Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Cheminform ; 15(1): 42, 2023 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-37031191

RESUMO

Artificial intelligence (AI)-based molecular design methods, especially deep generative models for generating novel molecule structures, have gratified our imagination to explore unknown chemical space without relying on brute-force exploration. However, whether designed by AI or human experts, the molecules need to be accessibly synthesized and biologically evaluated, and the trial-and-error process remains a resources-intensive endeavor. Therefore, AI-based drug design methods face a major challenge of how to prioritize the molecular structures with potential for subsequent drug development. This study indicates that common filtering approaches based on traditional screening metrics fail to differentiate AI-designed molecules. To address this issue, we propose a novel molecular filtering method, MolFilterGAN, based on a progressively augmented generative adversarial network. Comparative analysis shows that MolFilterGAN outperforms conventional screening approaches based on drug-likeness or synthetic ability metrics. Retrospective analysis of AI-designed discoidin domain receptor 1 (DDR1) inhibitors shows that MolFilterGAN significantly increases the efficiency of molecular triaging. Further evaluation of MolFilterGAN on eight external ligand sets suggests that MolFilterGAN is useful in triaging or enriching bioactive compounds across a wide range of target types. These results highlighted the importance of MolFilterGAN in evaluating molecules integrally and further accelerating molecular discovery especially combined with advanced AI generative models.

3.
J Chem Theory Comput ; 18(9): 5649-5658, 2022 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-35939398

RESUMO

The traditional approach of computational biology consists of calculating molecule properties by using approximate classical potentials. Interactions between atoms are described by an energy function derived from physical principles or fitted to experimental data. Their functional form is usually limited to pairwise interactions between atoms and does not consider complex multibody effects. More recently, neural networks have emerged as an alternative way of describing the interactions between biomolecules. In this approach, the energy function does not have an explicit functional form and is learned bottom-up from simulations at the atomistic or quantum level. In this study, we attempt a top-down approach and use deep learning methods to obtain an energy function by exploiting the large amount of experimental data acquired with years in the field of structural biology. The energy function is represented by a probability density model learned from a large repertoire of building blocks representing local clusters of amino acids paired with their sequence signature. We demonstrated the feasibility of this approach by generating a neural network energy function and testing its validity on several applications such as discriminating decoys, assessing qualities of structural models, sampling structural conformations, and designing new protein sequences. We foresee that, in the future, our methodology could exploit the continuously increasing availability of experimental data and simulations and provide a new method for the parametrization of protein energy functions.


Assuntos
Redes Neurais de Computação , Proteínas , Biologia Computacional/métodos , Física , Proteínas/química , Termodinâmica
4.
Sci China Life Sci ; 65(3): 529-539, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34319533

RESUMO

Artificial intelligence (AI) models usually require large amounts of high-quality training data, which is in striking contrast to the situation of small and biased data faced by current drug discovery pipelines. The concept of federated learning has been proposed to utilize distributed data from different sources without leaking sensitive information of the data. This emerging decentralized machine learning paradigm is expected to dramatically improve the success rate of AI-powered drug discovery. Here, we simulated the federated learning process with different property and activity datasets from different sources, among which overlapping molecules with high or low biases exist in the recorded values. Beyond the benefit of gaining more data, we also demonstrated that federated training has a regularization effect superior to centralized training on the pooled datasets with high biases. Moreover, different network architectures for clients and aggregation algorithms for coordinators have been compared on the performance of federated learning, where personalized federated learning shows promising results. Our work demonstrates the applicability of federated learning in predicting drug-related properties and highlights its promising role in addressing the small and biased data dilemma in drug discovery.


Assuntos
Inteligência Artificial , Descoberta de Drogas , Algoritmos , Conjuntos de Dados como Assunto , Canal de Potássio ERG1/antagonistas & inibidores
5.
J Med Chem ; 64(19): 14011-14027, 2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34533311

RESUMO

Artificial intelligence (AI) is booming. Among various AI approaches, generative models have received much attention in recent years. Inspired by these successes, researchers are now applying generative model techniques to de novo drug design, which has been considered as the "holy grail" of drug discovery. In this Perspective, we first focus on describing models such as recurrent neural network, autoencoder, generative adversarial network, transformer, and hybrid models with reinforcement learning. Next, we summarize the applications of generative models to drug design, including generating various compounds to expand the compound library and designing compounds with specific properties, and we also list a few publicly available molecular design tools based on generative models which can be used directly to generate molecules. In addition, we also introduce current benchmarks and metrics frequently used for generative models. Finally, we discuss the challenges and prospects of using generative models to aid drug design.


Assuntos
Inteligência Artificial , Desenho de Fármacos , Estrutura Molecular
6.
PLoS Comput Biol ; 17(9): e1009302, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34520464

RESUMO

A continuing challenge in modern medicine is the identification of safer and more efficacious drugs. Precision therapeutics, which have one molecular target, have been long promised to be safer and more effective than traditional therapies. This approach has proven to be challenging for multiple reasons including lack of efficacy, rapidly acquired drug resistance, and narrow patient eligibility criteria. An alternative approach is the development of drugs that address the overall disease network by targeting multiple biological targets ('polypharmacology'). Rational development of these molecules will require improved methods for predicting single chemical structures that target multiple drug targets. To address this need, we developed the Multi-Targeting Drug DREAM Challenge, in which we challenged participants to predict single chemical entities that target pro-targets but avoid anti-targets for two unrelated diseases: RET-based tumors and a common form of inherited Tauopathy. Here, we report the results of this DREAM Challenge and the development of two neural network-based machine learning approaches that were applied to the challenge of rational polypharmacology. Together, these platforms provide a potentially useful first step towards developing lead therapeutic compounds that address disease complexity through rational polypharmacology.


Assuntos
Desenvolvimento de Medicamentos , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-ret/antagonistas & inibidores , Tauopatias/tratamento farmacológico , Humanos , Neoplasias/metabolismo , Redes Neurais de Computação , Polifarmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
7.
Drug Discov Today ; 26(6): 1382-1393, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33609779

RESUMO

The goal of de novo drug design is to create novel chemical entities with desired biological activities and pharmacokinetics (PK) properties. Over recent years, with the development of artificial intelligence (AI) technologies, data-driven methods have rapidly gained in popularity in this field. Among them, graph neural networks (GNNs), a type of neural network directly operating on the graph structure data, have received extensive attention. In this review, we introduce the applications of GNNs in de novo drug design from three aspects: molecule scoring, molecule generation and optimization, and synthesis planning. Furthermore, we also discuss the current challenges and future directions of GNNs in de novo drug design.


Assuntos
Inteligência Artificial , Desenho de Fármacos/métodos , Redes Neurais de Computação , Descoberta de Drogas/métodos , Humanos , Tecnologia/métodos
8.
Eur J Med Chem ; 204: 112572, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32711293

RESUMO

Complex neuropsychiatric diseases such as schizophrenia require drugs that can target multiple G protein-coupled receptors (GPCRs) to modulate complex neuropsychiatric functions. Here, we report an automated system comprising a deep recurrent neural network (RNN) and a multitask deep neural network (MTDNN) to design and optimize multitarget antipsychotic drugs. The system has successfully generated novel molecule structures with desired multiple target activities, among which high-ranking compound 3 was synthesized, and demonstrated potent activities against dopamine D2, serotonin 5-HT1A and 5-HT2A receptors. Hit expansion based on the MTDNN was performed, 6 analogs of compound 3 were evaluated experimentally, among which compound 8 not only exhibited specific polypharmacology profiles but also showed antipsychotic effect in animal models with low potential for sedation and catalepsy, highlighting their suitability for further preclinical studies. The approach can be an efficient tool for designing lead compounds with multitarget profiles to achieve the desired efficacy in the treatment of complex neuropsychiatric diseases.


Assuntos
Aprendizado Profundo , Descoberta de Drogas/métodos , Terapia de Alvo Molecular , Esquizofrenia/tratamento farmacológico , Animais , Automação , Camundongos , Receptor 5-HT1A de Serotonina/metabolismo , Receptor 5-HT2A de Serotonina/metabolismo , Receptores de Dopamina D2/metabolismo , Esquizofrenia/metabolismo
9.
J Med Chem ; 63(16): 8723-8737, 2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31364850

RESUMO

The kinome-wide virtual profiling of small molecules with high-dimensional structure-activity data is a challenging task in drug discovery. Here, we present a virtual profiling model against a panel of 391 kinases based on large-scale bioactivity data and the multitask deep neural network algorithm. The obtained model yields excellent internal prediction capability with an auROC of 0.90 and consistently outperforms conventional single-task models on external tests, especially for kinases with insufficient activity data. Moreover, more rigorous experimental validations including 1410 kinase-compound pairs showed a high-quality average auROC of 0.75 and confirmed many novel predicted "off-target" activities. Given the verified generalizability, the model was further applied to various scenarios for depicting the kinome-wide selectivity and the association with certain diseases. Overall, the computational model enables us to create a comprehensive kinome interaction network for designing novel chemical modulators or drug repositioning and is of practical value for exploring previously less studied kinases.


Assuntos
Aprendizado Profundo , Polifarmacologia , Inibidores de Proteínas Quinases/química , Proteínas Quinases/química , Bases de Dados de Compostos Químicos , Conjuntos de Dados como Assunto , Descoberta de Drogas/métodos
10.
J Med Chem ; 63(16): 8749-8760, 2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31408336

RESUMO

Hunting for chemicals with favorable pharmacological, toxicological, and pharmacokinetic properties remains a formidable challenge for drug discovery. Deep learning provides us with powerful tools to build predictive models that are appropriate for the rising amounts of data, but the gap between what these neural networks learn and what human beings can comprehend is growing. Moreover, this gap may induce distrust and restrict deep learning applications in practice. Here, we introduce a new graph neural network architecture called Attentive FP for molecular representation that uses a graph attention mechanism to learn from relevant drug discovery data sets. We demonstrate that Attentive FP achieves state-of-the-art predictive performances on a variety of data sets and that what it learns is interpretable. The feature visualization for Attentive FP suggests that it automatically learns nonlocal intramolecular interactions from specified tasks, which can help us gain chemical insights directly from data beyond human perception.


Assuntos
Aprendizado Profundo , Descoberta de Drogas/métodos , Compostos Orgânicos/química , Bases de Dados de Compostos Químicos , Conjuntos de Dados como Assunto , Modelos Moleculares , Estudo de Prova de Conceito , Solubilidade
11.
Front Pharmacol ; 10: 924, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31507420

RESUMO

Scoring functions play an important role in structure-based virtual screening. It has been widely accepted that target-specific scoring functions (TSSFs) may achieve better performance compared with universal scoring functions in actual drug research and development processes. A method that can effectively construct TSSFs will be of great value to drug design and discovery. In this work, we proposed a deep learning-based model named DeepScore to achieve this goal. DeepScore adopted the form of PMF scoring function to calculate protein-ligand binding affinity. However, different from PMF scoring function, in DeepScore, the score for each protein-ligand atom pair was calculated using a feedforward neural network. Our model significantly outperformed Glide Gscore on validation data set DUD-E. The average ROC-AUC on 102 targets was 0.98. We also combined Gscore and DeepScore together using a consensus method and put forward a consensus model named DeepScoreCS. The comparison results showed that DeepScore outperformed other machine learning-based TSSFs building methods. Furthermore, we presented a strategy to visualize the prediction of DeepScore. All of these results clearly demonstrated that DeepScore would be a useful model in constructing TSSFs and represented a novel way incorporating deep learning and drug design.

12.
J Med Chem ; 62(16): 7473-7488, 2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31335138

RESUMO

Alterations of fibroblast growth factor receptors (FGFRs) play key roles in numerous cancer progression and development, which makes FGFRs attractive targets in the cancer therapy. In the present study, based on a newly devised FGFR target-specific scoring function, a novel FGFR inhibitor hit was identified through virtual screening. Hit-to-lead optimization was then performed by integrating molecular docking and site-of-metabolism predictions with an array of in vitro evaluations and X-ray cocrystal structure determination, leading to a covalent FGFR inhibitor 15, which showed a highly selective and potent FGFR inhibition profile. Pharmacokinetic assessment, protein kinase profiling, and hERG inhibition evaluation were also conducted, and they confirmed the value of 15 as a lead for further investigation. Overall, this study exemplifies the importance of the integrative use of computational methods and experimental techniques in drug discovery.


Assuntos
Desenho de Fármacos , Descoberta de Drogas , Inibidores de Proteínas Quinases/farmacologia , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Sequência de Aminoácidos , Humanos , Cinética , Simulação de Acoplamento Molecular , Estrutura Molecular , Terapia de Alvo Molecular/métodos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Fosforilação/efeitos dos fármacos , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacocinética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
13.
Bioinformatics ; 35(24): 5354-5356, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31228181

RESUMO

MOTIVATION: The large-scale kinome-wide virtual profiling for small molecules is a daunting task by experimental and traditional in silico drug design approaches. Recent advances in deep learning algorithms have brought about new opportunities in promoting this process. RESULTS: KinomeX is an online platform to predict kinome-wide polypharmacology effect of small molecules based solely on their chemical structures. The prediction is made by a multi-task deep neural network model trained with over 140 000 bioactivity data points for 391 kinases. Extensive computational and experimental validations have been performed. Overall, KinomeX enables users to create a comprehensive kinome interaction network for designing novel chemical modulators, and is of practical value on exploring the previously less studied or untargeted kinases. AVAILABILITY AND IMPLEMENTATION: KinomeX is available at: https://kinome.dddc.ac.cn. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Polifarmacologia , Algoritmos , Desenho de Fármacos , Software
14.
Sci China Life Sci ; 61(10): 1191-1204, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30054833

RESUMO

Thanks to the fast improvement of the computing power and the rapid development of the computational chemistry and biology, the computer-aided drug design techniques have been successfully applied in almost every stage of the drug discovery and development pipeline to speed up the process of research and reduce the cost and risk related to preclinical and clinical trials. Owing to the development of machine learning theory and the accumulation of pharmacological data, the artificial intelligence (AI) technology, as a powerful data mining tool, has cut a figure in various fields of the drug design, such as virtual screening, activity scoring, quantitative structure-activity relationship (QSAR) analysis, de novo drug design, and in silico evaluation of absorption, distribution, metabolism, excretion and toxicity (ADME/T) properties. Although it is still challenging to provide a physical explanation of the AI-based models, it indeed has been acting as a great power to help manipulating the drug discovery through the versatile frameworks. Recently, due to the strong generalization ability and powerful feature extraction capability, deep learning methods have been employed in predicting the molecular properties as well as generating the desired molecules, which will further promote the application of AI technologies in the field of drug design.


Assuntos
Inteligência Artificial , Desenho Assistido por Computador , Desenho de Fármacos , Descoberta de Drogas/métodos , Biologia Computacional/métodos , Simulação por Computador , Humanos , Preparações Farmacêuticas/administração & dosagem , Preparações Farmacêuticas/química , Preparações Farmacêuticas/metabolismo , Farmacocinética , Relação Estrutura-Atividade
15.
Q Rev Biophys ; 48(4): 488-515, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26328949

RESUMO

In recent decades, in silico absorption, distribution, metabolism, excretion (ADME), and toxicity (T) modelling as a tool for rational drug design has received considerable attention from pharmaceutical scientists, and various ADME/T-related prediction models have been reported. The high-throughput and low-cost nature of these models permits a more streamlined drug development process in which the identification of hits or their structural optimization can be guided based on a parallel investigation of bioavailability and safety, along with activity. However, the effectiveness of these tools is highly dependent on their capacity to cope with needs at different stages, e.g. their use in candidate selection has been limited due to their lack of the required predictability. For some events or endpoints involving more complex mechanisms, the current in silico approaches still need further improvement. In this review, we will briefly introduce the development of in silico models for some physicochemical parameters, ADME properties and toxicity evaluation, with an emphasis on the modelling approaches thereof, their application in drug discovery, and the potential merits or deficiencies of these models. Finally, the outlook for future ADME/T modelling based on big data analysis and systems sciences will be discussed.


Assuntos
Disponibilidade Biológica , Desenho de Fármacos , Animais , Sítios de Ligação , Transporte Biológico , Barreira Hematoencefálica , Cristalografia por Raios X , Descoberta de Drogas , Humanos , Intestinos/patologia , Ligantes , Camundongos , Modelos Biológicos , Conformação Molecular , Ligação Proteica , Relação Quantitativa Estrutura-Atividade , Solubilidade , Termodinâmica
16.
Yi Chuan ; 36(9): 857-63, 2014 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-25252302

RESUMO

Cardiovascular disease (CVD) has become an increased risk to human health, and the abnormal cholesterol metabolism will increase the risk of developing CVD. Along with the development of high-throughput sequencing technology and population genomics, the scanning for genes or mutations related to complex traits (or diseases) has been greatly promoted. Also, it becomes possible to explore the genetic mechanism of cholesterol metabolism. In this review, we summarize the progress of molecular genetic studies of cholesterol metabolism, based on the results of traditional genetic method and GWAS screening. Finally, the functional background of abnormal cholesterol metabolism was explored by pathway enrichment analysis. All these analyses will contribute to a better understanding of cholesterol molecular mechanism, and will also provide clues for prevention and treatment of cholesterol metabolism disorders.


Assuntos
Doenças Cardiovasculares/genética , Doenças Cardiovasculares/metabolismo , Colesterol/metabolismo , Animais , Doenças Cardiovasculares/epidemiologia , Ligação Genética , Estudo de Associação Genômica Ampla , Humanos , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA