Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Chem Inf Model ; 64(7): 2488-2495, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38113513

ABSTRACT

Deep learning methods that predict protein-ligand binding have recently been used for structure-based virtual screening. Many such models have been trained using protein-ligand complexes with known crystal structures and activities from the PDBBind data set. However, because PDBbind only includes 20K complexes, models typically fail to generalize to new targets, and model performance is on par with models trained with only ligand information. Conversely, the ChEMBL database contains a wealth of chemical activity information but includes no information about binding poses. We introduce BigBind, a data set that maps ChEMBL activity data to proteins from the CrossDocked data set. BigBind comprises 583 K ligand activities and includes 3D structures of the protein binding pockets. Additionally, we augmented the data by adding an equal number of putative inactives for each target. Using this data, we developed Banana (basic neural network for binding affinity), a neural network-based model to classify active from inactive compounds, defined by a 10 µM cutoff. Our model achieved an AUC of 0.72 on BigBind's test set, while a ligand-only model achieved an AUC of 0.59. Furthermore, Banana achieved competitive performance on the LIT-PCBA benchmark (median EF1% 1.81) while running 16,000 times faster than molecular docking with Gnina. We suggest that Banana, as well as other models trained on this data set, will significantly improve the outcomes of prospective virtual screening tasks.


Subject(s)
Proteins , Ubiquitin-Protein Ligases , Molecular Docking Simulation , Ligands , Prospective Studies , Proteins/chemistry , Protein Binding , Ubiquitin-Protein Ligases/metabolism
2.
ACS Omega ; 8(2): 2389-2397, 2023 Jan 17.
Article in English | MEDLINE | ID: mdl-36687059

ABSTRACT

Drug design involves the process of identifying and designing molecules that bind well to a given receptor. A vital computational component of this process is the protein-ligand interaction scoring functions that evaluate the binding ability of various molecules or ligands with a given protein receptor binding pocket reasonably accurately. With the publicly available protein-ligand binding affinity data sets in both sequential and structural forms, machine learning methods have gained traction as a top choice for developing such scoring functions. While the performance shown by these models is optimistic, there are several hidden biases present in these data sets themselves that affect the utility of such models for practical purposes such as virtual screening. In this work, we use published methods to systematically investigate several such factors or biases present in these data sets. In our analysis, we highlight the importance of considering sequence, protein-ligand interaction, and pocket structure similarity while constructing data splits and provide an explanation for good protein-only and ligand-only performances in some data sets. Through this study, we provide to the community several pointers for the design of binding affinity predictors and data sets for reliable applicability.

3.
J Chem Inf Model ; 62(9): 2064-2076, 2022 05 09.
Article in English | MEDLINE | ID: mdl-34694798

ABSTRACT

Application of deep learning techniques for de novo generation of molecules, termed as inverse molecular design, has been gaining enormous traction in drug design. The representation of molecules in SMILES notation as a string of characters enables the usage of state of the art models in natural language processing, such as Transformers, for molecular design in general. Inspired by generative pre-training (GPT) models that have been shown to be successful in generating meaningful text, we train a transformer-decoder on the next token prediction task using masked self-attention for the generation of druglike molecules in this study. We show that our model, MolGPT, performs on par with other previously proposed modern machine learning frameworks for molecular generation in terms of generating valid, unique, and novel molecules. Furthermore, we demonstrate that the model can be trained conditionally to control multiple properties of the generated molecules. We also show that the model can be used to generate molecules with desired scaffolds as well as desired molecular properties by conditioning the generation on scaffold SMILES strings of desired scaffolds and property values. Using saliency maps, we highlight the interpretability of the generative process of the model.


Subject(s)
Drug Design , Machine Learning
4.
J Chem Inf Model ; 62(21): 5069-5079, 2022 11 14.
Article in English | MEDLINE | ID: mdl-34374539

ABSTRACT

A structure-based drug design pipeline involves the development of potential drug molecules or ligands that form stable complexes with a given receptor at its binding site. A prerequisite to this is finding druggable and functionally relevant binding sites on the 3D structure of the protein. Although several methods for detecting binding sites have been developed beforehand, a majority of them surprisingly fail in the identification and ranking of binding sites accurately. The rapid adoption and success of deep learning algorithms in various sections of structural biology beckons the usage of such algorithms for accurate binding site detection. As a combination of geometry based software and deep learning, we report a novel framework, DeepPocket that utilizes 3D convolutional neural networks for the rescoring of pockets identified by Fpocket and further segments these identified cavities on the protein surface. Apart from this, we also propose another data set SC6K containing protein structures submitted in the Protein Data Bank (PDB) from January 1st, 2018, until February 28th, 2020, for ligand binding site (LBS) detection. DeepPocket's results on various binding site data sets and SC6K highlight its better performance over current state-of-the-art methods and good generalization ability over novel structures.


Subject(s)
Neural Networks, Computer , Proteins , Ligands , Binding Sites , Proteins/chemistry , Software , Algorithms
5.
J Cheminform ; 13(1): 43, 2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34108002

ABSTRACT

Molecular docking computationally predicts the conformation of a small molecule when binding to a receptor. Scoring functions are a vital piece of any molecular docking pipeline as they determine the fitness of sampled poses. Here we describe and evaluate the 1.0 release of the Gnina docking software, which utilizes an ensemble of convolutional neural networks (CNNs) as a scoring function. We also explore an array of parameter values for Gnina 1.0 to optimize docking performance and computational cost. Docking performance, as evaluated by the percentage of targets where the top pose is better than 2Å root mean square deviation (Top1), is compared to AutoDock Vina scoring when utilizing explicitly defined binding pockets or whole protein docking. GNINA, utilizing a CNN scoring function to rescore the output poses, outperforms AutoDock Vina scoring on redocking and cross-docking tasks when the binding pocket is defined (Top1 increases from 58% to 73% and from 27% to 37%, respectively) and when the whole protein defines the binding pocket (Top1 increases from 31% to 38% and from 12% to 16%, respectively). The derived ensemble of CNNs generalizes to unseen proteins and ligands and produces scores that correlate well with the root mean square deviation to the known binding pose. We provide the 1.0 version of GNINA under an open source license for use as a molecular docking tool at https://github.com/gnina/gnina .

SELECTION OF CITATIONS
SEARCH DETAIL
...