Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Brief Bioinform ; 25(3)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38762789

RESUMEN

Identifying drug-target interactions (DTIs) holds significant importance in drug discovery and development, playing a crucial role in various areas such as virtual screening, drug repurposing and identification of potential drug side effects. However, existing methods commonly exploit only a single type of feature from drugs and targets, suffering from miscellaneous challenges such as high sparsity and cold-start problems. We propose a novel framework called MSI-DTI (Multi-Source Information-based Drug-Target Interaction Prediction) to enhance prediction performance, which obtains feature representations from different views by integrating biometric features and knowledge graph representations from multi-source information. Our approach involves constructing a Drug-Target Knowledge Graph (DTKG), obtaining multiple feature representations from diverse information sources for SMILES sequences and amino acid sequences, incorporating network features from DTKG and performing an effective multi-source information fusion. Subsequently, we employ a multi-head self-attention mechanism coupled with residual connections to capture higher-order interaction information between sparse features while preserving lower-order information. Experimental results on DTKG and two benchmark datasets demonstrate that our MSI-DTI outperforms several state-of-the-art DTIs prediction methods, yielding more accurate and robust predictions. The source codes and datasets are publicly accessible at https://github.com/KEAML-JLU/MSI-DTI.


Asunto(s)
Descubrimiento de Drogas , Biología Computacional/métodos , Algoritmos , Humanos
2.
Brief Bioinform ; 24(2)2023 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-36892155

RESUMEN

Drug-target interaction (DTI) prediction can identify novel ligands for specific protein targets, and facilitate the rapid screening of effective new drug candidates to speed up the drug discovery process. However, the current methods are not sensitive enough to complex topological structures, and complicated relations between multiple node types are not fully captured yet. To address the above challenges, we construct a metapath-based heterogeneous bioinformatics network, and then propose a DTI prediction method with metapath-based hierarchical transformer and attention network for drug-target interaction prediction (MHTAN-DTI), applying metapath instance-level transformer, single-semantic attention and multi-semantic attention to generate low-dimensional vector representations of drugs and proteins. Metapath instance-level transformer performs internal aggregation on the metapath instances, and models global context information to capture long-range dependencies. Single-semantic attention learns the semantics of a certain metapath type, introduces the central node weight and assigns different weights to different metapath instances to obtain the semantic-specific node embedding. Multi-semantic attention captures the importance of different metapath types and performs weighted fusion to attain the final node embedding. The hierarchical transformer and attention network weakens the influence of noise data on the DTI prediction results, and enhances the robustness and generalization ability of MHTAN-DTI. Compared with the state-of-the-art DTI prediction methods, MHTAN-DTI achieves significant performance improvements. In addition, we also conduct sufficient ablation studies and visualize the experimental results. All the results demonstrate that MHTAN-DTI can offer a powerful and interpretable tool for integrating heterogeneous information to predict DTIs and provide new insights into drug discovery.


Asunto(s)
Desarrollo de Medicamentos , Descubrimiento de Drogas , Simulación por Computador , Descubrimiento de Drogas/métodos , Proteínas/química , Aprendizaje
3.
Brief Bioinform ; 24(1)2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36592060

RESUMEN

Drug-target interaction (DTI) prediction is an essential step in drug repositioning. A few graph neural network (GNN)-based methods have been proposed for DTI prediction using heterogeneous biological data. However, existing GNN-based methods only aggregate information from directly connected nodes restricted in a drug-related or a target-related network and are incapable of capturing high-order dependencies in the biological heterogeneous graph. In this paper, we propose a metapath-aggregated heterogeneous graph neural network (MHGNN) to capture complex structures and rich semantics in the biological heterogeneous graph for DTI prediction. Specifically, MHGNN enhances heterogeneous graph structure learning and high-order semantics learning by modeling high-order relations via metapaths. Additionally, MHGNN enriches high-order correlations between drug-target pairs (DTPs) by constructing a DTP correlation graph with DTPs as nodes. We conduct extensive experiments on three biological heterogeneous datasets. MHGNN favorably surpasses 17 state-of-the-art methods over 6 evaluation metrics, which verifies its efficacy for DTI prediction. The code is available at https://github.com/Zora-LM/MHGNN-DTI.


Asunto(s)
Benchmarking , Reposicionamiento de Medicamentos , Sistemas de Liberación de Medicamentos , Aprendizaje , Redes Neurales de la Computación
4.
Brief Bioinform ; 24(2)2023 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-36907663

RESUMEN

The discovery of drug-target interactions (DTIs) is a pivotal process in pharmaceutical development. Computational approaches are a promising and efficient alternative to tedious and costly wet-lab experiments for predicting novel DTIs from numerous candidates. Recently, with the availability of abundant heterogeneous biological information from diverse data sources, computational methods have been able to leverage multiple drug and target similarities to boost the performance of DTI prediction. Similarity integration is an effective and flexible strategy to extract crucial information across complementary similarity views, providing a compressed input for any similarity-based DTI prediction model. However, existing similarity integration methods filter and fuse similarities from a global perspective, neglecting the utility of similarity views for each drug and target. In this study, we propose a Fine-Grained Selective similarity integration approach, called FGS, which employs a local interaction consistency-based weight matrix to capture and exploit the importance of similarities at a finer granularity in both similarity selection and combination steps. We evaluate FGS on five DTI prediction datasets under various prediction settings. Experimental results show that our method not only outperforms similarity integration competitors with comparable computational costs, but also achieves better prediction performance than state-of-the-art DTI prediction approaches by collaborating with conventional base models. Furthermore, case studies on the analysis of similarity weights and on the verification of novel predictions confirm the practical ability of FGS.


Asunto(s)
Desarrollo de Medicamentos , Descubrimiento de Drogas , Descubrimiento de Drogas/métodos , Interacciones Farmacológicas
5.
BMC Bioinformatics ; 25(1): 59, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38321386

RESUMEN

The prediction of interactions between novel drugs and biological targets is a vital step in the early stage of the drug discovery pipeline. Many deep learning approaches have been proposed over the last decade, with a substantial fraction of them sharing the same underlying two-branch architecture. Their distinction is limited to the use of different types of feature representations and branches (multi-layer perceptrons, convolutional neural networks, graph neural networks and transformers). In contrast, the strategy used to combine the outputs (embeddings) of the branches has remained mostly the same. The same general architecture has also been used extensively in the area of recommender systems, where the choice of an aggregation strategy is still an open question. In this work, we investigate the effectiveness of three different embedding aggregation strategies in the area of drug-target interaction (DTI) prediction. We formally define these strategies and prove their universal approximator capabilities. We then present experiments that compare the different strategies on benchmark datasets from the area of DTI prediction, showcasing conditions under which specific strategies could be the obvious choice.


Asunto(s)
Benchmarking , Descubrimiento de Drogas , Suministros de Energía Eléctrica , Redes Neurales de la Computación
6.
Brief Bioinform ; 23(5)2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36070659

RESUMEN

The discovery of drug-target interactions (DTIs) is a very promising area of research with great potential. The accurate identification of reliable interactions among drugs and proteins via computational methods, which typically leverage heterogeneous information retrieved from diverse data sources, can boost the development of effective pharmaceuticals. Although random walk and matrix factorization techniques are widely used in DTI prediction, they have several limitations. Random walk-based embedding generation is usually conducted in an unsupervised manner, while the linear similarity combination in matrix factorization distorts individual insights offered by different views. To tackle these issues, we take a multi-layered network approach to handle diverse drug and target similarities, and propose a novel optimization framework, called Multiple similarity DeepWalk-based Matrix Factorization (MDMF), for DTI prediction. The framework unifies embedding generation and interaction prediction, learning vector representations of drugs and targets that not only retain higher order proximity across all hyper-layers and layer-specific local invariance, but also approximate the interactions with their inner product. Furthermore, we develop an ensemble method (MDMF2A) that integrates two instantiations of the MDMF model, optimizing the area under the precision-recall curve (AUPR) and the area under the receiver operating characteristic curve (AUC), respectively. The empirical study on real-world DTI datasets shows that our method achieves statistically significant improvement over current state-of-the-art approaches in four different settings. Moreover, the validation of highly ranked non-interacting pairs also demonstrates the potential of MDMF2A to discover novel DTIs.


Asunto(s)
Desarrollo de Medicamentos , Descubrimiento de Drogas , Algoritmos , Descubrimiento de Drogas/métodos , Interacciones Farmacológicas , Preparaciones Farmacéuticas , Proteínas
7.
Brief Bioinform ; 23(6)2022 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-36242566

RESUMEN

MOTIVATION: Discovering the drug-target interactions (DTIs) is a crucial step in drug development such as the identification of drug side effects and drug repositioning. Since identifying DTIs by web-biological experiments is time-consuming and costly, many computational-based approaches have been proposed and have become an efficient manner to infer the potential interactions. Although extensive effort is invested to solve this task, the prediction accuracy still needs to be improved. More especially, heterogeneous network-based approaches do not fully consider the complex structure and rich semantic information in these heterogeneous networks. Therefore, it is still a challenge to predict DTIs efficiently. RESULTS: In this study, we develop a novel method via Multiview heterogeneous information network embedding with Hierarchical Attention mechanisms to discover potential Drug-Target Interactions (MHADTI). Firstly, MHADTI constructs different similarity networks for drugs and targets by utilizing their multisource information. Combined with the known DTI network, three drug-target heterogeneous information networks (HINs) with different views are established. Secondly, MHADTI learns embeddings of drugs and targets from multiview HINs with hierarchical attention mechanisms, which include the node-level, semantic-level and graph-level attentions. Lastly, MHADTI employs the multilayer perceptron to predict DTIs with the learned deep feature representations. The hierarchical attention mechanisms could fully consider the importance of nodes, meta-paths and graphs in learning the feature representations of drugs and targets, which makes their embeddings more comprehensively. Extensive experimental results demonstrate that MHADTI performs better than other SOTA prediction models. Moreover, analysis of prediction results for some interested drugs and targets further indicates that MHADTI has advantages in discovering DTIs. AVAILABILITY AND IMPLEMENTATION: https://github.com/pxystudy/MHADTI.


Asunto(s)
Reposicionamiento de Medicamentos , Redes Neurales de la Computación , Interacciones Farmacológicas , Desarrollo de Medicamentos , Servicios de Información
8.
Methods ; 218: 176-188, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37586602

RESUMEN

Drug-target interaction (DTI) prediction serves as the foundation of new drug findings and drug repositioning. For drugs/targets, the sequence data contains the biological structural information, while the heterogeneous network contains the biochemical functional information. These two types of information describe different aspects of drugs and targets. Due to the complexity of DTI machinery, it is necessary to learn the representation from multiple perspectives. We hereby try to design a way to leverage information from multi-source data to the maximum extent and find a strategy to fuse them. To address the above challenges, we propose a model, named MOVE (short for integrating multi-source information for predicting DTI via cross-view contrastive learning), for learning comprehensive representations of each drug and target from multi-source data. MOVE extracts information from the sequence view and the network view, then utilizes a fusion module with auxiliary contrastive learning to facilitate the fusion of representations. Experimental results on the benchmark dataset demonstrate that MOVE is effective in DTI prediction.


Asunto(s)
Desarrollo de Medicamentos , Reposicionamiento de Medicamentos , Simulación por Computador , Desarrollo de Medicamentos/métodos
9.
BMC Bioinformatics ; 24(1): 276, 2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37407927

RESUMEN

BACKGROUND: In many applications of bioinformatics, data stem from distinct heterogeneous sources. One of the well-known examples is the identification of drug-target interactions (DTIs), which is of significant importance in drug discovery. In this paper, we propose a novel framework, manifold optimization based kernel preserving embedding (MOKPE), to efficiently solve the problem of modeling heterogeneous data. Our model projects heterogeneous drug and target data into a unified embedding space by preserving drug-target interactions and drug-drug, target-target similarities simultaneously. RESULTS: We performed ten replications of ten-fold cross validation on four different drug-target interaction network data sets for predicting DTIs for previously unseen drugs. The classification evaluation metrics showed better or comparable performance compared to previous similarity-based state-of-the-art methods. We also evaluated MOKPE on predicting unknown DTIs of a given network. Our implementation of the proposed algorithm in R together with the scripts that replicate the reported experiments is publicly available at https://github.com/ocbinatli/mokpe .


Asunto(s)
Algoritmos , Desarrollo de Medicamentos , Desarrollo de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Biología Computacional/métodos , Interacciones Farmacológicas
10.
BMC Bioinformatics ; 24(1): 488, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38114937

RESUMEN

BACKGROUND: The pharmaceutical field faces a significant challenge in validating drug target interactions (DTIs) due to the time and cost involved, leading to only a fraction being experimentally verified. To expedite drug discovery, accurate computational methods are essential for predicting potential interactions. Recently, machine learning techniques, particularly graph-based methods, have gained prominence. These methods utilize networks of drugs and targets, employing knowledge graph embedding (KGE) to represent structured information from knowledge graphs in a continuous vector space. This phenomenon highlights the growing inclination to utilize graph topologies as a means to improve the precision of predicting DTIs, hence addressing the pressing requirement for effective computational methodologies in the field of drug discovery. RESULTS: The present study presents a novel approach called DTIOG for the prediction of DTIs. The methodology employed in this study involves the utilization of a KGE strategy, together with the incorporation of contextual information obtained from protein sequences. More specifically, the study makes use of Protein Bidirectional Encoder Representations from Transformers (ProtBERT) for this purpose. DTIOG utilizes a two-step process to compute embedding vectors using KGE techniques. Additionally, it employs ProtBERT to determine target-target similarity. Different similarity measures, such as Cosine similarity or Euclidean distance, are utilized in the prediction procedure. In addition to the contextual embedding, the proposed unique approach incorporates local representations obtained from the Simplified Molecular Input Line Entry Specification (SMILES) of drugs and the amino acid sequences of protein targets. CONCLUSIONS: The effectiveness of the proposed approach was assessed through extensive experimentation on datasets pertaining to Enzymes, Ion Channels, and G-protein-coupled Receptors. The remarkable efficacy of DTIOG was showcased through the utilization of diverse similarity measures in order to calculate the similarities between drugs and targets. The combination of these factors, along with the incorporation of various classifiers, enabled the model to outperform existing algorithms in its ability to predict DTIs. The consistent observation of this advantage across all datasets underlines the robustness and accuracy of DTIOG in the domain of DTIs. Additionally, our case study suggests that the DTIOG can serve as a valuable tool for discovering new DTIs.


Asunto(s)
Desarrollo de Medicamentos , Reconocimiento de Normas Patrones Automatizadas , Desarrollo de Medicamentos/métodos , Proteínas/química , Algoritmos , Bases del Conocimiento , Interacciones Farmacológicas
11.
BMC Bioinformatics ; 24(1): 278, 2023 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-37415176

RESUMEN

MOTIVATION: Accurate identification of Drug-Target Interactions (DTIs) plays a crucial role in many stages of drug development and drug repurposing. (i) Traditional methods do not consider the use of multi-source data and do not consider the complex relationship between data sources. (ii) How to better mine the hidden features of drug and target space from high-dimensional data, and better solve the accuracy and robustness of the model. RESULTS: To solve the above problems, a novel prediction model named VGAEDTI is proposed in this paper. We constructed a heterogeneous network with multiple sources of information using multiple types of drug and target dataIn order to obtain deeper features of drugs and targets, we use two different autoencoders. One is variational graph autoencoder (VGAE) which is used to infer feature representations from drug and target spaces. The second is graph autoencoder (GAE) propagating labels between known DTIs. Experimental results on two public datasets show that the prediction accuracy of VGAEDTI is better than that of six DTIs prediction methods. These results indicate that model can predict new DTIs and provide an effective tool for accelerating drug development and repurposing.


Asunto(s)
Desarrollo de Medicamentos , Reposicionamiento de Medicamentos , Interacciones Farmacológicas
12.
Brief Bioinform ; 22(5)2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-33517357

RESUMEN

Accurately identifying potential drug-target interactions (DTIs) is a key step in drug discovery. Although many related experimental studies have been carried out for identifying DTIs in the past few decades, the biological experiment-based DTI identification is still timeconsuming and expensive. Therefore, it is of great significance to develop effective computational methods for identifying DTIs. In this paper, we develop a novel 'end-to-end' learning-based framework based on heterogeneous 'graph' convolutional networks for 'DTI' prediction called end-to-end graph (EEG)-DTI. Given a heterogeneous network containing multiple types of biological entities (i.e. drug, protein, disease, side-effect), EEG-DTI learns the low-dimensional feature representation of drugs and targets using a graph convolutional networks-based model and predicts DTIs based on the learned features. During the training process, EEG-DTI learns the feature representation of nodes in an end-to-end mode. The evaluation test shows that EEG-DTI performs better than existing state-of-art methods. The data and source code are available at: https://github.com/MedicineBiology-AI/EEG-DTI.


Asunto(s)
Simulación por Computador , Desarrollo de Medicamentos , Descubrimiento de Drogas , Aprendizaje Automático , Preparaciones Farmacéuticas/química , Programas Informáticos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Proteínas/química , Proteínas/metabolismo
13.
Brief Bioinform ; 22(2): 2141-2150, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-32367110

RESUMEN

Identification of new drug-target interactions (DTIs) is an important but a time-consuming and costly step in drug discovery. In recent years, to mitigate these drawbacks, researchers have sought to identify DTIs using computational approaches. However, most existing methods construct drug networks and target networks separately, and then predict novel DTIs based on known associations between the drugs and targets without accounting for associations between drug-protein pairs (DPPs). To incorporate the associations between DPPs into DTI modeling, we built a DPP network based on multiple drugs and proteins in which DPPs are the nodes and the associations between DPPs are the edges of the network. We then propose a novel learning-based framework, 'graph convolutional network (GCN)-DTI', for DTI identification. The model first uses a graph convolutional network to learn the features for each DPP. Second, using the feature representation as an input, it uses a deep neural network to predict the final label. The results of our analysis show that the proposed framework outperforms some state-of-the-art approaches by a large margin.


Asunto(s)
Aprendizaje Profundo , Sistemas de Liberación de Medicamentos , Redes Neurales de la Computación , Algoritmos , Humanos
14.
Brief Bioinform ; 22(1): 247-269, 2021 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-31950972

RESUMEN

The task of predicting the interactions between drugs and targets plays a key role in the process of drug discovery. There is a need to develop novel and efficient prediction approaches in order to avoid costly and laborious yet not-always-deterministic experiments to determine drug-target interactions (DTIs) by experiments alone. These approaches should be capable of identifying the potential DTIs in a timely manner. In this article, we describe the data required for the task of DTI prediction followed by a comprehensive catalog consisting of machine learning methods and databases, which have been proposed and utilized to predict DTIs. The advantages and disadvantages of each set of methods are also briefly discussed. Lastly, the challenges one may face in prediction of DTI using machine learning approaches are highlighted and we conclude by shedding some lights on important future research directions.


Asunto(s)
Biología Computacional/métodos , Descubrimiento de Drogas/métodos , Aprendizaje Automático , Bases de Datos Factuales , Humanos
15.
BMC Bioinformatics ; 23(1): 564, 2022 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-36581822

RESUMEN

BACKGROUND: Identifying drug-target interactions (DTIs) plays a key role in drug development. Traditional wet experiments to identify DTIs are expensive and time consuming. Effective computational methods to predict DTIs are useful to narrow the searching scope of potential drugs and speed up the process of drug discovery. There are a variety of non-negativity matrix factorization based methods to predict DTIs, but the convergence of the algorithms used in the matrix factorization are often overlooked and the results can be further improved. RESULTS: In order to predict DTIs more accurately and quickly, we propose an alternating direction algorithm to solve graph regularized non-negative matrix factorization with prior knowledge consistency constraint (ADA-GRMFC). Based on known DTIs, drug chemical structures and target sequences, ADA-GRMFC at first constructs a DTI matrix, a drug similarity matrix and a target similarity matrix. Then DTI prediction is modeled as the non-negative factorization of the DTI matrix with graph dual regularization terms and a prior knowledge consistency constraint. The graph dual regularization terms are used to integrate the information from the drug similarity matrix and the target similarity matrix, and the prior knowledge consistency constraint is used to ensure the matrix decomposition result should be consistent with the prior knowledge of known DTIs. Finally, an alternating direction algorithm is used to solve the matrix factorization. Furthermore, we prove that the algorithm can converge to a stationary point. Extensive experimental results of 10-fold cross-validation show that ADA-GRMFC has better performance than other state-of-the-art methods. In the case study, ADA-GRMFC is also used to predict the targets interacting with the drug olanzapine, and all of the 10 highest-scoring targets have been accurately predicted. In predicting drug interactions with target estrogen receptors alpha, 17 of the 20 highest-scoring drugs have been validated.


Asunto(s)
Desarrollo de Medicamentos , Descubrimiento de Drogas , Descubrimiento de Drogas/métodos , Interacciones Farmacológicas , Algoritmos
16.
BMC Bioinformatics ; 23(1): 245, 2022 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-35729494

RESUMEN

BACKGROUND: Drug-target interactions (DTIs) are critical for drug repurposing and elucidation of drug mechanisms, and are manually curated by large databases, such as ChEMBL, BindingDB, DrugBank and DrugTargetCommons. However, the number of curated articles likely constitutes only a fraction of all the articles that contain experimentally determined DTIs. Finding such articles and extracting the experimental information is a challenging task, and there is a pressing need for systematic approaches to assist the curation of DTIs. To this end, we applied Bidirectional Encoder Representations from Transformers (BERT) to identify such articles. Because DTI data intimately depends on the type of assays used to generate it, we also aimed to incorporate functions to predict the assay format. RESULTS: Our novel method identified 0.6 million articles (along with drug and protein information) which are not previously included in public DTI databases. Using 10-fold cross-validation, we obtained ~ 99% accuracy for identifying articles containing quantitative drug-target profiles. The F1 micro for the prediction of assay format is 88%, which leaves room for improvement in future studies. CONCLUSION: The BERT model in this study is robust and the proposed pipeline can be used to identify previously overlooked articles containing quantitative DTIs. Overall, our method provides a significant advancement in machine-assisted DTI extraction and curation. We expect it to be a useful addition to drug mechanism discovery and repurposing.


Asunto(s)
Reposicionamiento de Medicamentos , Proteínas , Bases de Datos Factuales , Interacciones Farmacológicas , Proteínas/metabolismo , PubMed
17.
Adv Exp Med Biol ; 1361: 119-141, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35230686

RESUMEN

The wealth of knowledge and multi-omics data available in drug research has allowed the rise of several computational methods in the drug discovery field, resulting in a novel and exciting strategy called drug repurposing. Drug repurposing consists in finding new applications for existing drugs. Numerous computational methods perform a high-level integration of different knowledge sources to facilitate the discovery of unknown mechanisms. In this chapter, we present a survey of data resources and computational tools available for drug repositioning.


Asunto(s)
Descubrimiento de Drogas , Reposicionamiento de Medicamentos , Biología Computacional/métodos , Descubrimiento de Drogas/métodos , Reposicionamiento de Medicamentos/métodos
18.
BMC Bioinformatics ; 22(1): 418, 2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34479477

RESUMEN

BACKGROUND: Prediction of the drug-target interaction (DTI) is a critical step in the drug repurposing process, which can effectively reduce the following workload for experimental verification of potential drugs' properties. In recent studies, many machine-learning-based methods have been proposed to discover unknown interactions between drugs and protein targets. A recent trend is to use graph-based machine learning, e.g., graph embedding to extract features from drug-target networks and then predict new drug-target interactions. However, most of the graph embedding methods are not specifically designed for DTI predictions; thus, it is difficult for these methods to fully utilize the heterogeneous information of drugs and targets (e.g., the respective vertex features of drugs and targets and path-based interactive features between drugs and targets). RESULTS: We propose a DTI prediction method DTI-HeNE (DTI based on Heterogeneous Network Embedding), which is specifically designed to cope with the bipartite DTI relations for generating high-quality embeddings of drug-target pairs. This method splits a heterogeneous DTI network into a bipartite DTI network, multiple drug homogeneous networks and target homogeneous networks, and extracts features from these sub-networks separately to better utilize the characteristics of bipartite DTI relations as well as the auxiliary similarity information related to drugs and targets. The features extracted from each sub-network are integrated using pathway information between these sub-networks to acquire new features, i.e., embedding vectors of drug-target pairs. Finally, these features are fed into a random forest (RF) model to predict novel DTIs. CONCLUSIONS: Our experimental results show that, the proposed DTI network embedding method can learn higher-quality features of heterogeneous drug-target interaction networks for novel DTIs discovery.


Asunto(s)
Desarrollo de Medicamentos , Preparaciones Farmacéuticas , Interacciones Farmacológicas , Reposicionamiento de Medicamentos , Aprendizaje Automático
19.
Brief Bioinform ; 20(4): 1337-1357, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-29377981

RESUMEN

Computational prediction of drug-target interactions (DTIs) has become an essential task in the drug discovery process. It narrows down the search space for interactions by suggesting potential interaction candidates for validation via wet-lab experiments that are well known to be expensive and time-consuming. In this article, we aim to provide a comprehensive overview and empirical evaluation on the computational DTI prediction techniques, to act as a guide and reference for our fellow researchers. Specifically, we first describe the data used in such computational DTI prediction efforts. We then categorize and elaborate the state-of-the-art methods for predicting DTIs. Next, an empirical comparison is performed to demonstrate the prediction performance of some representative methods under different scenarios. We also present interesting findings from our evaluation study, discussing the advantages and disadvantages of each method. Finally, we highlight potential avenues for further enhancement of DTI prediction performance as well as related research directions.


Asunto(s)
Desarrollo de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Teorema de Bayes , Quimioinformática , Biología Computacional , Simulación por Computador , Árboles de Decisión , Desarrollo de Medicamentos/estadística & datos numéricos , Descubrimiento de Drogas/estadística & datos numéricos , Interacciones Farmacológicas , Reposicionamiento de Medicamentos/métodos , Reposicionamiento de Medicamentos/estadística & datos numéricos , Lógica Difusa , Humanos , Análisis de los Mínimos Cuadrados , Aprendizaje Automático , Modelos Estadísticos , Pruebas de Farmacogenómica/métodos , Pruebas de Farmacogenómica/estadística & datos numéricos , Máquina de Vectores de Soporte , Encuestas y Cuestionarios
20.
BMC Bioinformatics ; 20(Suppl 8): 287, 2019 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-31182006

RESUMEN

BACKGROUND: Predicting drug-target interactions is time-consuming and expensive. It is important to present the accuracy of the calculation method. There are many algorithms to predict global interactions, some of which use drug-target networks for prediction (ie, a bipartite graph of bound drug pairs and targets known to interact). Although these algorithms can predict some drug-target interactions to some extent, there is little effect for some new drugs or targets that have no known interaction. RESULTS: Since the datasets are usually located at or near low-dimensional nonlinear manifolds, we propose an improved GRMF (graph regularized matrix factorization) method to learn these flow patterns in combination with the previous matrix-decomposition method. In addition, we use one of the pre-processing steps previously proposed to improve the accuracy of the prediction. CONCLUSIONS: Cross-validation is used to evaluate our method, and simulation experiments are used to predict new interactions. In most cases, our method is superior to other methods. Finally, some examples of new drugs and new targets are predicted by performing simulation experiments. And the improved GRMF method can better predict the remaining drug-target interactions.


Asunto(s)
Algoritmos , Interacciones Farmacológicas , Bases de Datos como Asunto , Humanos , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda