitors. The SAR optimization, cocrystal structures of small mols. with SMYD3, and mode of inhibition (MOI) characterization of compds. are described. The synthesis and biol. and pharmacokinetic profiles of compds. are also presented.
- 5Schindler, C. E. M.; Baumann, H.; Blum, A.; Bose, D.; Buchstaller, H. P.; Burgdorf, L.; Cappel, D.; Chekler, E.; Czodrowski, P.; Dorsch, D.; Eguida, M. K. I.; Follows, B.; Fuchss, T.; Gradler, U.; Gunera, J.; Johnson, T.; Jorand Lebrun, C.; Karra, S.; Klein, M.; Knehans, T.; Koetzner, L.; Krier, M.; Leiendecker, M.; Leuthner, B.; Li, L.; Mochalkin, I.; Musil, D.; Neagu, C.; Rippmann, F.; Schiemann, K.; Schulz, R.; Steinbrecher, T.; Tanzer, E. M.; Unzue Lopez, A.; Viacava Follis, A.; Wegener, A.; Kuhn, D. Large-Scale Assessment of Binding Free Energy Calculations in Active Drug Discovery Projects. J. Chem. Inf. Model. 2020, 60, 5457– 5474, DOI: 10.1021/acs.jcim.0c009005https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1CktrbE&md5=f2372d56cd501717435ed8095dd0dbb0Large-Scale Assessment of Binding Free Energy Calculations in Active Drug Discovery ProjectsSchindler, Christina E. M.; Baumann, Hannah; Blum, Andreas; Boese, Dietrich; Buchstaller, Hans-Peter; Burgdorf, Lars; Cappel, Daniel; Chekler, Eugene; Czodrowski, Paul; Dorsch, Dieter; Eguida, Merveille K. I.; Follows, Bruce; Fuchss, Thomas; Graedler, Ulrich; Gunera, Jakub; Johnson, Theresa; Jorand Lebrun, Catherine; Karra, Srinivasa; Klein, Markus; Knehans, Tim; Koetzner, Lisa; Krier, Mireille; Leiendecker, Matthias; Leuthner, Birgitta; Li, Liwei; Mochalkin, Igor; Musil, Djordje; Neagu, Constantin; Rippmann, Friedrich; Schiemann, Kai; Schulz, Robert; Steinbrecher, Thomas; Tanzer, Eva-Maria; Unzue Lopez, Andrea; Viacava Follis, Ariele; Wegener, Ansgar; Kuhn, DanielJournal of Chemical Information and Modeling (2020), 60 (11), 5457-5474CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Accurate ranking of compds. with regards to their binding affinity to a protein using computational methods is of great interest to pharmaceutical research. Physics-based free energy calcns. are regarded as the most rigorous way to est. binding affinity. In recent years, many retrospective studies carried out both in academia and industry have demonstrated its potential. Here, we present the results of large-scale prospective application of the FEP+ method in active drug discovery projects in an industry setting at Merck KGaA, Darmstadt, Germany. We compare these prospective data to results obtained on a new diverse, public benchmark of eight pharmaceutically relevant targets. Our results offer insights into the challenges faced when using free energy calcns. in real-life drug discovery projects and identify limitations that could be tackled by future method development. The new public data set we provide to the community can support further method development and comparative benchmarking of free energy calcns.
- 6Ciordia, M.; Pérez-Benito, L.; Delgado, F.; Trabanco, A. A.; Tresadern, G. Application of Free Energy Perturbation for the Design of Bace1 Inhibitors. J. Chem. Inf. Model. 2016, 56, 1856– 1871, DOI: 10.1021/acs.jcim.6b002206https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xht12mu7%252FK&md5=b7577a89492867fc334cb1ec937a4eb6Application of Free Energy Perturbation for the Design of BACE1 InhibitorsCiordia, Myriam; Perez-Benito, Laura; Delgado, Francisca; Trabanco, Andres A.; Tresadern, GaryJournal of Chemical Information and Modeling (2016), 56 (9), 1856-1871CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Novel spiroaminodihydropyrroles probing for optimized interactions at the P3 pocket of β-secretase 1 (BACE1) were designed with the use of free energy perturbation (FEP) calcns. The resulting mols. showed pIC50 potencies in enzymic BACE1 inhibition assays ranging from approx. 5 to 7. Good correlation was obsd. between the predicted activity from the FEP calcns. and exptl. activity. Simulations run with a default 5 ns approach delivered a mean unsigned error (MUE) between prediction and expt. of 0.58 and 0.91 kcal/mol for retrospective and prospective applications, resp. With longer simulations of 10 and 20 ns, the MUE was in both cases 0.57 kcal/mol for the retrospective application, and 0.69 and 0.59 kcal/mol for the prospective application. Other considerations that impact the quality of the calcns. are discussed. This work provides an example of the value of FEP as a computational tool for drug discovery.
- 7Wang, 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/ja512751q7https://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.
- 8Wan, S.; Bhati, A. P.; Zasada, S. J.; Coveney, P. V. Rapid, Accurate, Precise and Reproducible Ligand-Protein Binding Free Energy Prediction. Interface Focus 2020, 10, 20200007 DOI: 10.1098/rsfs.2020.00078https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3s3js1Wiuw%253D%253D&md5=9626db15b14559e08c35bfabc0716e4cRapid, accurate, precise and reproducible ligand-protein binding free energy predictionWan Shunzhou; Bhati Agastya P; Zasada Stefan J; Coveney Peter V; Coveney Peter VInterface focus (2020), 10 (6), 20200007 ISSN:2042-8898.A central quantity of interest in molecular biology and medicine is the free energy of binding of a molecule to a target biomacromolecule. Until recently, the accurate prediction of binding affinity had been widely regarded as out of reach of theoretical methods owing to the lack of reproducibility of the available methods, not to mention their complexity, computational cost and time-consuming procedures. The lack of reproducibility stems primarily from the chaotic nature of classical molecular dynamics (MD) and the associated extreme sensitivity of trajectories to their initial conditions. Here, we review computational approaches for both relative and absolute binding free energy calculations, and illustrate their application to a diverse set of ligands bound to a range of proteins with immediate relevance in a number of medical domains. We focus on ensemble-based methods which are essential in order to compute statistically robust results, including two we have recently developed, namely thermodynamic integration with enhanced sampling and enhanced sampling of MD with an approximation of continuum solvent. Together, these form a set of rapid, accurate, precise and reproducible free energy methods. They can be used in real-world problems such as hit-to-lead and lead optimization stages in drug discovery, and in personalized medicine. These applications show that individual binding affinities equipped with uncertainty quantification may be computed in a few hours on a massive scale given access to suitable high-end computing resources and workflow automation. A high level of accuracy can be achieved using these approaches.
- 9Wan, S.; Tresadern, G.; Pérez-Benito, L.; Vlijmen, H.; Coveney, P. V. Accuracy and Precision of Alchemical Relative Free Energy Predictions with and without Replica-Exchange. Adv. Theory Simul. 2019, 3, 1900195 DOI: 10.1002/adts.201900195There is no corresponding record for this reference.
- 10Wan, 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.5b0017910https://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.
- 11Bhati, 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.6b0097911https://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.
- 12Kollman, 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., 3rd Calculating Structures and Free Energies of Complex Molecules: Combining Molecular Mechanics and Continuum Models. Acc. Chem. Res. 2000, 33, 889– 897, DOI: 10.1021/ar000033j12https://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.
- 13Coveney, 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/C6CP02349E13https://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.
- 14Vassaux, M.; Wan, S.; Edeling, W.; Coveney, P. V. Ensembles Are Required to Handle Aleatoric and Parametric Uncertainty in Molecular Dynamics Simulation. J. Chem. Theory Comput. 2021, 17, 5187– 5197, DOI: 10.1021/acs.jctc.1c0052614https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsFKktbjI&md5=12376b0c3d728bb7b54be27e450867f8Aleatoric and parametric uncertainty in molecular dynamics simulationVassaux, Maxime; Wan, Shunzhou; Edeling, Wouter; Coveney, Peter V.Journal of Chemical Theory and Computation (2021), 17 (8), 5187-5197CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Classical mol. dynamics is a computer simulation technique that is in widespread use across many areas of science, from physics and chem. to materials, biol. and medicine. The method continues to attract criticism due its oft-reported lack of reproducibility which is in part due to a failure to submit it to reliable uncertainty quantification (UQ). Here we show that the uncertainty arises from a combination of (i) the input parameters and (ii) the intrinsic stochasticity of the method controlled by the random seeds. To illustrate the situation, we make a systematic UQ anal. of a widely used mol. dynamics code (NAMD), applied to est. binding free energy of a ligand-bound to a protein. In particular, we replace the usually fixed input parameters with random variables, systematically distributed about their mean values, and study the resulting distribution of the simulation output. We also perform a sensitivity anal., which reveals that, out of a total of 175 parameters, just six dominate the variance in the code output. Furthermore, we show that binding energy calcn. damps the input uncertainty, in the sense that the variation around the mean output free energy is less than the variation around the mean of the assumed input distributions, if the output is ensemble-averaged over the random seeds. Without such ensemble averaging, the predicted free energy is five times more uncertain. The distribution of the predicted properties is thus strongly dependent upon the random seed. Owing to this substantial uncertainty, robust statistical measures of uncertainty in mol. dynamics simulation require the use of ensembles in all contexts.
- 15Wade, A.; Bhati, A. P.; Wan, S.; Coveney, P. V. Alchemical Free Energy Estimators and Molecular Dynamics Engines: Accuracy Precision and Reproducibility. ChemRxiv 2021, DOI: 10.26434/chemrxiv-2021-nqp8r .There is no corresponding record for this reference.
- 16Bhati, A. P.; Coveney, P. V. Large Scale Study of Ligand-Protein Relative Binding Free Energy Calculations: Actionable Predictions from Statistically Robust Protocols. J. Chem. Theory Comput. 2022, 18, 2687– 2702, DOI: 10.1021/acs.jctc.1c0128816https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XntVaqtL8%253D&md5=2b4f884275deced8f66f6b4a6b067b36Large Scale Study of Ligand-Protein Relative Binding Free Energy Calculations: Actionable Predictions from Statistically Robust ProtocolsBhati, Agastya P.; Coveney, Peter V.Journal of Chemical Theory and Computation (2022), 18 (4), 2687-2702CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The accurate and reliable prediction of protein-ligand binding affinities can play a central role in the drug discovery process as well as in personalized medicine. Of considerable importance during lead optimization are the alchem. free energy methods that furnish an estn. of relative binding free energies (RBFE) of similar mols. Recent advances in these methods have increased their speed, accuracy, and precision. This is evident from the increasing no. of retrospective as well as prospective studies employing them. However, such methods still have limited applicability in real-world scenarios due to a no. of important yet unresolved issues. Here, we report the findings from a large data set comprising over 500 ligand transformations spanning over 300 ligands binding to a diverse set of 14 different protein targets which furnish statistically robust results on the accuracy, precision, and reproducibility of RBFE calcns. We use ensemble-based methods which are the only way to provide reliable uncertainty quantification given that the underlying mol. dynamics is chaotic. These are implemented using TIES (Thermodn. Integration with Enhanced Sampling). Results achieve chem. accuracy in all cases. Ensemble simulations also furnish information on the statistical distributions of the free energy calcns. which exhibit non-normal behavior. We find that the "enhanced sampling" method known as replica exchange with solute tempering degrades RBFE predictions. We also report definitively on numerous assocd. alchem. factors including the choice of ligand charge method, flexibility in ligand structure, and the size of the alchem. region including the no. of atoms involved in transforming one ligand into another. Our findings provide a key set of recommendations that should be adopted for the reliable application of RBFE methods.
- 17Knapp, B.; Ospina, L.; Deane, C. M. Avoiding False Positive Conclusions in Molecular Simulation: The Importance of Replicas. J. Chem. Theory Comput. 2018, 14, 6127– 6138, DOI: 10.1021/acs.jctc.8b0039117https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFygsb%252FN&md5=6ed83fe1ea4a74d6739c2cc521fcfc70Avoiding False Positive Conclusions in Molecular Simulation: The Importance of ReplicasKnapp, Bernhard; Ospina, Luis; Deane, Charlotte M.Journal of Chemical Theory and Computation (2018), 14 (12), 6127-6138CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Mol. simulations are a computational technique used to investigate the dynamics of proteins and other mols. The free energy landscape of these simulations is often rugged, and minor differences in the initial velocities, floating-point precision, or underlying hardware can cause identical simulations (replicas) to take different paths in the landscape. In this study the authors investigated the magnitude of these effects based on 310 000 ns of simulation time. The authors performed 100 identically parametrized replicas of 3000 ns each for a small 10 amino acid system as well as 100 identically parametrized replicas of 100 ns each for an 827 residue T-cell receptor/MHC system. Comparing randomly chosen subgroups within these replica sets, the authors estd. the reproducibility and reliability that can be achieved by a given no. of replicas at a given simulation time. These results demonstrate that conclusions drawn from single simulations are often not reproducible and that conclusions drawn from multiple shorter replicas are more reliable than those from a single longer simulation. The actual no. of replicas needed will always depend on the question asked and the level of reliability sought. On the basis of the data, it appears that a good rule of thumb is to perform a min. of five to 10 replicas.
- 18Wan, S.; Sinclair, R. C.; Coveney, P. V. Uncertainty Quantification in Classical Molecular Dynamics. Philos. Trans. R. Soc., A 2021, 379, 20200082 DOI: 10.1098/rsta.2020.008218https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXptFentrY%253D&md5=d2b00cf950ff1e1b69ab4d468dfa5cfeUncertainty quantification in classical molecular dynamicsWan, Shunzhou; Sinclair, Robert C.; Coveney, Peter V.Philosophical Transactions of the Royal Society, A: Mathematical, Physical & Engineering Sciences (2021), 379 (2197), 20200082CODEN: PTRMAD; ISSN:1364-503X. (Royal Society)Mol. dynamics simulation is now a widespread approach for understanding complex systems on the atomistic scale. It finds applications from physics and chem. to engineering, life and medical science. In the last decade, the approach has begun to advance from being a computer-based means of rationalizing exptl. observations to producing apparently credible predictions for a no. of real-world applications within industrial sectors such as advanced materials and drug discovery. However, key aspects concerning the reproducibility of the method have not kept pace with the speed of its uptake in the scientific community. Here, we present a discussion of uncertainty quantification for mol. dynamics simulation designed to endow the method with better error ests. that will enable it to be used to report actionable results. The approach adopted is a std. one in the field of uncertainty quantification, namely using ensemble methods, in which a sufficiently large no. of replicas are run concurrently, from which reliable statistics can be extd. Indeed, because mol. dynamics is intrinsically chaotic, the need to use ensemble methods is fundamental and holds regardless of the duration of the simulations performed. We discuss the approach and illustrate it in a range of applications from materials science to ligand-protein binding free energy estn.
- 19Bieniek, M. K.; Bhati, A. P.; Wan, S.; Coveney, P. V. TIES 20: Relative Binding Free Energy with a Flexible Superimposition Algorithm and Partial Ring Morphing. J. Chem. Theory Comput. 2021, 17, 1250– 1265, DOI: 10.1021/acs.jctc.0c0117919https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVWks7w%253D&md5=fdeea48a835674ad89813380561ad2bfTIES 20: Relative Binding Free Energy with a Flexible Superimposition Algorithm and Partial Ring MorphingBieniek, Matuesz K.; Bhati, Agastya P.; Wan, Shunzhou; Coveney, Peter V.Journal of Chemical Theory and Computation (2021), 17 (2), 1250-1265CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The TIES (Thermodn. Integration with Enhanced Sampling) protocol is a formally exact alchem. approach in computational chem. to the calcn. of relative binding free energies. The validity of TIES relies on the correctness of matching atoms across compared pairs of ligands, laying the foundation for the transformation along an alchem. pathway. We implement a flexible topol. superimposition algorithm which uses an exhaustive joint-traversal for computing the largest common component(s). The algorithm is employed to enable matching and morphing of partial rings in the TIES protocol along with a validation study using 55 transformations and five different proteins from our previous work. We find that TIES 20 with the RESP charge system, using the new superimposition algorithm, reproduces the previous results with mean unsigned error of 0.75 kcal/mol with respect to the exptl. data. Enabling the morphing of partial rings decreases the size of the alchem. region in the dual-topol. transformations resulting in a significant improvement in the prediction precision. We find that increasing the ensemble size from 5 to 20 replicas per λ window only has a minimal impact on the accuracy. However, the non-normal nature of the relative free energy distributions underscores the importance of ensemble simulation. We further compare the results with the AM1-BCC charge system and show that it improves agreement with the exptl. data by slightly over 10%. This improvement is partly due to AM1-BCC affecting only the charges of the atoms local to the mutation, which translates to even fewer morphed atoms, consequently reducing issues with sampling and therefore ensemble averaging. TIES 20, in conjunction with the enablement of ring morphing, reduces the size of the alchem. region and significantly improves the precision of the predicted free energies.
- 20Sadiq, 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/ci800093720https://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.
- 21Suruzhon, M.; Bodnarchuk, M. S.; Ciancetta, A.; Viner, R.; Wall, I. D.; Essex, J. W. Sensitivity of Binding Free Energy Calculations to Initial Protein Crystal Structure. J. Chem. Theory Comput. 2021, 17, 1806– 1821, DOI: 10.1021/acs.jctc.0c0097221https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXislGjsr8%253D&md5=7e1754a44e1387726c524896f071247fSensitivity of binding free energy calculations to initial protein crystal structureSuruzhon, Miroslav; Bodnarchuk, Michael S.; Ciancetta, Antonella; Viner, Russell; Wall, Ian D.; Essex, Jonathan W.Journal of Chemical Theory and Computation (2021), 17 (3), 1806-1821CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Binding free energy calcns. using alchem. free energy (AFE) methods are widely considered to be the most rigorous tool in the computational drug discovery arsenal. Despite this, the calcns. suffer from accuracy, precision, and reproducibility issues. In this publication, the authors perform a high-throughput study of more than a thousand AFE calcns., utilizing over 220μs of total sampling time, on three different protein systems to investigate the impact of the initial crystal structure on the resulting binding free energy values. The authors also consider the influence of equilibration time and discover that the initial crystal structure can have a significant effect on free energy values obtained at short timescales that can manifest itself as a free energy difference of more than 1 kcal/mol. At longer timescales, these differences are largely overtaken by important rare events, such as torsional ligand motions, typically resulting in a much higher uncertainty in the obtained values. This work emphasizes the importance of rare event sampling and long-timescale dynamics in free energy calcns. even for routinely performed alchem. perturbations. The authors conclude that an optimal protocol should not only conc. computational resources on achieving convergence in the alchem. coupling parameter (λ) space but also on longer simulations and multiple repeats.
- 22Pérez-Benito, L.; Keränen, H.; van Vlijmen, H.; Tresadern, G. Predicting Binding Free Energies of PDE2 Inhibitors. The Difficulties of Protein Conformation. Sci. Rep. 2018, 8, 4883, DOI: 10.1038/s41598-018-23039-522https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MnjtFKlsA%253D%253D&md5=44c26c5bd2fe3e35bba344abb8262eebPredicting Binding Free Energies of PDE2 Inhibitors. The Difficulties of Protein ConformationPerez-Benito Laura; Keranen Henrik; van Vlijmen Herman; Tresadern Gary; Keranen HenrikScientific reports (2018), 8 (1), 4883 ISSN:.A congeneric series of 21 phosphodiesterase 2 (PDE2) inhibitors are reported. Crystal structures show how the molecules can occupy a 'top-pocket' of the active site. Molecules with small substituents do not enter the pocket, a critical leucine (Leu770) is closed and water molecules are present. Large substituents enter the pocket, opening the Leu770 conformation and displacing the waters. We also report an X-ray structure revealing a new conformation of the PDE2 active site domain. The relative binding affinities of these compounds were studied with free energy perturbation (FEP) methods and it represents an attractive real-world test case. In general, the calculations could predict the energy of small-to-small, or large-to-large molecule perturbations. However, accurately capturing the transition from small-to-large proved challenging. Only when using alternative protein conformations did results improve. The new X-ray structure, along with a modelled dimer, conferred stability to the catalytic domain during the FEP molecular dynamics (MD) simulations, increasing the convergence and thereby improving the prediction of ΔΔG of binding for some small-to-large transitions. In summary, we found the most significant improvement in results when using different protein structures, and this data set is useful for future free energy validation studies.
- 23Friesner, R. A.; Murphy, R. B.; Repasky, M. P.; Frye, L. L.; Greenwood, J. R.; Halgren, T. A.; Sanschagrin, P. C.; Mainz, D. T. Extra Precision Glide: Docking and Scoring Incorporating a Model of Hydrophobic Enclosure for Protein-Ligand Complexes. J. Med. Chem. 2006, 49, 6177– 6196, DOI: 10.1021/jm051256o23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XpvVGmurg%253D&md5=ea428c82ead0d8c27f8c1a7b694a1edfExtra Precision Glide: Docking and Scoring Incorporating a Model of Hydrophobic Enclosure for Protein-Ligand ComplexesFriesner, Richard A.; Murphy, Robert B.; Repasky, Matthew P.; Frye, Leah L.; Greenwood, Jeremy R.; Halgren, Thomas A.; Sanschagrin, Paul C.; Mainz, Daniel T.Journal of Medicinal Chemistry (2006), 49 (21), 6177-6196CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A novel scoring function to est. protein-ligand binding affinities has been developed and implemented as the Glide 4.0 XP scoring function and docking protocol. In addn. to unique water desolvation energy terms, protein-ligand structural motifs leading to enhanced binding affinity are included:(1) hydrophobic enclosure where groups of lipophilic ligand atoms are enclosed on opposite faces by lipophilic protein atoms, (2) neutral-neutral single or correlated hydrogen bonds in a hydrophobically enclosed environment, and (3) five categories of charged-charged hydrogen bonds. The XP scoring function and docking protocol have been developed to reproduce exptl. binding affinities for a set of 198 complexes (RMSDs of 2.26 and 1.73 kcal/mol over all and well-docked ligands, resp.) and to yield quality enrichments for a set of fifteen screens of pharmaceutical importance. Enrichment results demonstrate the importance of the novel XP mol. recognition and water scoring in sepg. active and inactive ligands and avoiding false positives.
- 24Case, D. A.; Cheatham, T. E., 3rd; Darden, T.; Gohlke, H.; Luo, R.; Merz, K. M., Jr.; Onufriev, A.; Simmerling, C.; Wang, B.; Woods, R. J. The Amber Biomolecular Simulation Programs. J. Comput. Chem. 2005, 26, 1668– 1688, DOI: 10.1002/jcc.2029024https://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.
- 25Zhang, X.; Perez-Sanchez, H.; Lightstone, F. C. A Comprehensive Docking and MM/GBSA Rescoring Study of Ligand Recognition Upon Binding Antithrombin. Curr. Top. Med. Chem. 2017, 17, 1631– 1639, DOI: 10.2174/156802661666616111711260425https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXmt1eht7s%253D&md5=c1baa4455076543f619a194bc5559bffA Comprehensive Docking and MM/GBSA Rescoring Study of Ligand Recognition upon Binding AntithrombinZhang, Xiaohua; Perez-Sanchez, Horacio; Lightstone, Felice C.Current Topics in Medicinal Chemistry (Sharjah, United Arab Emirates) (2017), 17 (14), 1631-1639CODEN: CTMCCL; ISSN:1568-0266. (Bentham Science Publishers Ltd.)Background: A high-throughput virtual screening pipeline has been extended from single energetically minimized structure Mol. Mechanics/Generalized Born Surface Area (MM/GBSA) rescoring to ensemble-av. MM/GBSA rescoring. The correlation coeff. (R2) of calcd. and exptl. binding free energies for a series of antithrombin ligands has been improved from 0.36 to 0.69 when switching from the single-structure MM/GBSA rescoring to ensemble-av. one. The electrostatic interactions in both solute and solvent are identified to play an important role in detg. the binding free energy after the decompn. of the calcd. binding free energy. The increasing neg. charge of the compds. provides a more favorable electrostatic energy change but creates a higher penalty for the solvation free energy. Such a penalty is compensated by the electrostatic energy change, which results in a better binding affinity. A highly hydrophobic ligand is detd. by the docking program to be a non-specific binder. Results: Our results have demonstrated that it is very important to keep a few top poses for rescoring, if the binding is non-specific or the binding mode is not well detd. by the docking calcn.
- 26Wright, D. W.; Husseini, F.; Wan, S.; Meyer, C.; van Vlijmen, H.; Tresadern, G.; Coveney, P. V. Application of the ESMACS Binding Free Energy Protocol to a Multi-Binding Site Lactate Dehydogenase a Ligand Dataset. Adv. Theory Simul. 2019, 3, 1900194 DOI: 10.1002/adts.201900194There is no corresponding record for this reference.
- 27Wright, D. W.; Wan, S.; Meyer, C.; van Vlijmen, H.; Tresadern, G.; Coveney, P. V. Application of ESMACS Binding Free Energy Protocols to Diverse Datasets: Bromodomain-Containing Protein 4. Sci. Rep. 2019, 9, 6017, DOI: 10.1038/s41598-019-41758-127https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M%252FkvFaqsQ%253D%253D&md5=b62c5a234b09797af7f8e21846c7815fApplication of ESMACS binding free energy protocols to diverse datasets: Bromodomain-containing protein 4Wright David W; Wan Shunzhou; Coveney Peter V; Meyer Christophe; van Vlijmen Herman; Tresadern GaryScientific reports (2019), 9 (1), 6017 ISSN:.As the application of computational methods in drug discovery pipelines becomes more widespread it is increasingly important to understand how reproducible their results are and how sensitive they are to choices made in simulation setup and analysis. Here we use ensemble simulation protocols, termed ESMACS (enhanced sampling of molecular dynamics with approximation of continuum solvent), to investigate the sensitivity of the popular molecular mechanics Poisson-Boltzmann surface area (MMPBSA) methodology. Using the bromodomain-containing protein 4 (BRD4) system bound to a diverse set of ligands as our target, we show that robust rankings can be produced only through combining ensemble sampling with multiple trajectories and enhanced solvation via an explicit ligand hydration shell.
- 28Wan, S.; Potterton, A.; Husseini, F. S.; Wright, D. W.; Heifetz, A.; Malawski, M.; Townsend-Nicholson, A.; Coveney, P. V. Hit-to-Lead and Lead Optimization Binding Free Energy Calculations for G Protein-Coupled Receptors. Interface Focus 2020, 10, 20190128 DOI: 10.1098/rsfs.2019.012828https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3s3js1Witw%253D%253D&md5=cd117abc40d9d90d9878a35d49729109Hit-to-lead and lead optimization binding free energy calculations for G protein-coupled receptorsWan Shunzhou; Husseini Fouad S; Wright David W; Coveney Peter V; Potterton Andrew; Heifetz Alexander; Townsend-Nicholson Andrea; Heifetz Alexander; Malawski Maciej; Coveney Peter VInterface focus (2020), 10 (6), 20190128 ISSN:2042-8898.We apply the hit-to-lead ESMACS (enhanced sampling of molecular dynamics with approximation of continuum solvent) and lead-optimization TIES (thermodynamic integration with enhanced sampling) methods to compute the binding free energies of a series of ligands at the A1 and A2A adenosine receptors, members of a subclass of the GPCR (G protein-coupled receptor) superfamily. Our predicted binding free energies, calculated using ESMACS, show a good correlation with previously reported experimental values of the ligands studied. Relative binding free energies, calculated using TIES, accurately predict experimentally determined values within a mean absolute error of approximately 1 kcal mol(-1). Our methodology may be applied widely within the GPCR superfamily and to other small molecule-receptor protein systems.
- 29Wright, 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/ct400703729https://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.
- 30Phillips, 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.2028930https://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.
- 31Beveridge, D. L.; Dicapua, F. M. Free-Energy Via Molecular Simulation - Applications to Chemical and Biomolecular Systems. Annu. Rev. Biophys. Bioeng. 1989, 18, 431– 492, DOI: 10.1146/annurev.bb.18.060189.00224331https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXlvVajsLs%253D&md5=396c2dbf6a5c78a579badf764f070c0dFree energy via molecular simulation: applications to chemical and biomolecular systemsBeveridge, D. L.; DiCapua, F. M.Annual Review of Biophysics and Biophysical Chemistry (1989), 18 (), 431-92CODEN: ARBCEY; ISSN:0883-9182.A review with over 250 refs. General approaches to free energy detns. by mol. simulation are formulated theor. and the practical uses are discussed. In general, the agreement between calcd. and exptl. data is good when some exptl. data are used in parametrization of the intermol. potentials. The applications of the methods are also discussed.
- 32Genheden, S.; Ryde, U. How to Obtain Statistically Converged MM/GBSA Results. J. Comput. Chem. 2010, 31, 837– 846, DOI: 10.1002/jcc.2136632https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtlentbY%253D&md5=173f37e54d4f0c80a8ec62df78f95127How to obtain statistically converged MM/GBSA resultsGenheden, Samuel; Ryde, UlfJournal of Computational Chemistry (2010), 31 (4), 837-846CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)The mol. mechanics/generalized Born surface area (MM/GBSA) method has been investigated with the aim of achieving a statistical precision of 1 kJ/mol for the results. The authors studied the binding of seven biotin analogs to avidin, taking advantage of the fact that the protein is a tetramer with four independent binding sites, which should give the same estd. binding affinities. The authors show that it is not enough to use a single long simulation (10 ns), because the std. error of such a calcn. underestimates the difference between the four binding sites. Instead, it is better to run several independent simulations and av. the results. With such an approach, the authors obtain the same results for the four binding sites, and any desired precision can be obtained by running a proper no. of simulations. The authors discuss how the simulations should be performed to optimize the use of computer time. The correlation time between the MM/GBSA energies is ∼5 ps and an equilibration time of 100 ps is needed. For MM/GBSA, the authors recommend a sampling time of 20-200 ps for each sep. simulation, depending on the protein. With 200 ps prodn. time, 5-50 sep. simulations are required to reach a statistical precision of 1 kJ/mol (800-8000 energy calcns. or 1.5-15 ns total simulation time per ligand) for the seven avidin ligands. This is an order of magnitude more than what is normally used, but such a no. of simulations is needed to obtain statistically valid results for the MM/GBSA method. © 2009 Wiley Periodicals, Inc. J Comput Chem 2010.
- 33Sadiq, S. K.; Wright, D. W.; Kenway, O. A.; Coveney, P. V. Accurate Ensemble Molecular Dynamics Binding Free Energy Ranking of Multidrug-Resistant HIV-1 Proteases. J. Chem. Inf. Model. 2010, 50, 890– 905, DOI: 10.1021/ci100007w33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksFeqtb4%253D&md5=4a9c06d974aad014a9a5e027a08a76acAccurate Ensemble Molecular Dynamics Binding Free Energy Ranking of Multidrug-Resistant HIV-1 ProteasesSadiq, S. Kashif; Wright, David W.; Kenway, Owain A.; Coveney, Peter V.Journal of Chemical Information and Modeling (2010), 50 (5), 890-905CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Accurate calcn. of important thermodn. properties, such as macromol. binding free energies, is one of the principal goals of mol. dynamics simulations. However, single long simulation frequently produces incorrectly converged quant. results due to inadequate sampling of conformational space in a feasible wall-clock time. Multiple short (ensemble) simulations have been shown to explore conformational space more effectively than single long simulations, but the two methods have not yet been thermodynamically compared. Here we show that, for end-state binding free energy detn. methods, ensemble simulations exhibit significantly enhanced thermodn. sampling over single long simulations and result in accurate and converged relative binding free energies that are reproducible to within 0.5 kcal/mol. Completely correct ranking is obtained for six HIV-1 protease variants bound to lopinavir with a correlation coeff. of 0.89 and a mean relative deviation from expt. of 0.9 kcal/mol. Multidrug resistance to lopinavir is enthalpically driven and increases through a decrease in the protein-ligand van der Waals interaction, principally due to the V82A/I84V mutation, and an increase in net electrostatic repulsion due to water-mediated disruption of protein-ligand interactions in the catalytic region. Furthermore, we correctly rank, to within 1 kcal/mol of expt., the substantially increased chem. potency of lopinavir binding to the wild-type protease compared to saquinavir and show that lopinavir takes advantage of a decreased net electrostatic repulsion to confer enhanced binding. Our approach is dependent on the combined use of petascale computing resources and on an automated simulation workflow to attain the required level of sampling and turn around time to obtain the results, which can be as little as three days. This level of performance promotes integration of such methodol. with clin. decision support systems for the optimization of patient-specific therapy.
- 34Wan, S.; Bhati, A. P.; Skerratt, S.; Omoto, K.; Shanmugasundaram, V.; Bagal, S. K.; Coveney, P. V. Evaluation and Characterization of Trk Kinase Inhibitors for the Treatment of Pain: Reliable Binding Affinity Predictions from Theory and Computation. J. Chem. Inf. Model. 2017, 57, 897– 909, DOI: 10.1021/acs.jcim.6b0078034https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXksVyrurc%253D&md5=2e366f56c1ee8c2f2b2760b9098e2030Evaluation and Characterization of Trk Kinase Inhibitors for the Treatment of Pain: Reliable Binding Affinity Predictions from Theory and ComputationWan, Shunzhou; Bhati, Agastya P.; Skerratt, Sarah; Omoto, Kiyoyuki; Shanmugasundaram, Veerabahu; Bagal, Sharan K.; Coveney, Peter V.Journal of Chemical Information and Modeling (2017), 57 (4), 897-909CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Optimization of ligand binding affinity to the target protein of interest is a primary objective in small-mol. drug discovery. Until now, the prediction of binding affinities by computational methods has not been widely applied in the drug discovery process, mainly due to its lack of accuracy and reproducibility, as well as the long turnaround times required to obtain results. Herein, the authors report on a collaborative study that compares tropomyosin receptor kinase A (TrkA) binding affinity predictions using two recently formulated fast computational approaches - namely ESMACS (Enhanced Sampling of Mol. dynamics with Approxn. of Continuum Solvent) and TIES (Thermodn. Integration with Enhanced Sampling) - to exptl. derived TrkA binding affinities for a set of Pfizer pan-Trk compds. ESMACS gives precise and reproducible results and is applicable to highly diverse sets of compds. It also provides detailed chem. insight into the nature of ligand-protein binding. TIES can predict and thus optimize more subtle changes in binding affinities between compds. of similar structure. Individual binding affinities were calcd. in a few hours, exhibiting good correlations with the exptl. data of 0.79 and 0.88 from ESMACS and TIES approaches resp. The speed, level of accuracy and precision of the calcns. are such that the affinity predictions can be used to rapidly explain the effects of compd. 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 compds. and targets.
- 35Wan, 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.6b0079435https://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.
- 36Scheen, J.; Wu, W.; Mey, A.; Tosco, P.; Mackey, M.; Michel, J. Hybrid Alchemical Free Energy/Machine-Learning Methodology for the Computation of Hydration Free Energies. J. Chem. Inf. Model. 2020, 60, 5331– 5339, DOI: 10.1021/acs.jcim.0c0060036https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlGnsbvF&md5=5168eabd0e51fb6ddb40f0d782171458Hybrid Alchemical Free Energy/Machine-Learning Methodology for the Computation of Hydration Free EnergiesScheen, Jenke; Wu, Wilson; Mey, Antonia S. J. S.; Tosco, Paolo; Mackey, Mark; Michel, JulienJournal of Chemical Information and Modeling (2020), 60 (11), 5331-5339CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)A methodol. that combines alchem. free energy calcns. (FEP) with machine learning (ML) has been developed to compute accurate abs. hydration free energies. The hybrid FEP/ML methodol. was trained on a subset of the FreeSolv database, and retrospectively shown to outperform most submissions from the SAMPL4 competition. Compared to pure machine-learning approaches, FEP/ML yields more precise ests. of free energies of hydration, and requires a fraction of the training set size to outperform standalone FEP calcns. The ML-derived correction terms are further shown to be transferable to a range of related FEP simulation protocols. The approach may be used to inexpensively improve the accuracy of FEP calcns., and to flag mols. which will benefit the most from bespoke forcefield parameterization efforts.
- 37Frost, C.; Thompson, S. G. Correcting for Regression Dilution Bias: Comparison of Methods for a Single Predictor Variable. J. R. Stat. Soc. 2000, 163, 173– 189, DOI: 10.1111/1467-985X.00164There is no corresponding record for this reference.
- 38Ruff, T. W.; Neelin, J. D. Long Tails in Regional Surface Temperature Probability Distributions with Implications for Extremes under Global Warming. Geophys. Res. Lett. 2012, 39, L04704, DOI: 10.1029/2011GL050610There is no corresponding record for this reference.
- 39Weitzman, M. L. Fat-Tailed Uncertainty in the Economics of Catastrophic Climate Change. Rev. Environ. Econ. Policy 2011, 5, 275– 292, DOI: 10.1093/reep/rer006There is no corresponding record for this reference.