Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo del documento
Publication year range
1.
Brief Bioinform ; 25(3)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38647154

RESUMEN

Molecular generative models have exhibited promising capabilities in designing molecules from scratch with high binding affinities in a predetermined protein pocket, offering potential synergies with traditional structural-based drug design strategy. However, the generative processes of such models are random and the atomic interaction information between ligand and protein are ignored. On the other hand, the ligand has high propensity to bind with residues called hotspots. Hotspot residues contribute to the majority of the binding free energies and have been recognized as appealing targets for designed molecules. In this work, we develop an interaction prompt guided diffusion model, InterDiff to deal with the challenges. Four kinds of atomic interactions are involved in our model and represented as learnable vector embeddings. These embeddings serve as conditions for individual residue to guide the molecular generative process. Comprehensive in silico experiments evince that our model could generate molecules with desired ligand-protein interactions in a guidable way. Furthermore, we validate InterDiff on two realistic protein-based therapeutic agents. Results show that InterDiff could generate molecules with better or similar binding mode compared to known targeted drugs.


Asunto(s)
Proteínas , Proteínas/química , Proteínas/metabolismo , Ligandos , Unión Proteica , Diseño de Fármacos , Modelos Moleculares , Algoritmos , Sitios de Unión , Simulación por Computador
2.
Brief Bioinform ; 23(4)2022 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-35830870

RESUMEN

We construct a protein-protein interaction (PPI) targeted drug-likeness dataset and propose a deep molecular generative framework to generate novel drug-likeness molecules from the features of the seed compounds. This framework gains inspiration from published molecular generative models, uses the key features associated with PPI inhibitors as input and develops deep molecular generative models for de novo molecular design of PPI inhibitors. For the first time, quantitative estimation index for compounds targeting PPI was applied to the evaluation of the molecular generation model for de novo design of PPI-targeted compounds. Our results estimated that the generated molecules had better PPI-targeted drug-likeness and drug-likeness. Additionally, our model also exhibits comparable performance to other several state-of-the-art molecule generation models. The generated molecules share chemical space with iPPI-DB inhibitors as demonstrated by chemical space analysis. The peptide characterization-oriented design of PPI inhibitors and the ligand-based design of PPI inhibitors are explored. Finally, we recommend that this framework will be an important step forward for the de novo design of PPI-targeted therapeutics.


Asunto(s)
Diseño de Fármacos , Redes Neurales de la Computación , Ligandos , Modelos Moleculares
3.
Methods ; 210: 52-59, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36682423

RESUMEN

The process of design/discovery of drugs involves the identification and design of novel molecules that have the desired properties and bind well to a given disease-relevant target. One of the main challenges to effectively identify potential drug candidates is to explore the vast drug-like chemical space to find novel chemical structures with desired physicochemical properties and biological characteristics. Moreover, the chemical space of currently available molecular libraries is only a small fraction of the total possible drug-like chemical space. Deep molecular generative models have received much attention and provide an alternative approach to the design and discovery of molecules. To efficiently explore the drug-like space, we first constructed the drug-like dataset and then performed the generative design of drug-like molecules using a Conditional Randomized Transformer approach with the molecular access system (MACCS) fingerprint as a condition and compared it with previously published molecular generative models. The results show that the deep molecular generative model explores the wider drug-like chemical space. The generated drug-like molecules share the chemical space with known drugs, and the drug-like space captured by the combination of quantitative estimation of drug-likeness (QED) and quantitative estimate of protein-protein interaction targeting drug-likeness (QEPPI) can cover a larger drug-like space. Finally, we show the potential application of the model in design of inhibitors of MDM2-p53 protein-protein interaction. Our results demonstrate the potential application of deep molecular generative models for guided exploration in drug-like chemical space and molecular design.


Asunto(s)
Diseño de Fármacos , Modelos Moleculares
4.
Methods ; 211: 10-22, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36764588

RESUMEN

Deep learning is improving and changing the process of de novo molecular design at a rapid pace. In recent years, great progress has been made in drug discovery and development by using deep generative models for de novo molecular design. However, most of the existing methods are string-based or graph-based and are limited by the lack of some very important properties, such as the three-dimensional information of molecules. We propose DNMG, a deep generative adversarial network (GAN) combined with transfer learning. Specifically, we use a Wasserstein-variant GAN based network architecture that considers the 3D grid spatial information of the ligand with atomic physicochemical properties to generate a representation of the molecule, which is then parsed into SMILES strings using an improved captioning network. Comprehensive in experiments demonstrate the ability of DNMG to generate valid and novel drug-like ligands. The DNMG model is used to design inhibitors for three targets, MK14, FNTA, and CDK2. The computational results show that the molecules generated by DNMG have better binding ability to the target proteins and better physicochemical properties. Overall, our deep generative model has excellent potential to generate molecules with high binding affinity for targets and explore the space of drug-like chemistry.


Asunto(s)
Diseño de Fármacos , Descubrimiento de Drogas , Modelos Moleculares , Descubrimiento de Drogas/métodos , Ligandos , Proteínas
5.
Brief Bioinform ; 22(6)2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34410338

RESUMEN

Artificial intelligence, such as deep generative methods, represents a promising solution to de novo design of molecules with the desired properties. However, generating new molecules with biological activities toward two specific targets remains an extremely difficult challenge. In this work, we conceive a novel computational framework, herein called dual-target ligand generative network (DLGN), for the de novo generation of bioactive molecules toward two given objectives. Via adversarial training and reinforcement learning, DLGN treats a sequence-based simplified molecular input line entry system (SMILES) generator as a stochastic policy for exploring chemical spaces. Two discriminators are then used to encourage the generation of molecules that belong to the intersection of two bioactive-compound distributions. In a case study, we employ our methods to design a library of dual-target ligands targeting dopamine receptor D2 and 5-hydroxytryptamine receptor 1A as new antipsychotics. Experimental results demonstrate that the proposed model can generate novel compounds with high similarity to both bioactive datasets in several structure-based metrics. Our model exhibits a performance comparable to that of various state-of-the-art multi-objective molecule generation models. We envision that this framework will become a generally applicable approach for designing dual-target drugs in silico.


Asunto(s)
Aprendizaje Profundo , Descubrimiento de Drogas/métodos , Ligandos , Algoritmos , Inteligencia Artificial , Biomarcadores , Fenómenos Químicos , Bases de Datos Farmacéuticas , Diseño de Fármacos , Proteínas , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda