Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Brief Bioinform ; 25(3)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38701419

RESUMEN

It is a vital step to recognize cyanobacteria promoters on a genome-wide scale. Computational methods are promising to assist in difficult biological identification. When building recognition models, these methods rely on non-promoter generation to cope with the lack of real non-promoters. Nevertheless, the factitious significant difference between promoters and non-promoters causes over-optimistic prediction. Moreover, designed for E. coli or B. subtilis, existing methods cannot uncover novel, distinct motifs among cyanobacterial promoters. To address these issues, this work first proposes a novel non-promoter generation strategy called phantom sampling, which can eliminate the factitious difference between promoters and generated non-promoters. Furthermore, it elaborates a novel promoter prediction model based on the Siamese network (SiamProm), which can amplify the hidden difference between promoters and non-promoters through a joint characterization of global associations, upstream and downstream contexts, and neighboring associations w.r.t. k-mer tokens. The comparison with state-of-the-art methods demonstrates the superiority of our phantom sampling and SiamProm. Both comprehensive ablation studies and feature space illustrations also validate the effectiveness of the Siamese network and its components. More importantly, SiamProm, upon our phantom sampling, finds a novel cyanobacterial promoter motif ('GCGATCGC'), which is palindrome-patterned, content-conserved, but position-shifted.


Asunto(s)
Cianobacterias , Regiones Promotoras Genéticas , Cianobacterias/genética , Biología Computacional/métodos , Algoritmos
2.
Brief Bioinform ; 24(1)2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36642408

RESUMEN

Current machine learning-based methods have achieved inspiring predictions in the scenarios of mono-type and multi-type drug-drug interactions (DDIs), but they all ignore enhancive and depressive pharmacological changes triggered by DDIs. In addition, these pharmacological changes are asymmetric since the roles of two drugs in an interaction are different. More importantly, these pharmacological changes imply significant topological patterns among DDIs. To address the above issues, we first leverage Balance theory and Status theory in social networks to reveal the topological patterns among directed pharmacological DDIs, which are modeled as a signed and directed network. Then, we design a novel graph representation learning model named SGRL-DDI (social theory-enhanced graph representation learning for DDI) to realize the multitask prediction of DDIs. SGRL-DDI model can capture the task-joint information by integrating relation graph convolutional networks with Balance and Status patterns. Moreover, we utilize task-specific deep neural networks to perform two tasks, including the prediction of enhancive/depressive DDIs and the prediction of directed DDIs. Based on DDI entries collected from DrugBank, the superiority of our model is demonstrated by the comparison with other state-of-the-art methods. Furthermore, the ablation study verifies that Balance and Status patterns help characterize directed pharmacological DDIs, and that the joint of two tasks provides better DDI representations than individual tasks. Last, we demonstrate the practical effectiveness of our model by a version-dependent test, where 88.47 and 81.38% DDI out of newly added entries provided by the latest release of DrugBank are validated in two predicting tasks respectively.


Asunto(s)
Aprendizaje Automático , Redes Neurales de la Computación , Interacciones Farmacológicas
3.
Brief Bioinform ; 24(4)2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37401373

RESUMEN

Recent advances and achievements of artificial intelligence (AI) as well as deep and graph learning models have established their usefulness in biomedical applications, especially in drug-drug interactions (DDIs). DDIs refer to a change in the effect of one drug to the presence of another drug in the human body, which plays an essential role in drug discovery and clinical research. DDIs prediction through traditional clinical trials and experiments is an expensive and time-consuming process. To correctly apply the advanced AI and deep learning, the developer and user meet various challenges such as the availability and encoding of data resources, and the design of computational methods. This review summarizes chemical structure based, network based, natural language processing based and hybrid methods, providing an updated and accessible guide to the broad researchers and development community with different domain knowledge. We introduce widely used molecular representation and describe the theoretical frameworks of graph neural network models for representing molecular structures. We present the advantages and disadvantages of deep and graph learning methods by performing comparative experiments. We discuss the potential technical challenges and highlight future directions of deep and graph learning models for accelerating DDIs prediction.


