Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
J Mol Biol ; : 168640, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38844044

ABSTRACT

Computational free energy-based methods have the potential to significantly improve throughput and decrease costs of protein design efforts. Such methods must reach a high level of reliability, accuracy, and automation to be effectively deployed in practical industrial settings in a way that impacts protein design projects. Here, we present a benchmark study for the calculation of relative changes in protein-protein binding affinity for single point mutations across a variety of systems from the literature, using free energy perturbation (FEP+) calculations. We describe a method for robust treatment of alternate protonation states for titratable amino acids, which yields improved correlation with and reduced error compared to experimental binding free energies. Following careful analysis of the largest outlier cases in our dataset, we assess limitations of the default FEP+ protocols and introduce an automated script which identifies probable outlier cases that may require additional scrutiny and calculates an empirical correction for a subset of charge-related outliers. Through a series of three additional case study systems, we discuss how protein FEP+ can be applied to real-world protein design projects, and suggest areas of further study.

2.
bioRxiv ; 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38712280

ABSTRACT

Computational free energy-based methods have the potential to significantly improve throughput and decrease costs of protein design efforts. Such methods must reach a high level of reliability, accuracy, and automation to be effectively deployed in practical industrial settings in a way that impacts protein design projects. Here, we present a benchmark study for the calculation of relative changes in protein-protein binding affinity for single point mutations across a variety of systems from the literature, using free energy perturbation (FEP+) calculations. We describe a method for robust treatment of alternate protonation states for titratable amino acids, which yields improved correlation with and reduced error compared to experimental binding free energies. Following careful analysis of the largest outlier cases in our dataset, we assess limitations of the default FEP+ protocols and introduce an automated script which identifies probable outlier cases that may require additional scrutiny and calculates an empirical correction for a subset of charge-related outliers. Through a series of three additional case study systems, we discuss how protein FEP+ can be applied to real-world protein design projects, and suggest areas of further study.

3.
Cell ; 187(3): 521-525, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38306979

ABSTRACT

High-quality predicted structures enable structure-based approaches to an expanding number of drug discovery programs. We propose that by utilizing free energy perturbation (FEP), predicted structures can be confidently employed to achieve drug design goals. We use structure-based modeling of hERG inhibition to illustrate this value of FEP.


Subject(s)
Drug Design , Drug Discovery , Thermodynamics , Entropy
4.
J Chem Theory Comput ; 19(22): 8414-8422, 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-37943175

ABSTRACT

For an effective drug, strong binding to the target protein is a prerequisite, but it is not enough. To produce a particular functional response, drugs need to either block the proteins' functions or modulate their activities by changing their conformational equilibrium. The binding free energy of a compound to its target is routinely calculated, but the timescales for the protein conformational changes are prohibitively long to be efficiently modeled via physics-based simulations. Thermodynamic principles suggest that the binding free energies of the ligands with different receptor conformations may infer their efficacy. However, this hypothesis has not been thoroughly validated. We present an actionable protocol and a comprehensive study to show that binding thermodynamics provides a strong predictor of the efficacy of a ligand. We apply the absolute binding free energy perturbation method to ligands bound to active and inactive states of eight G protein-coupled receptors and a nuclear receptor and then compare the resulting binding free energies. We find that carefully designed restraints are often necessary to efficiently model the corresponding conformational ensembles for each state. Our method achieves unprecedented performance in classifying ligands as agonists or antagonists across the various investigated receptors, all of which are important drug targets.


Subject(s)
Receptors, G-Protein-Coupled , Protein Conformation , Ligands , Receptors, G-Protein-Coupled/metabolism , Thermodynamics , Protein Binding
5.
J Med Chem ; 66(23): 15883-15893, 2023 12 14.
Article in English | MEDLINE | ID: mdl-38016916

ABSTRACT

Early assessment of crystalline thermodynamic solubility continues to be elusive for drug discovery and development despite its critical importance, especially for the ever-increasing fraction of poorly soluble drug candidates. Here we present a detailed evaluation of a physics-based free energy perturbation (FEP+) approach for computing the thermodynamic aqueous solubility. The predictive power of this approach is assessed across diverse chemical spaces, spanning pharmaceutically relevant literature compounds and more complex AbbVie compounds. Our approach achieves predictive (RMSE = 0.86) and differentiating power (R2 = 0.69) and therefore provides notably improved correlations to experimental solubility compared to state-of-the-art machine learning approaches that utilize quantum mechanics-based descriptors. The importance of explicit considerations of crystalline packing in predicting solubility by the FEP+ approach is also highlighted in this study. Finally, we show how computed energetics, including hydration and sublimation free energies, can provide further insights into molecule design to feed the medicinal chemistry DMTA cycle.


Subject(s)
Drug Discovery , Water , Solubility , Entropy , Thermodynamics , Water/chemistry
6.
Proc Natl Acad Sci U S A ; 120(41): e2304089120, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37792512

ABSTRACT

The serotonin transporter (SERT) tightly regulates synaptic serotonin levels and has been the primary target of antidepressants. Binding of inhibitors to the allosteric site of human SERT (hSERT) impedes the dissociation of antidepressants bound at the central site and may enhance the efficacy of such antidepressants to potentially reduce their dosage and side effects. Here, we report the identification of a series of high-affinity allosteric inhibitors of hSERT in a unique scaffold, with the lead compound, Lu AF88273 (3-(1-(2-(1H-indol-3-yl)ethyl)piperidin-4-yl)-6-chloro-1H-indole), having 2.1 nM allosteric potency in inhibiting imipramine dissociation. In addition, we find that Lu AF88273 also inhibits serotonin transport in a noncompetitive manner. The binding pose of Lu AF88273 in the allosteric site of hSERT is determined with extensive molecular dynamics simulations and rigorous absolute binding free energy perturbation (FEP) calculations, which show that a part of the compound occupies a dynamically formed small cavity. The predicted binding location and pose are validated by site-directed mutagenesis and can explain much of the structure-activity relationship of these inhibitors using the relative binding FEP calculations. Together, our findings provide a promising lead compound and the structural basis for the development of allosteric drugs targeting hSERT. Further, they demonstrate that the divergent allosteric sites of neurotransmitter transporters can be selectively targeted.


Subject(s)
Citalopram , Serotonin Plasma Membrane Transport Proteins , Humans , Antidepressive Agents/pharmacology , Citalopram/chemistry , Citalopram/pharmacology , Selective Serotonin Reuptake Inhibitors , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism
7.
Commun Chem ; 6(1): 222, 2023 Oct 14.
Article in English | MEDLINE | ID: mdl-37838760

ABSTRACT

Computational techniques can speed up the identification of hits and accelerate the development of candidate molecules for drug discovery. Among techniques for predicting relative binding affinities, the most consistently accurate is free energy perturbation (FEP), a class of rigorous physics-based methods. However, uncertainty remains about how accurate FEP is and can ever be. Here, we present what we believe to be the largest publicly available dataset of proteins and congeneric series of small molecules, and assess the accuracy of the leading FEP workflow. To ascertain the limit of achievable accuracy, we also survey the reproducibility of experimental relative affinity measurements. We find a wide variability in experimental accuracy and a correspondence between binding and functional assays. When careful preparation of protein and ligand structures is undertaken, FEP can achieve accuracy comparable to experimental reproducibility. Throughout, we highlight reliable protocols that can help maximize the accuracy of FEP in prospective studies.

8.
J Chem Theory Comput ; 19(11): 3080-3090, 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37219932

ABSTRACT

Structure-based drug design frequently operates under the assumption that a single holo structure is relevant. However, a large number of crystallographic examples clearly show that multiple conformations are possible. In those cases, the protein reorganization free energy must be known to accurately predict binding free energies for ligands. Only then can the energetic preference among these multiple protein conformations be utilized to design ligands with stronger binding potency and selectivity. Here, we present a computational method to quantify these protein reorganization free energies. We test it on two retrospective drug design cases, Abl kinase and HSP90, and illustrate how alternative holo conformations can be derisked and lead to large boosts in affinity. This method will allow computer-aided drug design to better support complex protein targets.


Subject(s)
Drug Design , HSP90 Heat-Shock Proteins , Ligands , Retrospective Studies , Protein Conformation , Protein Binding , Binding Sites
9.
J Chem Inf Model ; 63(10): 3171-3185, 2023 05 22.
Article in English | MEDLINE | ID: mdl-37167486

ABSTRACT

In the hit identification stage of drug discovery, a diverse chemical space needs to be explored to identify initial hits. Contrary to empirical scoring functions, absolute protein-ligand binding free-energy perturbation (ABFEP) provides a theoretically more rigorous and accurate description of protein-ligand binding thermodynamics and could, in principle, greatly improve the hit rates in virtual screening. In this work, we describe an implementation of an accurate and reliable ABFEP method in FEP+. We validated the ABFEP method on eight congeneric compound series binding to eight protein receptors including both neutral and charged ligands. For ligands with net charges, the alchemical ion approach is adopted to avoid artifacts in electrostatic potential energy calculations. The calculated binding free energies correlate with experimental results with a weighted average of R2 = 0.55 for the entire dataset. We also observe an overall root-mean-square error (RMSE) of 1.1 kcal/mol after shifting the zero-point of the simulation data to match the average experimental values. Through ABFEP calculations using apo versus holo protein structures, we demonstrated that the protein conformational and protonation state changes between the apo and holo proteins are the main physical factors contributing to the protein reorganization free energy manifested by the overestimation of raw ABFEP calculated binding free energies using the holo structures of the proteins. Furthermore, we performed ABFEP calculations in three virtual screening applications for hit enrichment. ABFEP greatly improves the hit rates as compared to docking scores or other methods like metadynamics. The good performance of ABFEP in rank ordering compounds demonstrated in this work confirms it as a useful tool to improve the hit rates in virtual screening, thus facilitating hit discovery.


Subject(s)
Proteins , Ligands , Protein Binding , Entropy , Proteins/chemistry , Thermodynamics
10.
J Chem Theory Comput ; 18(12): 7193-7204, 2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36384001

ABSTRACT

Accurate prediction of the pKa's of protein residues is crucial to many applications in biological simulation and drug discovery. Here, we present the use of free energy perturbation (FEP) calculations for the prediction of single-protein residue pKa values. We begin with an initial set of 191 residues with experimentally determined pKa values. To isolate sampling limitations from force field inaccuracies, we develop an algorithm to classify residues whose environments are significantly affected by crystal packing effects. We then report an approach to identify buried histidines that require significant sampling beyond what is achieved in typical FEP calculations. We therefore define a clean data set not requiring algorithms capable of predicting major conformational changes on which other pKa prediction methods can be tested. On this data set, we report an RMSE of 0.76 pKa units for 35 ASP residues, 0.51 pKa units for 44 GLU residues, and 0.67 pKa units for 76 HIS residues.


Subject(s)
Drug Discovery , Proteins , Entropy , Proteins/chemistry , Computer Simulation , Algorithms , Hydrogen-Ion Concentration
11.
J Chem Inf Model ; 62(3): 703-717, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35061383

ABSTRACT

The accurate prediction of binding affinity between protein and small molecules with free energy methods, particularly the difference in binding affinities via relative binding free energy calculations, has undergone a dramatic increase in use and impact over recent years. The improvements in methodology, hardware, and implementation can deliver results with less than 1 kcal/mol mean unsigned error between calculation and experiment. This is a remarkable achievement and beckons some reflection on the significance of calculation approaching the accuracy of experiment. In this article, we describe a statistical analysis of the implications of variance (standard deviation) of both experimental and calculated binding affinities with respect to the unknown true binding affinity. We reveal that plausible ratios of standard deviation in experiment and calculation can lead to unexpected outcomes for assessing the performance of predictions. The work extends beyond the case of binding free energies to other affinity or property prediction methods.


Subject(s)
Proteins , Entropy , Ligands , Protein Binding , Proteins/chemistry , Thermodynamics
12.
J Chem Theory Comput ; 17(7): 4291-4300, 2021 Jul 13.
Article in English | MEDLINE | ID: mdl-34096718

ABSTRACT

We report on the development and validation of the OPLS4 force field. OPLS4 builds upon our previous work with OPLS3e to improve model accuracy on challenging regimes of drug-like chemical space that includes molecular ions and sulfur-containing moieties. A novel parametrization strategy for charged species, which can be extended to other systems, is introduced. OPLS4 leads to improved accuracy on benchmarks that assess small-molecule solvation and protein-ligand binding.

13.
J Chem Inf Model ; 61(3): 1412-1426, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33661005

ABSTRACT

Drug design with patient centricity for ease of administration and pill burden requires robust understanding of the impact of chemical modifications on relevant physicochemical properties early in lead optimization. To this end, we have developed a physics-based ensemble approach to predict aqueous thermodynamic crystalline solubility, with a 2D chemical structure as the input. Predictions for the bromodomain and extraterminal domain (BET) inhibitor series show very close match (0.5 log unit) with measured thermodynamic solubility for cases with low crystal anisotropy and good match (1 log unit) for high anisotropy structures. The importance of thermodynamic solubility is clearly demonstrated by up to a 4 log unit drop in solubility compared to kinetic (amorphous) solubility in some cases and implications thereof, for instance on human dose. We have also demonstrated that incorporating predicted crystal structures in thermodynamic solubility prediction is necessary to differentiate (up to 4 log unit) between solubility of molecules within the series. Finally, our physics-based ensemble approach provides valuable structural insights into the origins of 3-D conformational landscapes, crystal polymorphism, and anisotropy that can be leveraged for both drug design and development.


Subject(s)
Physics , Water , Humans , Molecular Conformation , Solubility , Thermodynamics
14.
J Chem Theory Comput ; 17(1): 450-462, 2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33372778

ABSTRACT

Linking two fragments binding in nearby subpockets together has become an important technique in fragment-based drug discovery to optimize the binding potency of fragment hits. Despite the expected favorable translational and orientational entropic contribution to the binding free energy of the linked molecule, brute force enumeration of chemical linker for linking fragments is rarely successful, and the vast majority of linked molecules do not exhibit the expected gains of binding potency. In this paper, we examine the physical factors that contribute to the change of binding free energy from fragment linking and develop a method to rigorously calculate these different physical contributions. We find from these analyses that multiple confounding factors make successful fragment linking strategies rare, including (1) possible change of the binding mode of the fragments in the linked state compared to separate binding of the fragments, (2) unfavorable intramolecular strain energy of the bioactive conformation of the linked molecule, (3) unfavorable interaction between the linker and the protein, (4) favorable interaction energies between two fragments in solution when not chemically linked that offset the expected entropy loss for the formation of fragment pair, (5) complex compensating configurational entropic effects beyond the simplistic rotational and translational analysis. We here have applied a statistically mechanically rigorous approach to compute the fragment linking coefficients of 10 pharmaceutically interesting systems and quantify the contribution of each physical component to the binding free energy of the linked molecule. Based on these studies, we have found that the change in the relative configurational entropy of the two fragments in the protein binding pocket (a term neglected to our knowledge in all previous analyses) substantially offsets the favorable expected rotational and translational entropic contributions to the binding free energy of the linked molecule. This configurational restriction of the fragments in the binding pocket of the proteins is found to be, in our analysis, the dominant reason why most fragment linking strategies do not exhibit the expected gains of binding potency. These findings have further provided rich physical insights, which we expect should facilitate more successful fragment linking strategies to be formulated in the future.


Subject(s)
Drug Discovery , Proteins/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Binding Sites , Drug Design , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Protein Conformation , Proteins/chemistry , Thermodynamics
15.
J Chem Inf Model ; 60(12): 6211-6227, 2020 12 28.
Article in English | MEDLINE | ID: mdl-33119284

ABSTRACT

Alchemical free-energy calculations are now widely used to drive or maintain potency in small-molecule lead optimization with a roughly 1 kcal/mol accuracy. Despite this, the potential to use free-energy calculations to drive optimization of compound selectivity among two similar targets has been relatively unexplored in published studies. In the most optimistic scenario, the similarity of binding sites might lead to a fortuitous cancellation of errors and allow selectivity to be predicted more accurately than affinity. Here, we assess the accuracy with which selectivity can be predicted in the context of small-molecule kinase inhibitors, considering the very similar binding sites of human kinases CDK2 and CDK9 as well as another series of ligands attempting to achieve selectivity between the more distantly related kinases CDK2 and ERK2. Using a Bayesian analysis approach, we separate systematic from statistical errors and quantify the correlation in systematic errors between selectivity targets. We find that, in the CDK2/CDK9 case, a high correlation in systematic errors suggests that free-energy calculations can have significant impact in aiding chemists in achieving selectivity, while in more distantly related kinases (CDK2/ERK2), the correlation in systematic error suggests that fortuitous cancellation may even occur between systems that are not as closely related. In both cases, the correlation in systematic error suggests that longer simulations are beneficial to properly balance statistical error with systematic error to take full advantage of the increase in apparent free-energy calculation accuracy in selectivity prediction.


Subject(s)
Drug Design , Molecular Dynamics Simulation , Bayes Theorem , Binding Sites , Humans , Ligands , Protein Binding , Thermodynamics
16.
J Chem Theory Comput ; 16(11): 6926-6937, 2020 Nov 10.
Article in English | MEDLINE | ID: mdl-32910652

ABSTRACT

To address some of the inherent challenges in modeling metalloenzymes, we here report an extension to the functional form of the OPLS3e force field to include terms adopted from the ligand field molecular mechanics (LFMM) model, including the angular overlap and Morse potential terms. The integration of these terms with OPLS3e, herein referred to as OPLS3e+M, improves the description of metal-ligand interactions and provides accurate relative binding energies and geometric preferences of transition-metal complexes by training to gas-phase density functional theory (DFT) energies. For [Cu(H2O)4]2+, OPLS3e+M significantly improves H2O binding energies and the geometric preference of the tetra-aqua Cu2+ complex. In addition, we conduct free-energy perturbation calculations on two pharmaceutically relevant metalloenzyme targets, which include chemical modifications at varying proximity to the binding-site metals, including changes to the metal-binding moiety of the ligand itself. The extensions made to OPLS3e lead to accurate predicted relative binding free energies for these series (mean unsigned error of 1.29 kcal mol-1). Our results provide evidence that integration of the LFMM model with OPLS3e can be utilized to predict thermodynamic quantities for such systems near chemical accuracy. With these improvements, we anticipate that robust free-energy perturbation calculations can be employed to accelerate the drug development efforts for metalloenzyme targets.


Subject(s)
Density Functional Theory , Drug Discovery , Metalloproteins/chemistry , Metalloproteins/metabolism , Ligands , Molecular Dynamics Simulation , Thermodynamics
17.
J Chem Theory Comput ; 16(10): 6061-6076, 2020 Oct 13.
Article in English | MEDLINE | ID: mdl-32955877

ABSTRACT

The prediction of protein-ligand binding affinities using free energy perturbation (FEP) is becoming increasingly routine in structure-based drug discovery. Most FEP packages use molecular dynamics (MD) to sample the configurations of proteins and ligands, as MD is well-suited to capturing coupled motion. However, MD can be prohibitively inefficient at sampling water molecules that are buried within binding sites, which has severely limited the domain of applicability of FEP and its prospective usage in drug discovery. In this paper, we present an advancement of FEP that augments MD with grand canonical Monte Carlo (GCMC), an enhanced sampling method, to overcome the problem of sampling water. We accomplished this without degrading computational performance. On both old and newly assembled data sets of protein-ligand complexes, we show that the use of GCMC in FEP is essential for accurate and robust predictions for ligand perturbations that disrupt buried water.


Subject(s)
Density Functional Theory , Thermodynamics , Water/chemistry , Monte Carlo Method
18.
J Chem Inf Model ; 60(7): 3489-3498, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32539379

ABSTRACT

A tremendous research and development effort was exerted toward combating chronic hepatitis C, ultimately leading to curative oral treatments, all of which are targeting viral proteins. Despite the advantage of numerous targets allowing for broad hepatitis C virus (HCV) genotype coverage, the only host target inhibitors that advanced into clinical development were Cyclosporin A based cyclophilin inhibitors. While cyclosporin-based molecules typically require a fermentation process, Gilead successfully pursued a fully synthetic, oral program based on Sanglifehrin A. The drug discovery process, though greatly helped by facile crystallography, was still hampered by the limitations in the accuracy of predictive computational methods for prioritizing compound ideas. Recent advances in accuracy and speed of free energy perturbation (FEP) methods, however, are attractive for prioritizing and derisking synthetically challenging molecules and potentially could have had a significant impact on the speed of the development of this program. Here in our simulated prospective study, the binding free energies of 26 macrocyclic cyclophilin inhibitors were blindly predicted using FEP+ to test this hypothesis. The predictions had a low mean unsigned error (MUE) (1.1 kcal/mol) and accurately reproduced many design decisions from the program, suggesting that FEP+ has the potential to drive synthetic chemistry efforts by more accurately ranking compounds with nonintuitive structure-activity relationships (SARs).


Subject(s)
Drug Discovery , Entropy , Prospective Studies , Structure-Activity Relationship , Thermodynamics
19.
Biophys J ; 119(1): 115-127, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32533939

ABSTRACT

Accurately predicting the protein thermostability changes upon single point mutations in silico is a challenge that has implications for understanding diseases as well as industrial applications of protein engineering. Free energy perturbation (FEP) has been applied to predict the effect of single point mutations on protein stability for over 40 years and emerged as a potentially reliable prediction method with reasonable throughput. However, applications of FEP in protein stability calculations in industrial settings have been hindered by a number of limitations, including the inability to model mutations to and from prolines in which the bonded topology of the backbone is modified and the complexity in modeling charge-changing mutations. In this study, we have extended the FEP+ protocol to enable the accurate modeling of the effects on protein stability from proline mutations and from charge-changing mutations. We also evaluated the influence of the unfolded model in the stability calculations using increasingly longer peptides with native sequence and conformations. With the abovementioned improvements, the accuracy of FEP predictions of protein stability over a data set of 87 mutations on five different proteins has drastically improved compared with previous studies, with a mean unsigned error of 0.86 kcal/mol and root mean square error of 1.11 kcal/mol, comparable with the accuracy of previously published state-of-the-art small-molecule relative binding affinity calculations, which have been shown to be capable of driving discovery projects.


Subject(s)
Point Mutation , Proteins , Entropy , Peptides , Protein Stability , Proteins/genetics , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...