123 research outputs found

    Predicting Binding Affinity of CSAR Ligands Using Both Structure-Based and Ligand-Based Approaches

    Get PDF
    We report on the prediction accuracy of ligand-based (2D QSAR) and structure-based (MedusaDock) methods used both independently and in consensus for ranking the congeneric series of ligands binding to three protein targets (UK, ERK2, and CHK1) from the CSAR 2011 benchmark exercise. An ensemble of predictive QSAR models was developed using known binders of these three targets extracted from the publicly-available ChEMBL database. Selected models were used to predict the binding affinity of CSAR compounds towards the corresponding targets and rank them accordingly; the overall ranking accuracy evaluated by Spearman correlation was as high as 0.78 for UK, 0.60 for ERK2, and 0.56 for CHK1, placing our predictions in top-10% among all the participants. In parallel, MedusaDock designed to predict reliable docking poses was also used for ranking the CSAR ligands according to their docking scores; the resulting accuracy (Spearman correlation) for UK, ERK2, and CHK1 were 0.76, 0.31, and 0.26, respectively. In addition, performance of several consensus approaches combining MedusaDock and QSAR predicted ranks altogether has been explored; the best approach yielded Spearman correlation coefficients for UK, ERK2, and CHK1 of 0.82, 0.50, and 0.45, respectively. This study shows that (i) externally validated 2D QSAR models were capable of ranking CSAR ligands at least as accurately as more computationally intensive structure-based approaches used both by us and by other groups and (ii) ligand-based QSAR models can complement structure-based approaches by boosting the prediction performances when used in consensus

    Protein-Ligand Scoring with Convolutional Neural Networks

    Full text link
    Computational approaches to drug discovery can reduce the time and cost associated with experimental assays and enable the screening of novel chemotypes. Structure-based drug design methods rely on scoring functions to rank and predict binding affinities and poses. The ever-expanding amount of protein-ligand binding and structural data enables the use of deep machine learning techniques for protein-ligand scoring. We describe convolutional neural network (CNN) scoring functions that take as input a comprehensive 3D representation of a protein-ligand interaction. A CNN scoring function automatically learns the key features of protein-ligand interactions that correlate with binding. We train and optimize our CNN scoring functions to discriminate between correct and incorrect binding poses and known binders and non-binders. We find that our CNN scoring function outperforms the AutoDock Vina scoring function when ranking poses both for pose prediction and virtual screening

    Computational structure‐based drug design: Predicting target flexibility

    Get PDF
    The role of molecular modeling in drug design has experienced a significant revamp in the last decade. The increase in computational resources and molecular models, along with software developments, is finally introducing a competitive advantage in early phases of drug discovery. Medium and small companies with strong focus on computational chemistry are being created, some of them having introduced important leads in drug design pipelines. An important source for this success is the extraordinary development of faster and more efficient techniques for describing flexibility in three‐dimensional structural molecular modeling. At different levels, from docking techniques to atomistic molecular dynamics, conformational sampling between receptor and drug results in improved predictions, such as screening enrichment, discovery of transient cavities, etc. In this review article we perform an extensive analysis of these modeling techniques, dividing them into high and low throughput, and emphasizing in their application to drug design studies. We finalize the review with a section describing our Monte Carlo method, PELE, recently highlighted as an outstanding advance in an international blind competition and industrial benchmarks.We acknowledge the BSC-CRG-IRB Joint Research Program in Computational Biology. This work was supported by a grant from the Spanish Government CTQ2016-79138-R.J.I. acknowledges support from SVP-2014-068797, awarded by the Spanish Government.Peer ReviewedPostprint (author's final draft

    Pre-Training on Large-Scale Generated Docking Conformations with HelixDock to Unlock the Potential of Protein-ligand Structure Prediction Models

    Full text link
    Protein-ligand structure prediction is an essential task in drug discovery, predicting the binding interactions between small molecules (ligands) and target proteins (receptors). Although conventional physics-based docking tools are widely utilized, their accuracy is compromised by limited conformational sampling and imprecise scoring functions. Recent advances have incorporated deep learning techniques to improve the accuracy of structure prediction. Nevertheless, the experimental validation of docking conformations remains costly, it raises concerns regarding the generalizability of these deep learning-based methods due to the limited training data. In this work, we show that by pre-training a geometry-aware SE(3)-Equivariant neural network on a large-scale docking conformation generated by traditional physics-based docking tools and then fine-tuning with a limited set of experimentally validated receptor-ligand complexes, we can achieve outstanding performance. This process involved the generation of 100 million docking conformations, consuming roughly 1 million CPU core days. The proposed model, HelixDock, aims to acquire the physical knowledge encapsulated by the physics-based docking tools during the pre-training phase. HelixDock has been benchmarked against both physics-based and deep learning-based baselines, showing that it outperforms its closest competitor by over 40% for RMSD. HelixDock also exhibits enhanced performance on a dataset that poses a greater challenge, thereby highlighting its robustness. Moreover, our investigation reveals the scaling laws governing pre-trained structure prediction models, indicating a consistent enhancement in performance with increases in model parameters and pre-training data. This study illuminates the strategic advantage of leveraging a vast and varied repository of generated data to advance the frontiers of AI-driven drug discovery

    Modeling Protein-Ligand Interactions with Applications to Drug Design

    Get PDF

    Ranking docking poses by graph matching of protein–ligand interactions: lessons learned from the D3R Grand Challenge 2

    Get PDF
    International audienceA novel docking challenge has been set by the Drug Design Data Resource (D3R) in order to predict the pose and affinity ranking of a set of Farnesoid X receptor (FXR) agonists, prior to the public release of their bound X-ray structures and potencies. In a first phase, 36 agonists were docked to 26 Protein Data Bank (PDB) structures of the FXR receptor, and next rescored using the in-house developed GRIM method. GRIM aligns protein–ligand interaction patterns of docked poses to those of available PDB templates for the target protein, and rescore poses by a graph matching method. In agreement with results obtained during the previous 2015 docking challenge, we clearly show that GRIM rescoring improves the overall quality of top-ranked poses by prioritizing interaction patterns already visited in the PDB. Importantly, this challenge enables us to refine the applicability domain of the method by better defining the conditions of its success. We notably show that rescoring apolar ligands in hydrophobic pockets leads to frequent GRIM failures. In the second phase, 102 FXR agonists were ranked by decreasing affinity according to the Gibbs free energy of the corresponding GRIM-selected poses, computed by the HYDE scoring function. Interestingly, this fast and simple rescoring scheme provided the third most accurate ranking method among 57 contributions. Although the obtained ranking is still unsuitable for hit to lead optimization, the GRIM–HYDE scoring scheme is accurate and fast enough to post-process virtual screening dat

    DeepRLI: A Multi-objective Framework for Universal Protein--Ligand Interaction Prediction

    Full text link
    Protein (receptor)--ligand interaction prediction is a critical component in computer-aided drug design, significantly influencing molecular docking and virtual screening processes. Despite the development of numerous scoring functions in recent years, particularly those employing machine learning, accurately and efficiently predicting binding affinities for protein--ligand complexes remains a formidable challenge. Most contemporary methods are tailored for specific tasks, such as binding affinity prediction, binding pose prediction, or virtual screening, often failing to encompass all aspects. In this study, we put forward DeepRLI, a novel protein--ligand interaction prediction architecture. It encodes each protein--ligand complex into a fully connected graph, retaining the integrity of the topological and spatial structure, and leverages the improved graph transformer layers with cosine envelope as the central module of the neural network, thus exhibiting superior scoring power. In order to equip the model to generalize to conformations beyond the confines of crystal structures and to adapt to molecular docking and virtual screening tasks, we propose a multi-objective strategy, that is, the model outputs three scores for scoring and ranking, docking, and screening, and the training process optimizes these three objectives simultaneously. For the latter two objectives, we augment the dataset through a docking procedure, incorporate suitable physics-informed blocks and employ an effective contrastive learning approach. Eventually, our model manifests a balanced performance across scoring, ranking, docking, and screening, thereby demonstrating its ability to handle a range of tasks. Overall, this research contributes a multi-objective framework for universal protein--ligand interaction prediction, augmenting the landscape of structure-based drug design

    Ligand-Protein Binding Affinity Prediction Using Machine Learning Scoring Functions.

    Get PDF
    In recent years, artificial intelligence makes its appearance in extremely different fields with promising results able to produce enormous steps forward in some circumstances. In chemoinformatics the use of machine learning technique, in particular, allows the scientific community to build apparently accurate scoring functions for computational docking. These types of scoring functions can overperform classic ones, the type of scoring functions used until now. However the comparison between classic and machine learning scoring functions are based on particular tests which can favour these latter, as highlighted by some studies. In particular the machine learning scoring functions, per definition, must be trained on some data, passing to the model the instances chosen to describe the complexes and the relative ligand-protein affinity. In these conditions the scoring power of the machine learning scoring functions can be evaluated on different dataset and the scoring functions performance recorded can be different depending on it. In particular, datasets very similar to the one used for the training phase of the machine learning scoring function can facilitate in reaching high performance in the scoring power. The objective of the present study is to verify the real efficiency and the effective performances of the new born machine learning scoring functions. Our aim is to give an answer to the scientific community about the doubts on the fact that the machine learning scoring function can be or not the revolutionary road to be followed in the field of chemioinformatic and drug discovery. In order to do this many tests are conducted and a definitive test protocol to be executed to exhaustive validate a new machine learning scoring function is proposed . Here we investigate what are the circumstances in which a machine learning scoring function produces overestimated performances and why it can happen. As a possible solution we propose a tests protocol to be followed in order to guarantee a real performance descriptions of machine learning scoring functions. Eventually an effective and innovative solution in the field of machine learning scoring functions is proposed. It consists in the use of per-target scoring functions which are machine learning scoring functions created using complexes coming from a single protein and able to predict the affinity of complexes which use that target. The data used to build the model are synthetic and for this reason are easy to be created. The performances on the target chosen are better than the ones obtained with basic model of scoring functions and machine learning scoring functions trained on database composed by more than one protein
    • 

    corecore