Asunto(s)
Inteligencia Artificial , Redes Neurales de la Computación , Humanos , Interacciones Farmacológicas , Procesamiento de Lenguaje Natural , Descubrimiento de Drogas
4.
Methods ; 222: 51-56, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38184219

RESUMEN

The interaction between human microbes and drugs can significantly impact human physiological functions. It is crucial to identify potential microbe-drug associations (MDAs) before drug administration. However, conventional biological experiments to predict MDAs are plagued by drawbacks such as time-consuming, high costs, and potential risks. On the contrary, computational approaches can speed up the screening of MDAs at a low cost. Most computational models usually use a drug similarity matrix as the initial feature representation of drugs and stack the graph neural network layers to extract the features of network nodes. However, different calculation methods result in distinct similarity matrices, and message passing in graph neural networks (GNNs) induces phenomena of over-smoothing and over-squashing, thereby impacting the performance of the model. To address these issues, we proposed a novel graph representation learning model, dual-modal graph learning for microbe-drug association prediction (DMGL-MDA). It comprises a dual-modal embedding module, a bipartite graph network embedding module, and a predictor module. To assess the performance of DMGL-MDA, we compared it against state-of-the-art methods using two benchmark datasets. Through cross-validation, we illustrated the superiority of DMGL-MDA. Furthermore, we conducted ablation experiments and case studies to validate the effective performance of the model.


Asunto(s)
Benchmarking , Redes Neurales de la Computación , Humanos , Proyectos de Investigación
5.
Brief Bioinform ; 23(3)2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35470854

RESUMEN

It is tough to detect unexpected drug-drug interactions (DDIs) in poly-drug treatments because of high costs and clinical limitations. Computational approaches, such as deep learning-based approaches, are promising to screen potential DDIs among numerous drug pairs. Nevertheless, existing approaches neglect the asymmetric roles of two drugs in interaction. Such an asymmetry is crucial to poly-drug treatments since it determines drug priority in co-prescription. This paper designs a directed graph attention network (DGAT-DDI) to predict asymmetric DDIs. First, its encoder learns the embeddings of the source role, the target role and the self-roles of a drug. The source role embedding represents how a drug influences other drugs in DDIs. In contrast, the target role embedding represents how it is influenced by others. The self-role embedding encodes its chemical structure in a role-specific manner. Besides, two role-specific items, aggressiveness and impressionability, capture how the number of interaction partners of a drug affects its interaction tendency. Furthermore, the predictor of DGAT-DDI discriminates direction-specific interactions by the combination between two proximities and the above two role-specific items. The proximities measure the similarity between source/target embeddings and self-role embeddings. In the designated experiments, the comparison with state-of-the-art deep learning models demonstrates the superiority of DGAT-DDI across a direction-specific predicting task and a direction-blinded predicting task. An ablation study reveals how well each component of DGAT-DDI contributes to its ability. Moreover, a case study of finding novel DDIs confirms its practical ability, where 7 out of the top 10 candidates are validated in DrugBank.


Asunto(s)
Interacciones Farmacológicas
6.
Brief Bioinform ; 23(1)2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-34695842

RESUMEN

Drug-drug interactions (DDIs) are interactions with adverse effects on the body, manifested when two or more incompatible drugs are taken together. They can be caused by the chemical compositions of the drugs involved. We introduce gated message passing neural network (GMPNN), a message passing neural network which learns chemical substructures with different sizes and shapes from the molecular graph representations of drugs for DDI prediction between a pair of drugs. In GMPNN, edges are considered as gates which control the flow of message passing, and therefore delimiting the substructures in a learnable way. The final DDI prediction between a drug pair is based on the interactions between pairs of their (learned) substructures, each pair weighted by a relevance score to the final DDI prediction output. Our proposed method GMPNN-CS (i.e. GMPNN + prediction module) is evaluated on two real-world datasets, with competitive results on one, and improved performance on the other compared with previous methods. Source code is freely available at https://github.com/kanz76/GMPNN-CS.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Programas Informáticos , Interacciones Farmacológicas , Humanos , Redes Neurales de la Computación
7.
Bioinformatics ; 39(39 Suppl 1): i326-i336, 2023 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-37387157

RESUMEN

MOTIVATION: Deep learning-based molecule generation becomes a new paradigm of de novo molecule design since it enables fast and directional exploration in the vast chemical space. However, it is still an open issue to generate molecules, which bind to specific proteins with high-binding affinities while owning desired drug-like physicochemical properties. RESULTS: To address these issues, we elaborate a novel framework for controllable protein-oriented molecule generation, named CProMG, which contains a 3D protein embedding module, a dual-view protein encoder, a molecule embedding module, and a novel drug-like molecule decoder. Based on fusing the hierarchical views of proteins, it enhances the representation of protein binding pockets significantly by associating amino acid residues with their comprising atoms. Through jointly embedding molecule sequences, their drug-like properties, and binding affinities w.r.t. proteins, it autoregressively generates novel molecules having specific properties in a controllable manner by measuring the proximity of molecule tokens to protein residues and atoms. The comparison with state-of-the-art deep generative methods demonstrates the superiority of our CProMG. Furthermore, the progressive control of properties demonstrates the effectiveness of CProMG when controlling binding affinity and drug-like properties. After that, the ablation studies reveal how its crucial components contribute to the model respectively, including hierarchical protein views, Laplacian position encoding as well as property control. Last, a case study w.r.t. protein illustrates the novelty of CProMG and the ability to capture crucial interactions between protein pockets and molecules. It's anticipated that this work can boost de novo molecule design. AVAILABILITY AND IMPLEMENTATION: The code and data underlying this article are freely available at https://github.com/lijianing0902/CProMG.


Asunto(s)
Aminoácidos , Aprendizaje Profundo , Ingeniería de Proteínas
8.
Bioinformatics ; 39(8)2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37572298

RESUMEN

MOTIVATION: Metabolic stability plays a crucial role in the early stages of drug discovery and development. Accurately modeling and predicting molecular metabolic stability has great potential for the efficient screening of drug candidates as well as the optimization of lead compounds. Considering wet-lab experiment is time-consuming, laborious, and expensive, in silico prediction of metabolic stability is an alternative choice. However, few computational methods have been developed to address this task. In addition, it remains a significant challenge to explain key functional groups determining metabolic stability. RESULTS: To address these issues, we develop a novel cross-modality graph contrastive learning model named CMMS-GCL for predicting the metabolic stability of drug candidates. In our framework, we design deep learning methods to extract features for molecules from two modality data, i.e. SMILES sequence and molecule graph. In particular, for the sequence data, we design a multihead attention BiGRU-based encoder to preserve the context of symbols to learn sequence representations of molecules. For the graph data, we propose a graph contrastive learning-based encoder to learn structure representations by effectively capturing the consistencies between local and global structures. We further exploit fully connected neural networks to combine the sequence and structure representations for model training. Extensive experimental results on two datasets demonstrate that our CMMS-GCL consistently outperforms seven state-of-the-art methods. Furthermore, a collection of case studies on sequence data and statistical analyses of the graph structure module strengthens the validation of the interpretability of crucial functional groups recognized by CMMS-GCL. Overall, CMMS-GCL can serve as an effective and interpretable tool for predicting metabolic stability, identifying critical functional groups, and thus facilitating the drug discovery process and lead compound optimization. AVAILABILITY AND IMPLEMENTATION: The code and data underlying this article are freely available at https://github.com/dubingxue/CMMS-GCL.


Asunto(s)
Descubrimiento de Drogas , Redes Neurales de la Computación , Proyectos de Investigación
9.
J Chem Inf Model ; 64(1): 96-109, 2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38132638

RESUMEN

Detecting drug-drug interactions (DDIs) is an essential step in drug development and drug administration. Given the shortcomings of current experimental methods, the machine learning (ML) approach has become a reliable alternative, attracting extensive attention from the academic and industrial fields. With the rapid development of computational science and the growing popularity of cross-disciplinary research, a large number of DDI prediction studies based on ML methods have been published in recent years. To give an insight into the current situation and future direction of DDI prediction research, we systemically review these studies from three aspects: (1) the classic DDI databases, mainly including databases of drugs, side effects, and DDI information; (2) commonly used drug attributes, which focus on chemical, biological, and phenotypic attributes for representing drugs; (3) popular ML approaches, such as shallow learning-based, deep learning-based, recommender system-based, and knowledge graph-based methods for DDI detection. For each section, related studies are described, summarized, and compared, respectively. In the end, we conclude the research status of DDI prediction based on ML methods and point out the existing issues, future challenges, potential opportunities, and subsequent research direction.


Asunto(s)
Bases del Conocimiento , Aprendizaje Automático , Interacciones Farmacológicas , Preparaciones Farmacéuticas , Bases de Datos Factuales
10.
Brief Bioinform ; 22(6)2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33951725

RESUMEN

A major concern with co-administration of different drugs is the high risk of interference between their mechanisms of action, known as adverse drug-drug interactions (DDIs), which can cause serious injuries to the organism. Although several computational methods have been proposed for identifying potential adverse DDIs, there is still room for improvement. Existing methods are not explicitly based on the knowledge that DDIs are fundamentally caused by chemical substructure interactions instead of whole drugs' chemical structures. Furthermore, most of existing methods rely on manually engineered molecular representation, which is limited by the domain expert's knowledge.We propose substructure-substructure interaction-drug-drug interaction (SSI-DDI), a deep learning framework, which operates directly on the raw molecular graph representations of drugs for richer feature extraction; and, most importantly, breaks the DDI prediction task between two drugs down to identifying pairwise interactions between their respective substructures. SSI-DDI is evaluated on real-world data and improves DDI prediction performance compared to state-of-the-art methods. Source code is freely available at https://github.com/kanz76/SSI-DDI.


Asunto(s)
Biología Computacional , Interacciones Farmacológicas , Redes Neurales de la Computación , Programas Informáticos , Relación Estructura-Actividad
11.
Bioinformatics ; 38(Suppl 1): i325-i332, 2022 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-35758801

RESUMEN

MOTIVATION: During lead compound optimization, it is crucial to identify pathways where a drug-like compound is metabolized. Recently, machine learning-based methods have achieved inspiring progress to predict potential metabolic pathways for drug-like compounds. However, they neglect the knowledge that metabolic pathways are dependent on each other. Moreover, they are inadequate to elucidate why compounds participate in specific pathways. RESULTS: To address these issues, we propose a novel Multi-Label Graph Learning framework of Metabolic Pathway prediction boosted by pathway interdependence, called MLGL-MP, which contains a compound encoder, a pathway encoder and a multi-label predictor. The compound encoder learns compound embedding representations by graph neural networks. After constructing a pathway dependence graph by re-trained word embeddings and pathway co-occurrences, the pathway encoder learns pathway embeddings by graph convolutional networks. Moreover, after adapting the compound embedding space into the pathway embedding space, the multi-label predictor measures the proximity of two spaces to discriminate which pathways a compound participates in. The comparison with state-of-the-art methods on KEGG pathways demonstrates the superiority of our MLGL-MP. Also, the ablation studies reveal how its three components contribute to the model, including the pathway dependence, the adapter between compound embeddings and pathway embeddings, as well as the pre-training strategy. Furthermore, a case study illustrates the interpretability of MLGL-MP by indicating crucial substructures in a compound, which are significantly associated with the attending metabolic pathways. It is anticipated that this work can boost metabolic pathway predictions in drug discovery. AVAILABILITY AND IMPLEMENTATION: The code and data underlying this article are freely available at https://github.com/dubingxue/MLGL-MP.


Asunto(s)
Aprendizaje Automático , Redes Neurales de la Computación , Descubrimiento de Drogas , Redes y Vías Metabólicas , Programas Informáticos
12.
BMC Bioinformatics ; 23(1): 75, 2022 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-35172712

RESUMEN

BACKGROUND: Prediction of drug-drug interactions (DDIs) can reveal potential adverse pharmacological reactions between drugs in co-medication. Various methods have been proposed to address this issue. Most of them focus on the traditional link prediction between drugs, however, they ignore the cold-start scenario, which requires the prediction between known drugs having approved DDIs and new drugs having no DDI. Moreover, they're restricted to infer whether DDIs occur, but are not able to deduce diverse DDI types, which are important in clinics. RESULTS: In this paper, we propose a cold start prediction model for both single-type and multiple-type drug-drug interactions, referred to as CSMDDI. CSMDDI predict not only whether two drugs trigger pharmacological reactions but also what reaction types they induce in the cold start scenario. We implement several embedding methods in CSMDDI, including SVD, GAE, TransE, RESCAL and compare it with the state-of-the-art multi-type DDI prediction method DeepDDI and DDIMDL to verify the performance. The comparison shows that CSMDDI achieves a good performance of DDI prediction in the case of both the occurrence prediction and the multi-type reaction prediction in cold start scenario. CONCLUSIONS: Our approach is able to predict not only conventional binary DDIs but also what reaction types they induce in the cold start scenario. More importantly, it learns a mapping function who can bridge the drugs attributes to their network embeddings to predict DDIs. The main contribution of CSMDDI contains the development of a generalized framework to predict the single-type and multi-type of DDIs in the cold start scenario, as well as the implementations of several embedding models for both single-type and multi-type of DDIs. The dataset and source code can be accessed at https://github.com/itsosy/csmddi .


Asunto(s)
Preparaciones Farmacéuticas , Programas Informáticos , Interacciones Farmacológicas
13.
Anal Biochem ; 646: 114631, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35227661

RESUMEN

It is crucial to identify DDIs and explore their underlying mechanism (e.g., DDIs types) for polypharmacy safety. However, the detection of DDIs in assays is still time-consuming and costly, due to the need for experimental search over a large space of drug combinations. Thus, many computational methods have been developed to predict DDIs, most of them focusing on whether a drug interacts with another or not. And a few deep learning-based methods address a more realistic screening task for identifying various DDI types, but they assume a DDI only triggers one pharmacological effect, while a DDI can trigger more types of pharmacological effects. Thus, here we proposed a novel end-to-end deep learning-based method (called deepMDDI) for the Multi-label prediction of Drug-Drug Interactions. deepMDDI contains an encoder derived from relational graph convolutional networks and a tensor-like decoder to uniformly model interactions. deepMDDI is not only efficient for DDI transductive prediction, but also inductive prediction. The experimental results show that our model is superior to other state-of-the-art deep learning-based methods. We also validated the power of deepMDDI in the DDIs multi-label prediction and found several new valid DDIs in the case study. In conclusion, deepMDDI is beneficial to uncover the mechanism and regularity of DDIs.


Asunto(s)
Interacciones Farmacológicas
14.
BMC Bioinformatics ; 21(1): 419, 2020 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-32972364

RESUMEN

BACKGROUND: The treatment of complex diseases by taking multiple drugs becomes increasingly popular. However, drug-drug interactions (DDIs) may give rise to the risk of unanticipated adverse effects and even unknown toxicity. DDI detection in the wet lab is expensive and time-consuming. Thus, it is highly desired to develop the computational methods for predicting DDIs. Generally, most of the existing computational methods predict DDIs by extracting the chemical and biological features of drugs from diverse drug-related properties, however some drug properties are costly to obtain and not available in many cases. RESULTS: In this work, we presented a novel method (namely DPDDI) to predict DDIs by extracting the network structure features of drugs from DDI network with graph convolution network (GCN), and the deep neural network (DNN) model as a predictor. GCN learns the low-dimensional feature representations of drugs by capturing the topological relationship of drugs in DDI network. DNN predictor concatenates the latent feature vectors of any two drugs as the feature vector of the corresponding drug pairs to train a DNN for predicting the potential drug-drug interactions. Experiment results show that, the newly proposed DPDDI method outperforms four other state-of-the-art methods; the GCN-derived latent features include more DDI information than other features derived from chemical, biological or anatomical properties of drugs; and the concatenation feature aggregation operator is better than two other feature aggregation operators (i.e., inner product and summation). The results in case studies confirm that DPDDI achieves reasonable performance in predicting new DDIs. CONCLUSION: We proposed an effective and robust method DPDDI to predict the potential DDIs by utilizing the DDI network information without considering the drug properties (i.e., drug chemical and biological properties). The method should also be useful in other DDI-related scenarios, such as the detection of unexpected side effects, and the guidance of drug combination.


Asunto(s)
Interacciones Farmacológicas , Programas Informáticos , Bases de Datos como Asunto , Humanos , Redes Neurales de la Computación
15.
BMC Bioinformatics ; 19(Suppl 9): 281, 2018 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-30367598

RESUMEN

BACKGROUND: Human Microbiome Project reveals the significant mutualistic influence between human body and microbes living in it. Such an influence lead to an interesting phenomenon that many noninfectious diseases are closely associated with diverse microbes. However, the identification of microbe-noninfectious disease associations (MDAs) is still a challenging task, because of both the high cost and the limitation of microbe cultivation. Thus, there is a need to develop fast approaches to screen potential MDAs. The growing number of validated MDAs enables us to meet the demand in a new insight. Computational approaches, especially machine learning, are promising to predict MDA candidates rapidly among a large number of microbe-disease pairs with the advantage of no limitation on microbe cultivation. Nevertheless, a few computational efforts at predicting MDAs are made so far. RESULTS: In this paper, grouping a set of MDAs into a binary MDA matrix, we propose a novel predictive approach (BMCMDA) based on Binary Matrix Completion to predict potential MDAs. The proposed BMCMDA assumes that the incomplete observed MDA matrix is the summation of a latent parameterizing matrix and a noising matrix. It also assumes that the independently occurring subscripts of observed entries in the MDA matrix follows a binomial model. Adopting a standard mean-zero Gaussian distribution for the nosing matrix, we model the relationship between the parameterizing matrix and the MDA matrix under the observed microbe-disease pairs as a probit regression. With the recovered parameterizing matrix, BMCMDA deduces how likely a microbe would be associated with a particular disease. In the experiment under leave-one-out cross-validation, it exhibits the inspiring performance (AUC = 0.906, AUPR =0.526) and demonstrates its superiority by ~ 7% and ~ 5% improvements in terms of AUC and AUPR respectively in the comparison with the pioneering approach KATZHMDA. CONCLUSIONS: Our BMCMDA provides an effective approach for predicting MDAs and can be also extended to other similar predicting tasks of binary relationship (e.g. protein-protein interaction, drug-target interaction).


Asunto(s)
Algoritmos , Bacterias , Biología Computacional/métodos , Enfermedad , Microbiota , Modelos Biológicos , Fenómenos Fisiológicos Bacterianos , Interacciones Huésped-Patógeno , Humanos , Factores de Riesgo
16.
BMC Bioinformatics ; 19(Suppl 14): 411, 2018 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-30453924

RESUMEN

BACKGROUND: A significant number of adverse drug reactions is caused by unexpected Drug-drug interactions (DDIs). The identification of DDIs becomes crucial before the co-prescription of multiple drugs is made. Such a task in clinics or in drug discovery usually requires high costs and numerous limitations, while computational approaches are able to predict potential DDIs effectively by utilizing diverse drug attributes (e.g. side effects). Nevertheless, they're incapable when required to predict enhancive and degressive DDIs, which change increasingly and decreasingly the pharmacological behavior of interacting drugs respectively. The pharmacological change of DDIs is one of the most important factors when making a multi-drug prescription. RESULTS: In this work, we design a Triple Matrix Factorization-based Unified Framework (TMFUF) to address the above issue. By leveraging a group of side effect entries of drugs, TMFUF achieves the inspiring result (AUC = 0.842 and AUPR = 0.526) in the case of conventional DDI prediction under the traditional screening task. In the comparison with two state-of-the-art approaches, TMFUF demonstrates it superiority by ~ 7% and ~ 20% improvement in terms of AUC and AUPR respectively. More importantly, TMFUF shows its ability in the comprehensive DDI prediction under different screening tasks. Finally, a utilization TMFUF reveals the significant pairs of side effects, which contribute to form enhancive and degressive DDIs, for further clinical validation. CONCLUSIONS: The proposed TMFUF is first capable to predict both conventional binary DDIs and comprehensive DDIs such that it captures the pharmacological changes caused by DDIs. Furthermore, it provides a unified solution of DDI prediction for two screening scenarios, which involves newly given drugs having no prior interaction. Another advantage is its ability to indicate how significantly the pairs of drug features contribute to form DDIs.


Asunto(s)
Algoritmos , Interacciones Farmacológicas , Humanos , Análisis de los Mínimos Cuadrados , Curva ROC , Reproducibilidad de los Resultados
17.
BMC Bioinformatics ; 18(Suppl 12): 409, 2017 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-29072137

RESUMEN

BACKGROUND: Drug Combination is one of the effective approaches for treating complex diseases. However, determining combinative drug pairs in clinical trials is still costly. Thus, computational approaches are used to identify potential drug pairs in advance. Existing computational approaches have the following shortcomings: (i) the lack of an effective integration of heterogeneous features leads to a time-consuming training and even results in an over-fitted classifier; and (ii) the narrow consideration of predicting potential drug combinations only among known drugs having known combinations cannot meet the demand of realistic screenings, which pay more attention to potential combinative pairs among newly-coming drugs that have no approved combination with other drugs at all. RESULTS: In this paper, to tackle the above two problems, we propose a novel drug-driven approach for predicting potential combinative pairs on a large scale. We define four new features based on heterogeneous data and design an efficient fusion scheme to integrate these feature. Moreover importantly, we elaborate appropriate cross-validations towards realistic screening scenarios of drug combinations involving both known drugs and new drugs. In addition, we perform an extra investigation to show how each kind of heterogeneous features is related to combinative drug pairs. The investigation inspires the design of our approach. Experiments on real data demonstrate the effectiveness of our fusion scheme for integrating heterogeneous features and its predicting power in three scenarios of realistic screening. In terms of both AUC and AUPR, the prediction among known drugs achieves 0.954 and 0.821, that between known drugs and new drugs achieves 0.909 and 0.635, and that among new drugs achieves 0.809 and 0.592 respectively. CONCLUSIONS: Our approach provides not only an effective tool to integrate heterogeneous features but also the first tool to predict potential combinative pairs among new drugs.


Asunto(s)
Biología Computacional/métodos , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos , Bases de Datos como Asunto , Humanos
18.
BMC Bioinformatics ; 17 Suppl 8: 282, 2016 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-27585458

RESUMEN

BACKGROUND: There has been paid more and more attention to supervised classification models in the area of predicting drug-target interactions (DTIs). However, in terms of classification, unavoidable missing DTIs in data would cause three issues which have not yet been addressed appropriately by former approaches. Directly labeled as negatives (non-DTIs), missing DTIs increase the confusion of positives (DTIs) and negatives, aggravate the imbalance between few positives and many negatives, and are usually discriminated as highly-scored false positives, which influence the existing measures sharply. RESULTS: Under the framework of local classification model (LCM), this work focuses on the scenario of predicting how possibly a new drug interacts with known targets. To address the first two issues, two strategies, Spy and Super-target, are introduced accordingly and further integrated to form a two-layer LCM. In the bottom layer, Spy-based local classifiers for protein targets are built by positives, as well as reliable negatives identified among unlabeled drug-target pairs. In the top layer, regular local classifiers specific to super-targets are built with more positives generated by grouping similar targets and their interactions. Furthermore, to handle the third issue, an additional performance measure, Coverage, is presented for assessing DTI prediction. The experiments based on benchmark datasets are finally performed under five-fold cross validation of drugs to evaluate this approach. The main findings are concluded as follows. (1) Both two individual strategies and their combination are effective to missing DTIs, and the combination wins the best. (2) Having the advantages of less confusing decision boundary at the bottom layer and less biased decision boundary at the top layer, our two-layer LCM outperforms two former approaches. (3) Coverage is more robust to missing interactions than other measures and is able to evaluate how far one needs to go down the list of targets to cover all the proper targets of a drug. CONCLUSIONS: Proposing two strategies and one performance measure, this work has addressed the issues derived from missing interactions, which cause confusing and biased decision boundaries in classifiers, as well as the inappropriate measure of predicting performance, in the scenario of predicting interactions between new drugs and known targets.


Asunto(s)
Descubrimiento de Drogas , Interacciones Farmacológicas , Área Bajo la Curva , Humanos , Modelos Teóricos
19.
Methods ; 83: 98-104, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25957673

RESUMEN

Predicting drug-target interaction using computational approaches is an important step in drug discovery and repositioning. To predict whether there will be an interaction between a drug and a target, most existing methods identify similar drugs and targets in the database. The prediction is then made based on the known interactions of these drugs and targets. This idea is promising. However, there are two shortcomings that have not yet been addressed appropriately. Firstly, most of the methods only use 2D chemical structures and protein sequences to measure the similarity of drugs and targets respectively. However, this information may not fully capture the characteristics determining whether a drug will interact with a target. Secondly, there are very few known interactions, i.e. many interactions are "missing" in the database. Existing approaches are biased towards known interactions and have no good solutions to handle possibly missing interactions which affect the accuracy of the prediction. In this paper, we enhance the similarity measures to include non-structural (and non-sequence-based) information and introduce the concept of a "super-target" to handle the problem of possibly missing interactions. Based on evaluations on real data, we show that our similarity measure is better than the existing measures and our approach is able to achieve higher accuracy than the two best existing algorithms, WNN-GIP and KBMF2K. Our approach is available at http://web.hku.hk/∼liym1018/projects/drug/drug.html or http://www.bmlnwpu.org/us/tools/PredictingDTI_S2/METHODS.html.


Asunto(s)
Análisis por Conglomerados , Biología Computacional/métodos , Descubrimiento de Drogas/métodos , Genómica/métodos , Algoritmos , Inteligencia Artificial , Humanos , Preparaciones Farmacéuticas/química
20.
Comput Biol Med ; 178: 108699, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38870725

RESUMEN

Accurate prediction of drug-target binding affinity (DTA) plays a pivotal role in drug discovery and repositioning. Although deep learning methods are widely used in DTA prediction, two significant challenges persist: (i) how to effectively represent the complex structural information of proteins and drugs; (ii) how to precisely model the mutual interactions between protein binding sites and key drug substructures. To address these challenges, we propose a MSFFDTA (Multi-scale feature fusion for predicting drug target affinity) model, in which multi-scale encoders effectively capture multi-level structural information of drugs and proteins are designed. And then a Selective Cross Attention (SCA) mechanism is developed to filter out the trivial interactions between drug-protein substructure pairs and retain the important ones, which will make the proposed model better focusing on these key interactions and offering insights into their underlying mechanism. Experimental results on two benchmark datasets demonstrate that MSFFDTA is superior to several state-of-the-art methods across almost all comparison metrics. Finally, we provide the ablation and case studies with visualizations to verify the effectiveness and the interpretability of MSFFDTA. The source code is freely available at https://github.com/whitehat32/MSFF-DTA/.


Asunto(s)
Proteínas , Proteínas/química , Proteínas/metabolismo , Descubrimiento de Drogas/métodos , Aprendizaje Profundo , Preparaciones Farmacéuticas/metabolismo , Preparaciones Farmacéuticas/química , Humanos , Unión Proteica , Sitios de Unión , Biología Computacional/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA