Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Brief Bioinform ; 25(4)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-39007594

RESUMO

Artificial intelligence (AI)-driven methods can vastly improve the historically costly drug design process, with various generative models already in widespread use. Generative models for de novo drug design, in particular, focus on the creation of novel biological compounds entirely from scratch, representing a promising future direction. Rapid development in the field, combined with the inherent complexity of the drug design process, creates a difficult landscape for new researchers to enter. In this survey, we organize de novo drug design into two overarching themes: small molecule and protein generation. Within each theme, we identify a variety of subtasks and applications, highlighting important datasets, benchmarks, and model architectures and comparing the performance of top models. We take a broad approach to AI-driven drug design, allowing for both micro-level comparisons of various methods within each subtask and macro-level observations across different fields. We discuss parallel challenges and approaches between the two applications and highlight future directions for AI-driven de novo drug design as a whole. An organized repository of all covered sources is available at https://github.com/gersteinlab/GenAI4Drug.


Assuntos
Inteligência Artificial , Desenho de Fármacos , Proteínas , Humanos , Biologia Computacional/métodos , Proteínas/química
2.
Brief Bioinform ; 25(1)2023 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-38040493

RESUMO

Designing 3D molecules with high binding affinity for specific protein targets is crucial in drug design. One challenge is that the atomic interaction between molecules and proteins in 3D space has to be taken into account. However, the existing target-aware methods solely model the joint distribution between the molecules and proteins, disregarding the binding affinities between them, which leads to limited performance. In this paper, we propose an explainable diffusion model to generate molecules that can be bound to a given protein target with high affinity. Our method explicitly incorporates the chemical knowledge of protein-ligand binding affinity into the diffusion model, and uses the knowledge to guide the denoising process towards the direction of high binding affinity. Specifically, an SE(3)-invariant expert network is developed to fit the Vina scoring functions and jointly trained with the denoising network, while the domain knowledge is distilled and conveyed from Vina functions to the expert network. An effective guidance is proposed on both continuous atom coordinates and discrete atom types by taking advantages of the gradient of the expert network. Experiments on the benchmark CrossDocked2020 demonstrate the superiority of our method. Additionally, an atom-level explanation of the generated molecules is provided, and the connections with the domain knowledge are established.


Assuntos
Desenho de Fármacos , Proteínas , Proteínas/química , Ligação Proteica , Ligantes
3.
Brief Bioinform ; 25(1)2023 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-38189543

RESUMO

Recently, attention mechanism and derived models have gained significant traction in drug development due to their outstanding performance and interpretability in handling complex data structures. This review offers an in-depth exploration of the principles underlying attention-based models and their advantages in drug discovery. We further elaborate on their applications in various aspects of drug development, from molecular screening and target binding to property prediction and molecule generation. Finally, we discuss the current challenges faced in the application of attention mechanisms and Artificial Intelligence technologies, including data quality, model interpretability and computational resource constraints, along with future directions for research. Given the accelerating pace of technological advancement, we believe that attention-based models will have an increasingly prominent role in future drug discovery. We anticipate that these models will usher in revolutionary breakthroughs in the pharmaceutical domain, significantly accelerating the pace of drug development.


Assuntos
Inteligência Artificial , Descoberta de Drogas , Desenvolvimento de Medicamentos , Confiabilidade dos Dados
4.
J Comput Chem ; 45(22): 1886-1898, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-38698628

RESUMO

Reinforcement learning (RL) has been applied to various domains in computational chemistry and has found wide-spread success. In this review, we first motivate the application of RL to chemistry and list some broad application domains, for example, molecule generation, geometry optimization, and retrosynthetic pathway search. We set up some of the formalism associated with reinforcement learning that should help the reader translate their chemistry problems into a form where RL can be used to solve them. We then discuss the solution formulations and algorithms proposed in recent literature for these problems, the advantages of one over the other, together with the necessary details of the RL algorithms they employ. This article should help the reader understand the state of RL applications in chemistry, learn about some relevant actively-researched open problems, gain insight into how RL can be used to approach them and hopefully inspire innovative RL applications in Chemistry.

5.
Brief Bioinform ; 23(2)2022 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-35039853

RESUMO

Deep learning shortens the cycle of the drug discovery for its success in extracting features of molecules and proteins. Generating new molecules with deep learning methods could enlarge the molecule space and obtain molecules with specific properties. However, it is also a challenging task considering that the connections between atoms are constrained by chemical rules. Aiming at generating and optimizing new valid molecules, this article proposed Molecular Substructure Tree Generative Model, in which the molecule is generated by adding substructure gradually. The proposed model is based on the Variational Auto-Encoder architecture, which uses the encoder to map molecules to the latent vector space, and then builds an autoregressive generative model as a decoder to generate new molecules from Gaussian distribution. At the same time, for the molecular optimization task, a molecular optimization model based on CycleGAN was constructed. Experiments showed that the model could generate valid and novel molecules, and the optimized model effectively improves the molecular properties.


Assuntos
Desenho de Fármacos , Modelos Moleculares , Descoberta de Drogas
6.
Brief Bioinform ; 23(6)2022 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-36184256

RESUMO

Fentanyl and its analogues are psychoactive substances and the concern of fentanyl abuse has been existed in decades. Because the structure of fentanyl is easy to be modified, criminals may synthesize new fentanyl analogues to avoid supervision. The drug supervision is based on the structure matching to the database and too few kinds of fentanyl analogues are included in the database, so it is necessary to find out more potential fentanyl analogues and expand the sample space of fentanyl analogues. In this study, we introduced two deep generative models (SeqGAN and MolGPT) to generate potential fentanyl analogues, and a total of 11 041 valid molecules were obtained. The results showed that not only can we generate molecules with similar property distribution of original data, but the generated molecules also contain potential fentanyl analogues that are not pretty similar to any of original data. Ten molecules based on the rules of fentanyl analogues were selected for NMR, MS and IR validation. The results indicated that these molecules are all unreported fentanyl analogues. Furthermore, this study is the first to apply the deep learning to the generation of fentanyl analogues, greatly expands the exploring space of fentanyl analogues and provides help for the supervision of fentanyl.


Assuntos
Aprendizado Profundo , Fentanila , Fentanila/química , Analgésicos Opioides/química , Espectroscopia de Ressonância Magnética , Gerenciamento de Dados
7.
Brief Bioinform ; 23(1)2022 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-34734228

RESUMO

Artificial intelligence (AI) has been transforming the practice of drug discovery in the past decade. Various AI techniques have been used in many drug discovery applications, such as virtual screening and drug design. In this survey, we first give an overview on drug discovery and discuss related applications, which can be reduced to two major tasks, i.e. molecular property prediction and molecule generation. We then present common data resources, molecule representations and benchmark platforms. As a major part of the survey, AI techniques are dissected into model architectures and learning paradigms. To reflect the technical development of AI in drug discovery over the years, the surveyed works are organized chronologically. We expect that this survey provides a comprehensive review on AI in drug discovery. We also provide a GitHub repository with a collection of papers (and codes, if applicable) as a learning resource, which is regularly updated.


Assuntos
Inteligência Artificial , Descoberta de Drogas , Desenho de Fármacos , Descoberta de Drogas/métodos
8.
Molecules ; 29(7)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38611779

RESUMO

Drug discovery involves a crucial step of optimizing molecules with the desired structural groups. In the domain of computer-aided drug discovery, deep learning has emerged as a prominent technique in molecular modeling. Deep generative models, based on deep learning, play a crucial role in generating novel molecules when optimizing molecules. However, many existing molecular generative models have limitations as they solely process input information in a forward way. To overcome this limitation, we propose an improved generative model called BD-CycleGAN, which incorporates BiLSTM (bidirectional long short-term memory) and Mol-CycleGAN (molecular cycle generative adversarial network) to preserve the information of molecular input. To evaluate the proposed model, we assess its performance by analyzing the structural distribution and evaluation matrices of generated molecules in the process of structural transformation. The results demonstrate that the BD-CycleGAN model achieves a higher success rate and exhibits increased diversity in molecular generation. Furthermore, we demonstrate its application in molecular docking, where it successfully increases the docking score for the generated molecules. The proposed BD-CycleGAN architecture harnesses the power of deep learning to facilitate the generation of molecules with desired structural features, thus offering promising advancements in the field of drug discovery processes.


Assuntos
Fármacos Anti-HIV , Simulação de Acoplamento Molecular , Descoberta de Drogas , Hidrolases , Memória de Longo Prazo
9.
Int J Mol Sci ; 24(2)2023 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-36674658

RESUMO

Recent years have seen tremendous success in the design of novel drug molecules through deep generative models. Nevertheless, existing methods only generate drug-like molecules, which require additional structural optimization to be developed into actual drugs. In this study, a deep learning method for generating target-specific ligands was proposed. This method is useful when the dataset for target-specific ligands is limited. Deep learning methods can extract and learn features (representations) in a data-driven way with little or no human participation. Generative pretraining (GPT) was used to extract the contextual features of the molecule. Three different protein-encoding methods were used to extract the physicochemical properties and amino acid information of the target protein. Protein-encoding and molecular sequence information are combined to guide molecule generation. Transfer learning was used to fine-tune the pretrained model to generate molecules with better binding ability to the target protein. The model was validated using three different targets. The docking results show that our model is capable of generating new molecules with higher docking scores for the target proteins.


Assuntos
Desenho de Fármacos , Proteínas , Estrutura Molecular , Proteínas/química , Aminoácidos , Ligantes , Aprendizado de Máquina
10.
Sci Technol Adv Mater ; 23(1): 352-360, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35693890

RESUMO

Recently, artificial intelligence (AI)-enabled de novo molecular generators (DNMGs) have automated molecular design based on data-driven or simulation-based property estimates. In some domains like the game of Go where AI surpassed human intelligence, humans are trying to learn from AI about the best strategy of the game. To understand DNMG's strategy of molecule optimization, we propose an algorithm called characteristic functional group monitoring (CFGM). Given a time series of generated molecules, CFGM monitors statistically enriched functional groups in comparison to the training data. In the task of absorption wavelength maximization of pure organic molecules (consisting of H, C, N, and O), we successfully identified a strategic change from diketone and aniline derivatives to quinone derivatives. In addition, CFGM led us to a hypothesis that 1,2-quinone is an unconventional chromophore, which was verified with chemical synthesis. This study shows the possibility that human experts can learn from DNMGs to expand their ability to discover functional molecules.

11.
IEEE Trans Knowl Data Eng ; 34(11): 5459-5471, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36590707

RESUMO

The goal of molecular optimization is to generate molecules similar to a target molecule but with better chemical properties. Deep generative models have shown great success in molecule optimization. However, due to the iterative local generation process of deep generative models, the resulting molecules can significantly deviate from the input in molecular similarity and size, leading to poor chemical properties. The key issue here is that the existing deep generative models restrict their attention on substructure-level generation without considering the entire molecule as a whole. To address this challenge, we propose Molecule-Level Reward functions (MOLER) to encourage (1) the input and the generated molecule to be similar, and to ensure (2) the generated molecule has a similar size to the input. The proposed method can be combined with various deep generative models. Policy gradient technique is introduced to optimize reward-based objectives with small computational overhead. Empirical studies show that MOLER achieves up to 20.2% relative improvement in success rate over the best baseline method on several properties, including QED, DRD2 and LogP.

12.
Sci Technol Adv Mater ; 21(1): 552-561, 2020 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-32939179

RESUMO

Nuclear magnetic resonance (NMR) spectroscopy is an effective tool for identifying molecules in a sample. Although many previously observed NMR spectra are accumulated in public databases, they cover only a tiny fraction of the chemical space, and molecule identification is typically accomplished manually based on expert knowledge. Herein, we propose NMR-TS, a machine-learning-based python library, to automatically identify a molecule from its NMR spectrum. NMR-TS discovers candidate molecules whose NMR spectra match the target spectrum by using deep learning and density functional theory (DFT)-computed spectra. As a proof-of-concept, we identify prototypical metabolites from their computed spectra. After an average 5451 DFT runs for each spectrum, six of the nine molecules are identified correctly, and proximal molecules are obtained in the other cases. This encouraging result implies that de novo molecule generation can contribute to the fully automated identification of chemical structures. NMR-TS is available at https://github.com/tsudalab/NMR-TS.

13.
Drug Discov Today Technol ; 32-33: 55-63, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33386095

RESUMO

There has been a wave of generative models for molecules triggered by advances in the field of Deep Learning. These generative models are often used to optimize chemical compounds towards particular properties or a desired biological activity. The evaluation of generative models remains challenging and suggested performance metrics or scoring functions often do not cover all relevant aspects of drug design projects. In this work, we highlight some unintended failure modes in molecular generation and optimization and how these evade detection by current performance metrics.


Assuntos
Descoberta de Drogas , Modelos Moleculares , Humanos
14.
Comput Biol Med ; 171: 108073, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38359660

RESUMO

Large language models have made significant strides in natural language processing, enabling innovative applications in molecular science by processing textual representations of molecules. However, most existing language models cannot capture the rich information with complex molecular structures or images. In this paper, we introduce GIT-Mol, a multi-modal large language model that integrates the Graph, Image, and Text information. To facilitate the integration of multi-modal molecular data, we propose GIT-Former, a novel architecture that is capable of aligning all modalities into a unified latent space. We achieve a 5%-10% accuracy increase in properties prediction and a 20.2% boost in molecule generation validity compared to the baselines. With the any-to-language molecular translation strategy, our model has the potential to perform more downstream tasks, such as compound name recognition and chemical reaction prediction.


Assuntos
Idioma , Processamento de Linguagem Natural
15.
Interdiscip Sci ; 2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38683279

RESUMO

The structures of fentanyl and its analogues are easy to be modified and few types have been included in database so far, which allow criminals to avoid the supervision of relevant departments. This paper introduces a molecular graph-based transformer model, which is combined with a data augmentation method based on substructure replacement to generate novel fentanyl analogues. 140,000 molecules were generated, and after a set of screening, 36,799 potential fentanyl analogues were finally obtained. We calculated the molecular properties of 36,799 potential fentanyl analogues. The results showed that the model could learn some properties of original fentanyl molecules. We compared the generated molecules from transformer model and data augmentation method based on substructure replacement with those generated by the other two molecular generation models based on deep learning, and found that the model in this paper can generate more novel potential fentanyl analogues. Finally, the findings of the paper indicate that transformer model based on molecular graph helps us explore the structure of potential fentanyl molecules as well as understand distribution of original molecules of fentanyl.

16.
J Cheminform ; 16(1): 64, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38816825

RESUMO

Generative models are undergoing rapid research and application to de novo drug design. To facilitate their application and evaluation, we present MolScore. MolScore already contains many drug-design-relevant scoring functions commonly used in benchmarks such as, molecular similarity, molecular docking, predictive models, synthesizability, and more. In addition, providing performance metrics to evaluate generative model performance based on the chemistry generated. With this unification of functionality, MolScore re-implements commonly used benchmarks in the field (such as GuacaMol, MOSES, and MolOpt). Moreover, new benchmarks can be created trivially. We demonstrate this by testing a chemical language model with reinforcement learning on three new tasks of increasing complexity related to the design of 5-HT2a ligands that utilise either molecular descriptors, 266 pre-trained QSAR models, or dual molecular docking. Lastly, MolScore can be integrated into an existing Python script with just three lines of code. This framework is a step towards unifying generative model application and evaluation as applied to drug design for both practitioners and researchers. The framework can be found on GitHub and downloaded directly from the Python Package Index.Scientific ContributionMolScore is an open-source platform to facilitate generative molecular design and evaluation thereof for application in drug design. This platform takes important steps towards unifying existing benchmarks, providing a platform to share new benchmarks, and improves customisation, flexibility and usability for practitioners over existing solutions.

17.
Brief Funct Genomics ; 2024 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-38582610

RESUMO

Generative molecular models generate novel molecules with desired properties by searching chemical space. Traditional combinatorial optimization methods, such as genetic algorithms, have demonstrated superior performance in various molecular optimization tasks. However, these methods do not utilize docking simulation to inform the design process, and heavy dependence on the quality and quantity of available data, as well as require additional structural optimization to become candidate drugs. To address this limitation, we propose a novel model named DockingGA that combines Transformer neural networks and genetic algorithms to generate molecules with better binding affinity for specific targets. In order to generate high quality molecules, we chose the Self-referencing Chemical Structure Strings to represent the molecule and optimize the binding affinity of the molecules to different targets. Compared to other baseline models, DockingGA proves to be the optimal model in all docking results for the top 1, 10 and 100 molecules, while maintaining 100% novelty. Furthermore, the distribution of physicochemical properties demonstrates the ability of DockingGA to generate molecules with favorable and appropriate properties. This innovation creates new opportunities for the application of generative models in practical drug discovery.

18.
Acta Pharm Sin B ; 14(7): 3086-3109, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39027234

RESUMO

Multifunctional therapeutics have emerged as a solution to the constraints imposed by drugs with singular or insufficient therapeutic effects. The primary challenge is to integrate diverse pharmacophores within a single-molecule framework. To address this, we introduced DeepSA, a novel edit-based generative framework that utilizes deep simulated annealing for the modification of articaine, a well-known local anesthetic. DeepSA integrates deep neural networks into metaheuristics, effectively constraining molecular space during compound generation. This framework employs a sophisticated objective function that accounts for scaffold preservation, anti-inflammatory properties, and covalent constraints. Through a sequence of local editing to navigate the molecular space, DeepSA successfully identified AT-17, a derivative exhibiting potent analgesic properties and significant anti-inflammatory activity in various animal models. Mechanistic insights into AT-17 revealed its dual mode of action: selective inhibition of NaV1.7 and 1.8 channels, contributing to its prolonged local anesthetic effects, and suppression of inflammatory mediators via modulation of the NLRP3 inflammasome pathway. These findings not only highlight the efficacy of AT-17 as a multifunctional drug candidate but also highlight the potential of DeepSA in facilitating AI-enhanced drug discovery, particularly within stringent chemical constraints.

19.
Comput Biol Med ; 180: 108865, 2024 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-39067153

RESUMO

Designing drugs capable of binding to the structure of target proteins for treating diseases is essential in drug development. Recent remarkable advancements in geometric deep learning have led to unprecedented progress in three-dimensional (3D) generation of ligands that can bind to the protein pocket. However, most existing methods primarily focus on modeling the geometric information of ligands in 3D space. Consequently, these methods fail to consider that the binding of proteins and ligands is a phenomenon driven by intrinsic physicochemical principles. Motivated by this understanding, we propose PIDiff, a model for generating molecules by accounting in the physicochemical principles of protein-ligand binding. Our model learns not only the structural information of proteins and ligands but also to minimize the binding free energy between them. To evaluate the proposed model, we introduce an experimental framework that surpasses traditional assessment methods by encompassing various essential aspects for the practical application of generative models to actual drug development. The results confirm that our model outperforms baseline models on the CrossDocked2020 benchmark dataset, demonstrating its superiority. Through diverse experiments, we have illustrated the promising potential of the proposed model in practical drug development.

20.
J Cheminform ; 16(1): 77, 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38965600

RESUMO

SMILES-based generative models are amongst the most robust and successful recent methods used to augment drug design. They are typically used for complete de novo generation, however, scaffold decoration and fragment linking applications are sometimes desirable which requires a different grammar, architecture, training dataset and therefore, re-training of a new model. In this work, we describe a simple procedure to conduct constrained molecule generation with a SMILES-based generative model to extend applicability to scaffold decoration and fragment linking by providing SMILES prompts, without the need for re-training. In combination with reinforcement learning, we show that pre-trained, decoder-only models adapt to these applications quickly and can further optimize molecule generation towards a specified objective. We compare the performance of this approach to a variety of orthogonal approaches and show that performance is comparable or better. For convenience, we provide an easy-to-use python package to facilitate model sampling which can be found on GitHub and the Python Package Index.Scientific contributionThis novel method extends an autoregressive chemical language model to scaffold decoration and fragment linking scenarios. This doesn't require re-training, the use of a bespoke grammar, or curation of a custom dataset, as commonly required by other approaches.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA