Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lingle Wang is active.

Publication


Featured researches published by Lingle Wang.


Journal of Chemical Theory and Computation | 2016

OPLS3: A Force Field Providing Broad Coverage of Drug-like Small Molecules and Proteins

Edward Harder; Wolfgang Damm; Jon R. Maple; Chuanjie Wu; Mark Reboul; Jin Yu Xiang; Lingle Wang; Dmitry Lupyan; Markus K. Dahlgren; Jennifer L. Knight; Joseph W. Kaus; David S. Cerutti; Goran Krilov; William L. Jorgensen; Robert Abel

The parametrization and validation of the OPLS3 force field for small molecules and proteins are reported. Enhancements with respect to the previous version (OPLS2.1) include the addition of off-atom charge sites to represent halogen bonding and aryl nitrogen lone pairs as well as a complete refit of peptide dihedral parameters to better model the native structure of proteins. To adequately cover medicinal chemical space, OPLS3 employs over an order of magnitude more reference data and associated parameter types relative to other commonly used small molecule force fields (e.g., MMFF and OPLS_2005). As a consequence, OPLS3 achieves a high level of accuracy across performance benchmarks that assess small molecule conformational propensities and solvation. The newly fitted peptide dihedrals lead to significant improvements in the representation of secondary structure elements in simulated peptides and native structure stability over a number of proteins. Together, the improvements made to both the small molecule and protein force field lead to a high level of accuracy in predicting protein-ligand binding measured over a wide range of targets and ligands (less than 1 kcal/mol RMS error) representing a 30% improvement over earlier variants of the OPLS force field.


Journal of the American Chemical Society | 2015

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

Lingle Wang; Yujie Wu; Yuqing Deng; Byungchan Kim; Levi C. T. Pierce; Goran Krilov; Dmitry Lupyan; Shaughnessy Robinson; Markus K. Dahlgren; Jeremy R. Greenwood; Donna L. Romero; Craig E. Masse; Jennifer L. Knight; Thomas Steinbrecher; Thijs Beuming; Wolfgang Damm; Ed Harder; Woody Sherman; Mark L. Brewer; Ron Wester; Mark A. Murcko; Leah L. Frye; Ramy Farid; Teng-Yi Lin; David L. Mobley; William L. Jorgensen; B. J. Berne; Robert Abel

Designing tight-binding ligands is a primary objective of small-molecule drug discovery. Over the past few decades, free-energy calculations 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 commercial application of free-energy simulations has been limited due to the lack of large-scale validation coupled with the technical challenges traditionally associated with running these types of calculations. 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 chemical perturbations, many of which involve significant changes in ligand chemical structures. In addition, we have applied the method in prospective drug discovery projects and found a significant improvement in the quality of the compounds synthesized that have been predicted to be potent. Compounds predicted to be potent by this approach have a substantial reduction in false positives relative to compounds synthesized on the basis of other computational or medicinal chemistry 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.


Journal of Physical Chemistry B | 2011

Replica Exchange with Solute Scaling: A more efficient version of Replica Exchange with Solute Tempering (REST2)

Lingle Wang; B. J. Berne

A small change in the Hamiltonian scaling in Replica Exchange with Solute Tempering (REST) is found to improve its sampling efficiency greatly, especially for the sampling of aqueous protein solutions in which there are large-scale solute conformation changes. Like the original REST (REST1), the new version (which we call REST2) also bypasses the poor scaling with system size of the standard Temperature Replica Exchange Method (TREM), reducing the number of replicas (parallel processes) from what must be used in TREM. This reduction is accomplished by deforming the Hamiltonian function for each replica in such a way that the acceptance probability for the exchange of replica configurations does not depend on the number of explicit water molecules in the system. For proof of concept, REST2 is compared with TREM and with REST1 for the folding of the trpcage and β-hairpin in water. The comparisons confirm that REST2 greatly reduces the number of CPUs required by regular replica exchange and greatly increases the sampling efficiency over REST1. This method reduces the CPU time required for calculating thermodynamic averages and for the ab initio folding of proteins in explicit water.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Ligand binding to protein-binding pockets with wet and dry regions

Lingle Wang; B. J. Berne

Biological processes often depend on protein–ligand binding events, yet accurate calculation of the associated energetics remains as a significant challenge of central importance to structure-based drug design. Recently, we have proposed that the displacement of unfavorable waters by the ligand, replacing them with groups complementary to the protein surface, is the principal driving force for protein–ligand binding, and we have introduced the WaterMap method to account this effect. However, in spite of the adage “nature abhors vacuum,” one can occasionally observe situations in which a portion of the receptor active site is so unfavorable for water molecules that a void is formed there. In this paper, we demonstrate that the presence of dry regions in the receptor has a nontrivial effect on ligand binding affinity, and suggest that such regions may represent a general motif for molecular recognition between the dry region in the receptor and the hydrophobic groups in the ligands. With the introduction of a term attributable to the occupation of the dry regions by ligand atoms, combined with the WaterMap calculation, we obtain excellent agreement with experiment for the prediction of relative binding affinities for a number of congeneric ligand series binding to the major urinary protein receptor. In addition, WaterMap when combined with the cavity contribution is more predictive than at least one specific implementation [Abel R, Young T, Farid R, Berne BJ, Friesner RA (2008) J Am Chem Soc 130:2817–2831] of the popular MM-GBSA approach to binding affinity calculation.


Proceedings of the National Academy of Sciences of the United States of America | 2012

On achieving high accuracy and reliability in the calculation of relative protein-ligand binding affinities.

Lingle Wang; B. J. Berne

We apply a free energy perturbation simulation method, free energy perturbation/replica exchange with solute tempering, to two modifications of protein–ligand complexes that lead to significant conformational changes, the first in the protein and the second in the ligand. The approach is shown to facilitate sampling in these challenging cases where high free energy barriers separate the initial and final conformations and leads to superior convergence of the free energy as demonstrated both by consistency of the results (independence from the starting conformation) and agreement with experimental binding affinity data. The second case, consisting of two neutral thrombin ligands that are taken from a recent medicinal chemistry program for this interesting pharmaceutical target, is of particular significance in that it demonstrates that good results can be obtained for large, complex ligands, as opposed to relatively simple model systems. To achieve quantitative agreement with experiment in the thrombin case, a next generation force field, Optimized Potentials for Liquid Simulations 2.0, is required, which provides superior charges and torsional parameters as compared to earlier alternatives.


Journal of Chemical Theory and Computation | 2013

Modeling Local Structural Rearrangements Using FEP/REST: Application to Relative Binding Affinity Predictions of CDK2 Inhibitors.

Lingle Wang; Yuqing Deng; Jennifer L. Knight; Yujie Wu; Byungchan Kim; Woody Sherman; John C. Shelley; Teng Lin; Robert Abel

Accurate and reliable calculation of protein-ligand binding affinities remains a hotbed of computer-aided drug design research. Despite the potentially large impact FEP (free energy perturbation) may have in drug design projects, practical applications of FEP in industrial contexts have been limited. In this work, we use a recently developed method, FEP/REST (free energy perturbation/replica exchange with solute tempering), to calculate the relative binding affinities for a set of congeneric ligands binding to the CDK2 receptor. We compare the FEP/REST results with traditional FEP/MD (molecular dynamics) results and MM/GBSA (molecular mechanics/Generalized Born Surface Area model) results and examine why FEP/REST performed notably better than these other methods, as well as why certain ligand mutations lead to large increases of the binding affinity while others do not. We also introduce a mathematical framework for assessing the consistency and reliability of the calculations using cycle closures in FEP mutation paths.


Journal of Chemical Information and Modeling | 2015

Accurate Binding Free Energy Predictions in Fragment Optimization.

Thomas Steinbrecher; Markus K. Dahlgren; Daniel Cappel; Teng Lin; Lingle Wang; Goran Krilov; Robert Abel; Woody Sherman

Predicting protein-ligand binding free energies is a central aim of computational structure-based drug design (SBDD)--improved accuracy in binding free energy predictions could significantly reduce costs and accelerate project timelines in lead discovery and optimization. The recent development and validation of advanced free energy calculation methods represents a major step toward this goal. Accurately predicting the relative binding free energy changes of modifications to ligands is especially valuable in the field of fragment-based drug design, since fragment screens tend to deliver initial hits of low binding affinity that require multiple rounds of synthesis to gain the requisite potency for a project. In this study, we show that a free energy perturbation protocol, FEP+, which was previously validated on drug-like lead compounds, is suitable for the calculation of relative binding strengths of fragment-sized compounds as well. We study several pharmaceutically relevant targets with a total of more than 90 fragments and find that the FEP+ methodology, which uses explicit solvent molecular dynamics and physics-based scoring with no parameters adjusted, can accurately predict relative fragment binding affinities. The calculations afford R(2)-values on average greater than 0.5 compared to experimental data and RMS errors of ca. 1.1 kcal/mol overall, demonstrating significant improvements over the docking and MM-GBSA methods tested in this work and indicating that FEP+ has the requisite predictive power to impact fragment-based affinity optimization projects.


ACS Omega | 2016

Predicting Binding Affinities for GPCR Ligands Using Free-Energy Perturbation

Eelke B. Lenselink; Julien Louvel; Anna F. Forti; Jacobus P. D. van Veldhoven; Henk de Vries; Thea Mulder-Krieger; Fiona M. McRobb; Ana Negri; Joseph Goose; Robert Abel; Herman W. T. van Vlijmen; Lingle Wang; Edward Harder; Woody Sherman; Adriaan P. IJzerman; Thijs Beuming

The rapid growth of structural information for G-protein-coupled receptors (GPCRs) has led to a greater understanding of their structure, function, selectivity, and ligand binding. Although novel ligands have been identified using methods such as virtual screening, computationally driven lead optimization has been possible only in isolated cases because of challenges associated with predicting binding free energies for related compounds. Here, we provide a systematic characterization of the performance of free-energy perturbation (FEP) calculations to predict relative binding free energies of congeneric ligands binding to GPCR targets using a consistent protocol and no adjustable parameters. Using the FEP+ package, first we validated the protocol, which includes a full lipid bilayer and explicit solvent, by predicting the binding affinity for a total of 45 different ligands across four different GPCRs (adenosine A2AAR, β1 adrenergic, CXCR4 chemokine, and δ opioid receptors). Comparison with experimental binding affinity measurements revealed a highly predictive ranking correlation (average spearman ρ = 0.55) and low root-mean-square error (0.80 kcal/mol). Next, we applied FEP+ in a prospective project, where we predicted the affinity of novel, potent adenosine A2A receptor (A2AR) antagonists. Four novel compounds were synthesized and tested in a radioligand displacement assay, yielding affinity values in the nanomolar range. The affinity of two out of the four novel ligands (plus three previously reported compounds) was correctly predicted (within 1 kcal/mol), including one compound with approximately a tenfold increase in affinity compared to the starting compound. Detailed analyses of the simulations underlying the predictions provided insights into the structural basis for the two cases where the affinity was overpredicted. Taken together, these results establish a protocol for systematically applying FEP+ to GPCRs and provide guidelines for identifying potent molecules in drug discovery lead optimization projects.


Journal of Molecular Biology | 2017

Free Energy Perturbation Calculation of Relative Binding Free Energy between Broadly Neutralizing Antibodies and the gp120 Glycoprotein of HIV-1

Anthony J. Clark; Tatyana Gindin; Baoshan Zhang; Lingle Wang; Robert Abel; Colleen S. Murret; Fang Xu; Amy Bao; Nina J. Lu; Tongqing Zhou; Peter D. Kwong; Lawrence Shapiro; Barry Honig

Direct calculation of relative binding affinities between antibodies and antigens is a long-sought goal. However, despite substantial efforts, no generally applicable computational method has been described. Here, we describe a systematic free energy perturbation (FEP) protocol and calculate the binding affinities between the gp120 envelope glycoprotein of HIV-1 and three broadly neutralizing antibodies (bNAbs) of the VRC01 class. The protocol has been adapted from successful studies of small molecules to address the challenges associated with modeling protein–protein interactions. Specifically, we built homology models of the three antibody–gp120 complexes, extended the sampling times for large bulky residues, incorporated the modeling of glycans on the surface of gp120, and utilized continuum solvent-based loop prediction protocols to improve sampling. We present three experimental surface plasmon resonance data sets, in which antibody residues in the antibody/gp120 interface were systematically mutated to alanine. The RMS error in the large set (55 total cases) of FEP tests as compared to these experiments, 0.68 kcal/mol, is near experimental accuracy, and it compares favorably with the results obtained from a simpler, empirical methodology. The correlation coefficient for the combined data set including residues with glycan contacts, R2 = 0.49, should be sufficient to guide the choice of residues for antibody optimization projects, assuming that this level of accuracy can be realized in prospective prediction. More generally, these results are encouraging with regard to the possibility of using an FEP approach to calculate the magnitude of protein–protein binding affinities.


Journal of Medicinal Chemistry | 2017

Prospective Evaluation of Free Energy Calculations for the Prioritization of Cathepsin L Inhibitors

Bernd Kuhn; Michal Tichý; Lingle Wang; Shaughnessy Robinson; Rainer E. Martin; Andreas Kuglstatter; Jörg Benz; Maude Giroud; Tanja Schirmeister; Robert Abel; François Diederich; Jérôme Hert

Improving the binding affinity of a chemical series by systematically probing one of its exit vectors is a medicinal chemistry activity that can benefit from molecular modeling input. Herein, we compare the effectiveness of four approaches in prioritizing building blocks with better potency: selection by a medicinal chemist, manual modeling, docking followed by manual filtering, and free energy calculations (FEP). Our study focused on identifying novel substituents for the apolar S2 pocket of cathepsin L and was conducted entirely in a prospective manner with synthesis and activity determination of 36 novel compounds. We found that FEP selected compounds with improved affinity for 8 out of 10 picks compared to 1 out of 10 for the other approaches. From this result and other additional analyses, we conclude that FEP can be a useful approach to guide this type of medicinal chemistry optimization once it has been validated for the system under consideration.

Collaboration


Dive into the Lingle Wang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge