1
|
Gao X, Yan M, Zhang C, Wu G, Shang J, Zhang C, Yang K. MDNN-DTA: a multimodal deep neural network for drug-target affinity prediction. Front Genet 2025; 16:1527300. [PMID: 40182923 PMCID: PMC11965683 DOI: 10.3389/fgene.2025.1527300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Determining drug-target affinity (DTA) is a pivotal step in drug discovery, where in silico methods can significantly improve efficiency and reduce costs. Artificial intelligence (AI), especially deep learning models, can automatically extract high-dimensional features from the biological sequences of drug molecules and target proteins. This technology demonstrates lower complexity in DTA prediction compared to traditional experimental methods, particularly when handling large-scale data. In this study, we introduce a multimodal deep neural network model for DTA prediction, referred to as MDNN-DTA. This model employs Graph Convolutional Networks (GCN) and Convolutional Neural Networks (CNN) to extract features from the drug and protein sequences, respectively. One notable strength of our method is its ability to accurately predict DTA directly from the sequences of the target proteins, obviating the need for protein 3D structures, which are frequently unavailable in drug discovery. To comprehensively extract features from the protein sequence, we leverage an ESM pre-trained model for extracting biochemical features and design a specific Protein Feature Extraction (PFE) block for capturing both global and local features of the protein sequence. Furthermore, a Protein Feature Fusion (PFF) Block is engineered to augment the integration of multi-scale protein features derived from the abovementioned techniques. We then compare MDNN-DTA with other models on the same dataset, conducting a series of ablation experiments to assess the performance and efficacy of each component. The results highlight the advantages and effectiveness of the MDNN-DTA method.
Collapse
Affiliation(s)
- Xu Gao
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou, China
- National Supercomputing Center in Zhengzhou, Zhengzhou, China
| | - Mengfan Yan
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou, China
- National Supercomputing Center in Zhengzhou, Zhengzhou, China
| | - Chengwei Zhang
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou, China
- National Supercomputing Center in Zhengzhou, Zhengzhou, China
| | - Gang Wu
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou, China
- National Supercomputing Center in Zhengzhou, Zhengzhou, China
| | - Jiandong Shang
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou, China
- National Supercomputing Center in Zhengzhou, Zhengzhou, China
| | - Congxiang Zhang
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou, China
- National Supercomputing Center in Zhengzhou, Zhengzhou, China
| | - Kecheng Yang
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou, China
- National Supercomputing Center in Zhengzhou, Zhengzhou, China
| |
Collapse
|
2
|
Lu Z, Song G, Zhu H, Lei C, Sun X, Wang K, Qin L, Chen Y, Tang J, Li M. DTIAM: a unified framework for predicting drug-target interactions, binding affinities and drug mechanisms. Nat Commun 2025; 16:2548. [PMID: 40089473 PMCID: PMC11910601 DOI: 10.1038/s41467-025-57828-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Accurate and robust prediction of drug-target interactions (DTIs) plays a vital role in drug discovery but remains challenging due to limited labeled data, cold start problems, and insufficient understanding of mechanisms of action (MoA). Distinguishing activation and inhibition mechanisms is particularly critical in clinical applications. Here, we propose DTIAM, a unified framework for predicting interactions, binding affinities, and activation/inhibition mechanisms between drugs and targets. DTIAM learns drug and target representations from large amounts of label-free data through self-supervised pre-training, which accurately extracts their substructure and contextual information, and thus benefits the downstream prediction based on these representations. DTIAM achieves substantial performance improvement over other state-of-the-art methods in all tasks, particularly in the cold start scenario. Moreover, independent validation demonstrates the strong generalization ability of DTIAM. All these results suggest that DTIAM can provide a practically useful tool for predicting novel DTIs and further distinguishing the MoA of candidate drugs.
Collapse
Affiliation(s)
- Zhangli Lu
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
| | - Guoqiang Song
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China
| | - Huimin Zhu
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
| | - Chuqi Lei
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
| | - Xinliang Sun
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
| | - Kaili Wang
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
| | - Libo Qin
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
| | - Yafei Chen
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China
| | - Jing Tang
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Min Li
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China.
- Xiangjiang Laboratory, Changsha, 410205, China.
- Furong Laboratory, Central South University, Changsha, 410013, China.
| |
Collapse
|
3
|
Li J, Zhang J, Guo R, Dai J, Niu Z, Wang Y, Wang T, Jiang X, Hu W. Progress of machine learning in the application of small molecule druggability prediction. Eur J Med Chem 2025; 285:117269. [PMID: 39808972 DOI: 10.1016/j.ejmech.2025.117269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Machine learning (ML) has become an important tool for predicting the pharmaceutical properties of small molecules. Recent advancements in ML algorithms enable the rapid and accurate evaluation of solubility, activity, toxicity, pharmacokinetics, and other molecular properties through ML-based models. By conducting virtual screening of drug targets and elucidating drug-target protein interactions, researchers can conduct preliminary evaluations of the activity and safety of compounds from the ultra-large drug compound libraries, thereby accelerating the screening process for lead compounds. Moreover, ML leverages existing experimental data to train and generate new datasets, addressing the challenge of limited compounds and protein target data. This review provided a concise overview of ML applications in predicting small molecule properties, focusing on model construction principles, molecular feature selection, and other essential aspects. It also discussed the potential applications of ML in the screening of pharmaceutical small molecules.
Collapse
Affiliation(s)
- Junyao Li
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China; School of Life Sciences, Huaiyin Normal University, Huaian, 223300, China; Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Jianmei Zhang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China
| | - Rui Guo
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China; Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Jiawei Dai
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Zhiqiang Niu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Yan Wang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China
| | - Taoyun Wang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China.
| | - Xiaojian Jiang
- School of Life Sciences, Huaiyin Normal University, Huaian, 223300, China.
| | - Weicheng Hu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
4
|
Abbasi H, Lakizadeh A. Drug Repurposing Using Hypergraph Embedding Based on Common Therapeutic Targets of a Drug. J Comput Biol 2025; 32:316-329. [PMID: 39648844 DOI: 10.1089/cmb.2023.0427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024] Open
Abstract
Developing a new drug is a long and expensive process that typically takes 10-15 years and costs billions of dollars. This has led to an increasing interest in drug repositioning, which involves finding new therapeutic uses for existing drugs. Computational methods become an increasingly important tool for identifying associations between drugs and new diseases. Graph- and hypergraph-based approaches are a type of computational method that can be used to identify potential associations between drugs and new diseases. Here, we present a drug repurposing method based on hypergraph neural network for predicting drug-disease association in three stages. First, it constructs a heterogeneous graph that contains drug and disease nodes and links between them; in the second stage, it converts the heterogeneous simple graph to a hypergraph with only disease nodes. This is achieved by grouping diseases that use the same drug into a hyperedge. Indeed, all the diseases that are the common therapeutic goal of a drug are placed on a hyperedge. Finally, a graph neural network is used to predict drug-disease association based on the structure of the hypergraph. This model is more efficient than other methods because it uses a hypergraph to model relationships more effectively than graphs. Furthermore, it constructs the hypergraph using only a drug-disease association matrix, eliminating the need for extensive amounts of data. Experimental results show that the hypergraph-based approach effectively captures complex interrelationships between drugs and diseases, leading to improved accuracy of drug-disease association prediction compared to state-of-the-art methods.
Collapse
Affiliation(s)
- Hanieh Abbasi
- Computer Engineering Department, University of Qom, Qom, Iran
| | - Amir Lakizadeh
- Computer Engineering Department, University of Qom, Qom, Iran
| |
Collapse
|
5
|
Cheng Z, Xu D, Ding D, Ding Y. Prediction of Drug-Target Interactions With High- Quality Negative Samples and a Network-Based Deep Learning Framework. IEEE J Biomed Health Inform 2025; 29:1567-1578. [PMID: 38227407 DOI: 10.1109/jbhi.2024.3354953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Identification of drug-target interactions (DTIs) plays a crucial role in drug discovery. Compared to traditional experimental methods, computer-based methods for predicting DTIs can significantly reduce the time and financial burdens of drug development. In recent years, numerous machine learning-based methods have been proposed for predicting potential DTIs. However, a common limitation among these methods is the absence of high-quality negative samples. Moreover, the effective extraction of multisource information of drugs and proteins for DTI prediction remains a significant challenge. In this paper, we investigated two aspects: the selection of high-quality negative samples and the construction of a high-performance DTI prediction framework. Specifically, we found two types of hidden biases when randomly selecting negative samples from unlabeled drug-protein pairs and proposed a negative sample selection approach based on complex network theory. Furthermore, we proposed a novel DTI prediction method named HNetPa-DTI, which integrates topological information from the drug-protein-disease heterogeneous network and gene ontology (GO) and pathway annotation information of proteins. Specifically, we extracted topological information of the drug-protein-disease heterogeneous network using heterogeneous graph neural networks, and obtained GO and pathway annotation information of proteins from the GO term semantic similarity networks, GO term-protein bipartite networks, and pathway-protein bipartite network using graph neural networks. Experimental results show that HNetPa-DTI outperforms the baseline methods on four types of prediction tasks, demonstrating the superiority of our method.
Collapse
|
6
|
Yu H, Lu M, Li Z, Zhang Y. Semantic-Enhanced Graph Contrastive Learning With Adaptive Denoising for Drug Repositioning. IEEE J Biomed Health Inform 2025; 29:1635-1643. [PMID: 38109249 DOI: 10.1109/jbhi.2023.3344031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
The traditional drug development process requires a significant investment in workforce and financial resources. Drug repositioning as an efficient alternative has attracted much attention during the last few years. Despite the wide application and success of the method, there are still many shortcomings in the existing model. For example, sparse datasets will seriously affect the existing methods' performance. Additionally, these methods do not pay attention to the noise in datasets. In response to the above defects, we propose a semantic-enriched augmented graph contrastive learning with an adaptive denoising method, called SGCD. This method enhances data from the perspective of the embedding layer, deeply mines potential neighborhood relation-ships in semantic space, and combines similar drugs in the semantic neighborhoods into prototype comparison targets, thus effectively mitigating the impact of data sparsity on the model. Moreover, to enhance the model's robustness to noisy data, we use the adaptive denoising method, which can effectively identify noisy data in the training process. Exhaustive experiments on multiple real datasets show the effectiveness of the proposed model.
Collapse
|
7
|
Zuo Y, Wu X, Ge F, Yan H, Fei S, Liang J, Deng Z. Research progress on Drug-Target Interactions in the last five years. Anal Biochem 2025; 697:115691. [PMID: 39455038 DOI: 10.1016/j.ab.2024.115691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/06/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
The identification of Drug-Target Interaction (DTI) is an important step in drug discovery and drug repositioning, and has high application value in multiple fields such as drug discovery, drug repositioning, and repurposing. However, the high cost of experimental validation limits its identification. In contrast, computation-based approaches are both economical and efficient. This review first synthesizes existing chemical genomic approaches, provides a comprehensive summary of prevalent databases for predicting DTIs, and categorizes the feature encodings from recent years. This is followed by an overview and brief description of the methods currently in use for predicting DTIs. The strengths and weaknesses of newly proposed prediction methods in the last five years (2020-2024), including those based on network representation learning and graph neural networks, are then discussed in detail, evaluating the performance of the different methods on a wide range of datasets. Finally, this review explores potential directions for future DTI research, emphasizing how to improve prediction accuracy and efficiency by combining big data and emerging computing technologies.
Collapse
Affiliation(s)
- Yun Zuo
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China.
| | - Xubin Wu
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China
| | - Fei Ge
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China
| | - Hongjin Yan
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China
| | - Sirui Fei
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China
| | - Jingwen Liang
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China
| | - Zhaohong Deng
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214000, China.
| |
Collapse
|
8
|
Wang X, Zhang M, Xu J, Li X, Xiong J, Cao H, Dou F, Zhai X, Sun H. A novel approach for target deconvolution from phenotype-based screening using knowledge graph. Sci Rep 2025; 15:2414. [PMID: 39827292 PMCID: PMC11742725 DOI: 10.1038/s41598-025-86166-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
Deconvoluting drug targets is crucial in modern drug development, yet both traditional and artificial intelligence (AI)-driven methods face challenges in terms of completeness, accuracy, and efficiency. Identifying drug targets, especially within complex systems such as the p53 pathway, remains a formidable task. The regulation of this pathway by myriad stress signals and regulatory elements adds layers of complexity to the discovery of effective p53 pathway activators. Recent insights into p53 activation have led to two main screening strategies for p53 activators. The target-based approach focuses on p53 and its regulators (MDM2, MDMX, USP7, Sirt proteins), but requires separate systems for each target and may miss multi-target compounds. Phenotype-based screening can reveal new targets but involves a lengthy process to elucidate mechanisms and targets, hindering drug development. Knowledge graphs have emerged as powerful tools that offer strengths in link prediction and knowledge inference to address these issues. In this study, we constructed a protein-protein interaction knowledge graph (PPIKG) and pioneered an integrated drug target deconvolution system that combines AI with molecular docking techniques. Analysis based on the PPIKG narrowed down candidate proteins from 1088 to 35, significantly saving time and cost. Subsequent molecular docking led us to pinpoint USP7 as a direct target for the p53 pathway activator UNBS5162. Leveraging knowledge graphs and a multidisciplinary approach allows us to streamline the laborious and expensive process of reverse targeting drug discovery through phenotype screening. Our findings have the potential to revolutionize drug screening and open new avenues in pharmacological research, increasing the speed and efficiency of pursuing novel therapeutics. The code is available at https://github.com/Xiong-Jing/PPIKG .
Collapse
Affiliation(s)
- Xiaohong Wang
- Shandong Foreign Trade Vocational College, Qingdao, 266100, China
| | - Meifang Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
| | - Jianliang Xu
- Faculty of Information Science and Engineering, Ocean University of China, Qingdao, 266071, China
| | - Xin Li
- Gansu Health Vocational College, Lanzhou, 730000, China
| | - Jing Xiong
- School of Computer Science, Qufu Normal University, Rizhao, 276827, China.
- Rizhao-Qufu Normal University Joint Technology Transfer Center, Rizhao, 276827, China.
- International Joint Research Laboratory for Perception Data Intelligent Processing of Henan, Anyang Normal University, Anyang, 455000, China.
| | - Haowei Cao
- Shandong Computer Science Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 255000, China
- Shandong Provincial Key Laboratory of Computer Networks, Shandong Fundamental Research Center for Computer Science, Jinan, 255000, China
| | - Fangkun Dou
- Oceanographic Data Center, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Xue Zhai
- School of Engineering, Qufu Normal University, Rizhao, 276827, China
| | - Hua Sun
- International Joint Research Laboratory for Perception Data Intelligent Processing of Henan, Anyang Normal University, Anyang, 455000, China
| |
Collapse
|
9
|
Feng C, Cheng J, Sun M, Qiao C, Feng Q, Fang N, Ge Y, Rui M. Artificial intelligence-driven identification and mechanistic exploration of synergistic anti-breast cancer compound combinations from Prunella vulgaris L.- Taraxacum mongolicum Hand.-Mazz. herb pair. Front Pharmacol 2025; 15:1522787. [PMID: 39840098 PMCID: PMC11747269 DOI: 10.3389/fphar.2024.1522787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/13/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction The Prunella vulgaris L. (PVL) and Taraxacum mongolicum Hand.-Mazz. (TH) herb pair, which is commonly used in traditional Chinese medicine (TCM), has been applied for the treatment of breast cancer. Although its efficacy is validated, the synergistic anti-breast cancer compound combinations within this herb pair and their underlying mechanisms of action remain unclear. Methods This study aimed to identify and validate synergistic anti-breast cancer compound combinations within the PVL-TH pair using large-scale biomedical data, artificial intelligence and experimental methods. The first step was to investigate the anti-breast cancer effects of various PVL and TH extracts using in vitro cellular assays to identify the most effective superior extracts. These superior extracts were subjected to liquid chromatography-mass spectrometry (LC-MS) analysis to identify their constituent compounds. A deep learning-based prediction model, DeepMDS, was applied to predict synergistic anti-breast cancer multi-compound combinations. These predicted combinations were experimentally validated for their anti-breast cancer effects at actual content ratios found in the extracts. Preliminary bioinformatics analyses were conducted to explore the mechanisms of action of these superior combinations. We also compared the anti-breast cancer effects of superior extracts from different geographical origins and analyzed the contents of compounds to assess their representation of the anti-tumor effect of the corresponding TCM. Results The results revealed that LC-MS analysis identified 27 and 21 compounds in the superior extracts (50% ethanol extracts) of PVL and TH, respectively. Based on these compounds, DeepMDS model predicted synergistic anti-breast cancer compound combinations such as F973 (caffeic acid, rosmarinic acid, p-coumaric acid, and esculetin), T271 (chlorogenic acid, cichoric acid, and caffeic acid), and T1685 (chlorogenic acid, rosmarinic acid, and scopoletin) from single PVL, single TH and PVL-TH herb pair, respectively. These combinations, at their actual concentrations in extracts, demonstrated superior anti-breast cancer activity compared to the corresponding extracts. The bioinformatics analysis revealed that these compounds could regulate tumor-related pathways synergistically, inhibiting tumor cell growth, inducing cell apoptosis, and blocking cell cycle progression. Furthermore, the concentration ratio and total content of compounds in F973 and T271 were closely associated with their anti-breast cancer effects in extracts from various geographical origins. The compound combination T1685 could represent the synergistic anti-breast cancer effects of the PVL-TH pair. Discussion This study provides insights into exploring the representative synergistic anti-breast cancer compound combinations within the complex TCM.
Collapse
Affiliation(s)
- Chunlai Feng
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | | | | | | | | | | | | | - Mengjie Rui
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| |
Collapse
|
10
|
Shi W, Yang H, Xie L, Yin XX, Zhang Y. A review of machine learning-based methods for predicting drug-target interactions. Health Inf Sci Syst 2024; 12:30. [PMID: 38617016 PMCID: PMC11014838 DOI: 10.1007/s13755-024-00287-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/04/2024] [Indexed: 04/16/2024] Open
Abstract
The prediction of drug-target interactions (DTI) is a crucial preliminary stage in drug discovery and development, given the substantial risk of failure and the prolonged validation period associated with in vitro and in vivo experiments. In the contemporary landscape, various machine learning-based methods have emerged as indispensable tools for DTI prediction. This paper begins by placing emphasis on the data representation employed by these methods, delineating five representations for drugs and four for proteins. The methods are then categorized into traditional machine learning-based approaches and deep learning-based ones, with a discussion of representative approaches in each category and the introduction of a novel taxonomy for deep neural network models in DTI prediction. Additionally, we present a synthesis of commonly used datasets and evaluation metrics to facilitate practical implementation. In conclusion, we address current challenges and outline potential future directions in this research field.
Collapse
Affiliation(s)
- Wen Shi
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006 China
- School of Computer Science and Technology, Zhejiang Normal University, Jinhua, 321004 China
| | - Hong Yang
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006 China
| | - Linhai Xie
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing, 102206 China
| | - Xiao-Xia Yin
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006 China
| | - Yanchun Zhang
- School of Computer Science and Technology, Zhejiang Normal University, Jinhua, 321004 China
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, 518000 China
| |
Collapse
|
11
|
Kesimoglu ZN, Bozdag S. Fusing multiplex heterogeneous networks using graph attention-aware fusion networks. Sci Rep 2024; 14:29119. [PMID: 39582056 PMCID: PMC11586420 DOI: 10.1038/s41598-024-78555-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 10/31/2024] [Indexed: 11/26/2024] Open
Abstract
Graph Neural Networks (GNN) emerged as a deep learning framework to generate node and graph embeddings for downstream machine learning tasks. Popular GNN-based architectures operate on networks of single node and edge type. However, a large number of real-world networks include multiple types of nodes and edges. Enabling these architectures to work on networks with multiple node and edge types brings additional challenges due to the heterogeneity of the networks and the multiplicity of the existing associations. In this study, we present a framework, named GRAF (Graph Attention-aware Fusion Networks), to convert multiplex heterogeneous networks to homogeneous networks to make them more suitable for graph representation learning. Using attention-based neighborhood aggregation, GRAF learns the importance of each neighbor per node (called node-level attention) followed by the importance of each network layer (called network layer-level attention). Then, GRAF processes a network fusion step weighing each edge according to the learned attentions. After an edge elimination step based on edge weights, GRAF utilizes Graph Convolutional Networks (GCN) on the fused network and incorporates node features on graph-structured data for a node classification or a similar downstream task. To demonstrate GRAF's generalizability, we applied it to four datasets from different domains and observed that GRAF outperformed or was on par with the baselines and state-of-the-art (SOTA) methods. We were able to interpret GRAF's findings utilizing the attention weights. Source code for GRAF is publicly available at https://github.com/bozdaglab/GRAF .
Collapse
Affiliation(s)
- Ziynet Nesibe Kesimoglu
- Department of Computer Science and Engineering, University of North Texas, Denton, TX, USA
- BioDiscovery Institute, University of North Texas, Denton, TX, USA
| | - Serdar Bozdag
- Department of Computer Science and Engineering, University of North Texas, Denton, TX, USA.
- Department of Mathematics, University of North Texas, Denton, TX, USA.
- BioDiscovery Institute, University of North Texas, Denton, TX, USA.
| |
Collapse
|
12
|
Garg P, Singhal G, Kulkarni P, Horne D, Salgia R, Singhal SS. Artificial Intelligence-Driven Computational Approaches in the Development of Anticancer Drugs. Cancers (Basel) 2024; 16:3884. [PMID: 39594838 PMCID: PMC11593155 DOI: 10.3390/cancers16223884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
The integration of AI has revolutionized cancer drug development, transforming the landscape of drug discovery through sophisticated computational techniques. AI-powered models and algorithms have enhanced computer-aided drug design (CADD), offering unprecedented precision in identifying potential anticancer compounds. Traditionally, cancer drug design has been a complex, resource-intensive process, but AI introduces new opportunities to accelerate discovery, reduce costs, and optimize efficiency. This manuscript delves into the transformative applications of AI-driven methodologies in predicting and developing anticancer drugs, critically evaluating their potential to reshape the future of cancer therapeutics while addressing their challenges and limitations.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Gargi Singhal
- Department of Medical Sciences, S.N. Medical College, Agra 282002, Uttar Pradesh, India
| | - Prakash Kulkarni
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
13
|
Liu B, Tsoumakas G. Integrating Similarities via Local Interaction Consistency and Optimizing Area Under the Curve Measures via Matrix Factorization for Drug-Target Interaction Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:2212-2225. [PMID: 39226198 DOI: 10.1109/tcbb.2024.3453499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
In drug discovery, identifying drug-target interactions (DTIs) via experimental approaches is a tedious and expensive procedure. Computational methods efficiently predict DTIs and recommend a small part of potential interacting pairs for further experimental confirmation, accelerating the drug discovery process. Although fusing heterogeneous drug and target similarities can improve the prediction ability, the existing similarity combination methods ignore the interaction consistency for neighbour entities. Furthermore, area under the precision-recall curve (AUPR) and area under the receiver operating characteristic curve (AUC) are two widely used evaluation metrics in DTI prediction. However, the two metrics are seldom considered as losses within existing DTI prediction methods. We propose a local interaction consistency (LIC) aware similarity integration method to fuse vital information from diverse views for DTI prediction models. Furthermore, we propose two matrix factorization (MF) methods that optimize AUPR and AUC using convex surrogate losses respectively, and then develop an ensemble MF approach that takes advantage of the two area under the curve metrics by combining the two single metric based MF models. Experimental results under different prediction settings show that the proposed methods outperform various competitors in terms of the metric(s) they optimize and are reliable in discovering potential new DTIs.
Collapse
|
14
|
Xu P, Wei Z, Li C, Yuan J, Liu Z, Liu W. Drug-Target Prediction Based on Dynamic Heterogeneous Graph Convolutional Network. IEEE J Biomed Health Inform 2024; 28:6997-7005. [PMID: 39120984 DOI: 10.1109/jbhi.2024.3441324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2024]
Abstract
Novel drug-target interaction (DTI) prediction is crucial in drug discovery and repositioning. Recently, graph neural network (GNN) has shown promising results in identifying DTI by using thresholds to construct heterogeneous graphs. However, an empirically selected threshold can lead to loss of valuable information, especially in sparse networks, a common scenario in DTI prediction. To make full use of insufficient information, we propose a DTI prediction model based on Dynamic Heterogeneous Graph (DT-DHG). And progressive learning is introduced to adjust the receptive fields of node. The experimental results show that our method significantly improves the performance of the original GNNs and is robust against the choices of backbones. Meanwhile, DT-DHG outperforms the state-of-the-art methods and effectively predicts novel DTIs.
Collapse
|
15
|
Lu Q, Zhou Z, Wang Q. Multi-layer graph attention neural networks for accurate drug-target interaction mapping. Sci Rep 2024; 14:26119. [PMID: 39478027 PMCID: PMC11525987 DOI: 10.1038/s41598-024-75742-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
In the crucial process of drug discovery and repurposing, precise prediction of drug-target interactions (DTIs) is paramount. This study introduces a novel DTI prediction approach-Multi-Layer Graph Attention Neural Network (MLGANN), through a groundbreaking computational framework that effectively harnesses multi-source information to enhance prediction accuracy. MLGANN not only strides forward in constructing a multi-layer DTI network by capturing both direct interactions between drugs and targets as well as their multi-level information but also amalgamates Graph Convolutional Networks (GCN) with a self-attention mechanism to comprehensively integrate diverse data sources. This method exhibited significant performance surpassing existing approaches in comparative experiments, underscoring its immense potential in elevating the efficiency and accuracy of DTI predictions. More importantly, this study accentuates the significance of considering multi-source data information and network heterogeneity in the drug discovery process, offering new perspectives and tools for future pharmaceutical research.
Collapse
Affiliation(s)
- Qianwen Lu
- SDU-ANU Joint Science College, Shandong University, Weihai, 264209, Shandong, China
| | - Zhiheng Zhou
- Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, 100190, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Qi Wang
- College of Science, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
16
|
Wang Y, Yin Z. Drug-target interaction prediction through fine-grained selection and bidirectional random walk methodology. Sci Rep 2024; 14:18104. [PMID: 39103483 PMCID: PMC11300600 DOI: 10.1038/s41598-024-69186-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/01/2024] [Indexed: 08/07/2024] Open
Abstract
The study of drug-target interaction plays an important role in the process of drug development. The subject of DTI forecasting has advanced significantly in the last several years, yielding numerous significant research findings and methodologies. Heterogeneous data sources provide richer information and comprehensive perspectives for drug-target interaction prediction, so many existing methods rely on heterogeneous networks, and graph embedding technology becomes an important technology to extract information from heterogeneous networks. These approaches, however, are less concerned with potential noisy information in heterogeneous networks and more focused on the extent of information extraction in those networks. Based on this, a potential DTI predictive network model called FBRWPC is proposed in this paper. It uses a fine-grained similarity selection program to first integrate similarity on similar networks and then a bidirectional random walk graph embedding learning method with restart to obtain an updated drug target interaction matrix. Through the use of similarity selection and fine-grained selection similarity integration, the framework can effectively filter out the noise present in heterogeneous networks and enhance the model's prediction performance. The experimental findings demonstrate that, even after being split up into four distinct types of data sets, FBRWPC can still retain great prediction performance, a sign of the model's resilience and good generalization.
Collapse
Affiliation(s)
- YaPing Wang
- School of Mathematics, Physics and Statistics, Institute for Frontier Medical Technology, Center of Intelligent Computing and Applied Statistics, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - ZhiXiang Yin
- School of Mathematics, Physics and Statistics, Institute for Frontier Medical Technology, Center of Intelligent Computing and Applied Statistics, Shanghai University of Engineering Science, Shanghai, 201620, China.
| |
Collapse
|
17
|
Yu H, Xu WX, Tan T, Liu Z, Shi JY. Prediction of drug-target binding affinity based on multi-scale feature fusion. Comput Biol Med 2024; 178:108699. [PMID: 38870725 DOI: 10.1016/j.compbiomed.2024.108699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/05/2024] [Accepted: 06/01/2024] [Indexed: 06/15/2024]
Abstract
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/.
Collapse
Affiliation(s)
- Hui Yu
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Wen-Xin Xu
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Tian Tan
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Zun Liu
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Jian-Yu Shi
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
18
|
Goles M, Daza A, Cabas-Mora G, Sarmiento-Varón L, Sepúlveda-Yañez J, Anvari-Kazemabad H, Davari MD, Uribe-Paredes R, Olivera-Nappa Á, Navarrete MA, Medina-Ortiz D. Peptide-based drug discovery through artificial intelligence: towards an autonomous design of therapeutic peptides. Brief Bioinform 2024; 25:bbae275. [PMID: 38856172 PMCID: PMC11163380 DOI: 10.1093/bib/bbae275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/23/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024] Open
Abstract
With their diverse biological activities, peptides are promising candidates for therapeutic applications, showing antimicrobial, antitumour and hormonal signalling capabilities. Despite their advantages, therapeutic peptides face challenges such as short half-life, limited oral bioavailability and susceptibility to plasma degradation. The rise of computational tools and artificial intelligence (AI) in peptide research has spurred the development of advanced methodologies and databases that are pivotal in the exploration of these complex macromolecules. This perspective delves into integrating AI in peptide development, encompassing classifier methods, predictive systems and the avant-garde design facilitated by deep-generative models like generative adversarial networks and variational autoencoders. There are still challenges, such as the need for processing optimization and careful validation of predictive models. This work outlines traditional strategies for machine learning model construction and training techniques and proposes a comprehensive AI-assisted peptide design and validation pipeline. The evolving landscape of peptide design using AI is emphasized, showcasing the practicality of these methods in expediting the development and discovery of novel peptides within the context of peptide-based drug discovery.
Collapse
Affiliation(s)
- Montserrat Goles
- Departamento de Ingeniería en Computación, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
- Departamento de Ingeniería Química, Biotecnología y Materiales, Universidad de Chile, Beauchef 851, 8370456, Santiago, Chile
| | - Anamaría Daza
- Centre for Biotechnology and Bioengineering, CeBiB, Universidad de Chile, Beauchef 851, 8370456, Santiago, Chile
| | - Gabriel Cabas-Mora
- Departamento de Ingeniería en Computación, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
| | - Lindybeth Sarmiento-Varón
- Centro Asistencial de Docencia e Investigación, CADI, Universidad de Magallanes, Av. Los Flamencos 01364, 6210005, Punta Arenas, Chile
| | - Julieta Sepúlveda-Yañez
- Facultad de Ciencias de la Salud, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
| | - Hoda Anvari-Kazemabad
- Departamento de Ingeniería en Computación, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
| | - Mehdi D Davari
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120, Halle, Germany
| | - Roberto Uribe-Paredes
- Departamento de Ingeniería en Computación, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
| | - Álvaro Olivera-Nappa
- Centre for Biotechnology and Bioengineering, CeBiB, Universidad de Chile, Beauchef 851, 8370456, Santiago, Chile
| | - Marcelo A Navarrete
- Centro Asistencial de Docencia e Investigación, CADI, Universidad de Magallanes, Av. Los Flamencos 01364, 6210005, Punta Arenas, Chile
- Escuela de Medicina, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
| | - David Medina-Ortiz
- Departamento de Ingeniería en Computación, Universidad de Magallanes, Av. Pdte. Manuel Bulnes 01855, 6210427, Punta Arenas, Chile
- Centre for Biotechnology and Bioengineering, CeBiB, Universidad de Chile, Beauchef 851, 8370456, Santiago, Chile
| |
Collapse
|
19
|
Zhang Y, Deng Z, Xu X, Feng Y, Junliang S. Application of Artificial Intelligence in Drug-Drug Interactions Prediction: A Review. J Chem Inf Model 2024; 64:2158-2173. [PMID: 37458400 DOI: 10.1021/acs.jcim.3c00582] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Drug-drug interactions (DDI) are a critical aspect of drug research that can have adverse effects on patients and can lead to serious consequences. Predicting these events accurately can significantly improve clinicians' ability to make better decisions and establish optimal treatment regimens. However, manually detecting these interactions is time-consuming and labor-intensive. Utilizing the advancements in Artificial Intelligence (AI) is essential for achieving accurate forecasts of DDIs. In this review, DDI prediction tasks are classified into three types according to the type of DDI prediction: undirected DDI prediction, DDI events prediction, and Asymmetric DDI prediction. The paper then reviews the progress of AI for each of these three prediction tasks in DDI and provides a summary of the data sets used as well as the representative methods used in these three prediction directions. In this review, we aim to provide a comprehensive overview of drug interaction prediction. The first section introduces commonly used databases and presents an overview of current research advancements and techniques across three domains of DDI. Additionally, we introduce classical machine learning techniques for predicting undirected drug interactions and provide a timeline for the progression of the predicted drug interaction events. At last, we debate the difficulties and prospects of AI approaches at predicting DDI, emphasizing their potential for improving clinical decision-making and patient outcomes.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao,266000,China
| | - Zengqian Deng
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao,266000,China
| | - Xiaoyu Xu
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao,266000,China
| | - Yinfei Feng
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao,266000,China
| | - Shang Junliang
- School of Information Science and Engineering, Qufu Normal University, Rizhao, 276800, China
| |
Collapse
|
20
|
Liu Z, Chen Q, Lan W, Lu H, Zhang S. SSLDTI: A novel method for drug-target interaction prediction based on self-supervised learning. Artif Intell Med 2024; 149:102778. [PMID: 38462280 DOI: 10.1016/j.artmed.2024.102778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 12/01/2023] [Accepted: 01/14/2024] [Indexed: 03/12/2024]
Abstract
Many computational methods have been proposed to identify potential drug-target interactions (DTIs) to expedite drug development. Graph neural network (GNN) methods are considered to be one of the most effective approaches. However, shallow GNN methods can only aggregate local information from nodes. Also, deep GNN methods may result in over-smoothing while obtaining long-distance neighbourhood information. As a result, existing GNN methods struggle to extract the complete features of the graph. Additionally, the number of known DTIs is insufficient, and there are far more unknown drug-target pairs than known DTIs, leading to class imbalance. This article proposes a model that combines graph autoencoder and self-supervised learning to accurately encode multilevel features of graphs using only a small number of labelled samples. We introduce a positive sample compensation coefficient to the objective function to mitigate the impact of class imbalance. Experiments on two datasets demonstrated that our model outperforms the four baseline methods, and the new DTIs predicted by the SSLDTI model were verified by the DrugBank database.
Collapse
Affiliation(s)
- Zhixian Liu
- School of Electronics and Information Engineering, Beibu Gulf University, Qinzhou, Guangxi, China
| | - Qingfeng Chen
- School of Computer, Electronic and Information, Guangxi University, Nanning, Guangxi, China.
| | - Wei Lan
- School of Computer, Electronic and Information, Guangxi University, Nanning, Guangxi, China
| | - Huihui Lu
- School of Electronics and Information Engineering, Beibu Gulf University, Qinzhou, Guangxi, China
| | - Shichao Zhang
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China.
| |
Collapse
|
21
|
Wei J, Lu L, Shen T. Predicting drug-protein interactions by preserving the graph information of multi source data. BMC Bioinformatics 2024; 25:10. [PMID: 38177981 PMCID: PMC10768380 DOI: 10.1186/s12859-023-05620-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/15/2023] [Indexed: 01/06/2024] Open
Abstract
Examining potential drug-target interactions (DTIs) is a pivotal component of drug discovery and repurposing. Recently, there has been a significant rise in the use of computational techniques to predict DTIs. Nevertheless, previous investigations have predominantly concentrated on assessing either the connections between nodes or the consistency of the network's topological structure in isolation. Such one-sided approaches could severely hinder the accuracy of DTI predictions. In this study, we propose a novel method called TTGCN, which combines heterogeneous graph convolutional neural networks (GCN) and graph attention networks (GAT) to address the task of DTI prediction. TTGCN employs a two-tiered feature learning strategy, utilizing GAT and residual GCN (R-GCN) to extract drug and target embeddings from the diverse network, respectively. These drug and target embeddings are then fused through a mean-pooling layer. Finally, we employ an inductive matrix completion technique to forecast DTIs while preserving the network's node connectivity and topological structure. Our approach demonstrates superior performance in terms of area under the curve and area under the precision-recall curve in experimental comparisons, highlighting its significant advantages in predicting DTIs. Furthermore, case studies provide additional evidence of its ability to identify potential DTIs.
Collapse
Affiliation(s)
- Jiahao Wei
- School of Mathematical Sciences, Guizhou Normal University, Guiyang, 550025, China
| | - Linzhang Lu
- School of Mathematical Sciences, Guizhou Normal University, Guiyang, 550025, China.
- School of Mathematical Sciences, Xiamen University, Xiamen, 361005, China.
| | - Tie Shen
- Key Laboratory of Information and Computing Science Guizhou Province, Guizhou Normal University, Guizhou, 550001, China.
| |
Collapse
|
22
|
Wang W, Yu M, Sun B, Li J, Liu D, Zhang H, Wang X, Zhou Y. SMGCN: Multiple Similarity and Multiple Kernel Fusion Based Graph Convolutional Neural Network for Drug-Target Interactions Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:143-154. [PMID: 38051618 DOI: 10.1109/tcbb.2023.3339645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Accurately identifying potential drug-target interactions (DTIs) is a critical step in accelerating drug discovery. Despite many studies that have been conducted over the past decades, detecting DTIs remains a highly challenging and complicated process. Therefore, we propose a novel method called SMGCN, which combines multiple similarity and multiple kernel fusion based on Graph Convolutional Network (GCN) to predict DTIs. In order to capture the features of the network structure and fully explore direct or indirect relationships between nodes, we propose the method of multiple similarity, which combines similarity fusion matrices with Random Walk with Restart (RWR) and cosine similarity. Then, we use GCN to extract multi-layer low-dimensional embedding features. Unlike traditional GCN methods, we incorporate Multiple Kernel Learning (MKL). Finally, we use the Dual Laplace Regularized Least Squares method to predict novel DTIs through combinatorial kernels in drug and target spaces. We conduct experiments on a golden standard dataset, and demonstrate the effectiveness of our proposed model in predicting DTIs through showing significant improvements in Area Under the Curve (AUC) and Area Under the Precision-Recall Curve (AUPR). In addition, our model can also discover some new DTIs, which can be verified by the KEGG BRITE Database and relevant literature.
Collapse
|
23
|
Son J, Kim D. Applying network link prediction in drug discovery: an overview of the literature. Expert Opin Drug Discov 2024; 19:43-56. [PMID: 37794688 DOI: 10.1080/17460441.2023.2267020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/02/2023] [Indexed: 10/06/2023]
Abstract
INTRODUCTION Network representation can give a holistic view of relationships for biomedical entities through network topology. Link prediction estimates the probability of link formation between the pair of unconnected nodes. In the drug discovery process, the link prediction method not only enables the detection of connectivity patterns but also predicts the effects of one biomedical entity to multiple entities simultaneously and vice versa, which is useful for many applications. AREAS COVERED The authors provide a comprehensive overview of network link prediction in drug discovery. Link prediction methodologies such as similarity-based approaches, embedding-based approaches, probabilistic model-based approaches, and preprocessing methods are summarized with examples. In addition to describing their properties and limitations, the authors discuss the applications of link prediction in drug discovery based on the relationship between biomedical concepts. EXPERT OPINION Link prediction is a powerful method to infer the existence of novel relationships in drug discovery. However, link prediction has been hampered by the sparsity of data and the lack of negative links in biomedical networks. With preprocessing to balance positive and negative samples and the collection of more data, the authors believe it is possible to develop more reliable link prediction methods that can become invaluable tools for successful drug discovery.
Collapse
Affiliation(s)
- Jeongtae Son
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Dongsup Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
24
|
Abstract
Knowledge graphs represent information in the form of entities and relationships between those entities. Such a representation has multiple potential applications in drug discovery, including democratizing access to biomedical data, contextualizing or visualizing that data, and generating novel insights through the application of machine learning approaches. Knowledge graphs put data into context and therefore offer the opportunity to generate explainable predictions, which is a key topic in contemporary artificial intelligence. In this chapter, we outline some of the factors that need to be considered when constructing biomedical knowledge graphs, examine recent advances in mining such systems to gain insights for drug discovery, and identify potential future areas for further development.
Collapse
Affiliation(s)
- Tim James
- Evotec (UK) Ltd., Abingdon, Oxfordshire, UK.
| | | |
Collapse
|
25
|
Djeddi WE, Hermi K, Ben Yahia S, Diallo G. Advancing drug-target interaction prediction: a comprehensive graph-based approach integrating knowledge graph embedding and ProtBert pretraining. BMC Bioinformatics 2023; 24:488. [PMID: 38114937 PMCID: PMC10731821 DOI: 10.1186/s12859-023-05593-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/30/2023] [Indexed: 12/21/2023] Open
Abstract
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.
Collapse
Affiliation(s)
- Warith Eddine Djeddi
- LR11ES14, Faculty of Sciences of Tunis, University of Tunis El Manar, Campus Universitaire, 2092, Tunis, Tunisia.
- High Institute of Informatics in Kef, University of Jendouba, Saleh Ayech, 8189, Jendouba, Tunisia.
| | - Khalil Hermi
- High Institute of Informatics in Kef, University of Jendouba, Saleh Ayech, 8189, Jendouba, Tunisia
| | - Sadok Ben Yahia
- Department of Software Science, Tallinn University of Technology, Ehitajate tee-5, 12618, Tallinn, Estonia
- The Maersk Mc-Kinney Moller Institute, Southern Syddansk Universitet, Alsion 2, 6400, Sønderborg, Denmark
| | - Gayo Diallo
- Bordeaux Population Health Inserm 1219, University of Bordeaux, rue Léo Saignat, 33000, Bordeaux, France
| |
Collapse
|
26
|
Daza D, Alivanistos D, Mitra P, Pijnenburg T, Cochez M, Groth P. BioBLP: a modular framework for learning on multimodal biomedical knowledge graphs. J Biomed Semantics 2023; 14:20. [PMID: 38066573 PMCID: PMC10709903 DOI: 10.1186/s13326-023-00301-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Knowledge graphs (KGs) are an important tool for representing complex relationships between entities in the biomedical domain. Several methods have been proposed for learning embeddings that can be used to predict new links in such graphs. Some methods ignore valuable attribute data associated with entities in biomedical KGs, such as protein sequences, or molecular graphs. Other works incorporate such data, but assume that entities can be represented with the same data modality. This is not always the case for biomedical KGs, where entities exhibit heterogeneous modalities that are central to their representation in the subject domain. OBJECTIVE We aim to understand how to incorporate multimodal data into biomedical KG embeddings, and analyze the resulting performance in comparison with traditional methods. We propose a modular framework for learning embeddings in KGs with entity attributes, that allows encoding attribute data of different modalities while also supporting entities with missing attributes. We additionally propose an efficient pretraining strategy for reducing the required training runtime. We train models using a biomedical KG containing approximately 2 million triples, and evaluate the performance of the resulting entity embeddings on the tasks of link prediction, and drug-protein interaction prediction, comparing against methods that do not take attribute data into account. RESULTS In the standard link prediction evaluation, the proposed method results in competitive, yet lower performance than baselines that do not use attribute data. When evaluated in the task of drug-protein interaction prediction, the method compares favorably with the baselines. Further analyses show that incorporating attribute data does outperform baselines over entities below a certain node degree, comprising approximately 75% of the diseases in the graph. We also observe that optimizing attribute encoders is a challenging task that increases optimization costs. Our proposed pretraining strategy yields significantly higher performance while reducing the required training runtime. CONCLUSION BioBLP allows to investigate different ways of incorporating multimodal biomedical data for learning representations in KGs. With a particular implementation, we find that incorporating attribute data does not consistently outperform baselines, but improvements are obtained on a comparatively large subset of entities below a specific node-degree. Our results indicate a potential for improved performance in scientific discovery tasks where understudied areas of the KG would benefit from link prediction methods.
Collapse
Affiliation(s)
- Daniel Daza
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- University of Amsterdam, Amsterdam, The Netherlands.
- Discovery Lab, Elsevier, Amsterdam, The Netherlands.
| | - Dimitrios Alivanistos
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Discovery Lab, Elsevier, Amsterdam, The Netherlands.
| | | | | | - Michael Cochez
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Discovery Lab, Elsevier, Amsterdam, The Netherlands
| | - Paul Groth
- University of Amsterdam, Amsterdam, The Netherlands
- Discovery Lab, Elsevier, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Zhu Z, Yao Z, Zheng X, Qi G, Li Y, Mazur N, Gao X, Gong Y, Cong B. Drug-target affinity prediction method based on multi-scale information interaction and graph optimization. Comput Biol Med 2023; 167:107621. [PMID: 37907030 DOI: 10.1016/j.compbiomed.2023.107621] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023]
Abstract
Drug-target affinity (DTA) prediction as an emerging and effective method is widely applied to explore the strength of drug-target interactions in drug development research. By predicting these interactions, researchers can assess the potential efficacy and safety of candidate drugs at an early stage, narrowing down the search space for therapeutic targets and accelerating the discovery and development of new drugs. However, existing DTA prediction models mainly use graphical representations of drug molecules, which lack information on interactions between individual substructures, thus affecting prediction accuracy and model interpretability. Therefore, transformer and diffusion on drug graphs in DTA prediction (TDGraphDTA) are introduced to predict drug-target interactions using multi-scale information interaction and graph optimization. An interactive module is integrated into feature extraction of drug and target features at different granularity levels. A diffusion model-based graph optimization module is proposed to improve the representation of molecular graph structures and enhance the interpretability of graph representations while obtaining optimal feature representations. In addition, TDGraphDTA improves the accuracy and reliability of predictions by capturing relationships and contextual information between molecular substructures. The performance of the proposed TDGraphDTA in DTA prediction was verified on three publicly available benchmark datasets (Davis, Metz, and KIBA). Compared with state-of-the-art baseline models, it achieved better results in terms of consistency index, R-squared, etc. Furthermore, compared with some existing methods, the proposed TDGraphDTA is demonstrated to have better structure capturing capabilities by visualizing the feature capturing capabilities of the model using Grad-AAM toxicity labels in the ToxCast dataset. The corresponding source codes are available at https://github.com/Lamouryz/TDGraph.
Collapse
Affiliation(s)
- Zhiqin Zhu
- College of Automation, Chongqing University of Posts and Telecommunications, Chongqing, 400065, China.
| | - Zheng Yao
- College of Automation, Chongqing University of Posts and Telecommunications, Chongqing, 400065, China.
| | - Xin Zheng
- College of Automation, Chongqing University of Posts and Telecommunications, Chongqing, 400065, China.
| | - Guanqiu Qi
- Computer Information Systems Department, State University of New York at Buffalo State, Buffalo, NY 14222, USA.
| | - Yuanyuan Li
- College of Automation, Chongqing University of Posts and Telecommunications, Chongqing, 400065, China.
| | - Neal Mazur
- Computer Information Systems Department, State University of New York at Buffalo State, Buffalo, NY 14222, USA.
| | - Xinbo Gao
- College of Automation, Chongqing University of Posts and Telecommunications, Chongqing, 400065, China.
| | - Yifei Gong
- Faculty of applied science & engineering, the Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto at Toronto, ON M5S, Canada.
| | - Baisen Cong
- Diagnostics Digital, DH(Shanghai) Diagnostics Co, Ltd, a Danaher company, Shanghai, 200335, China.
| |
Collapse
|
28
|
Su Y, Hu Z, Wang F, Bin Y, Zheng C, Li H, Chen H, Zeng X. AMGDTI: drug-target interaction prediction based on adaptive meta-graph learning in heterogeneous network. Brief Bioinform 2023; 25:bbad474. [PMID: 38145949 PMCID: PMC10749791 DOI: 10.1093/bib/bbad474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/10/2023] [Accepted: 11/30/2023] [Indexed: 12/27/2023] Open
Abstract
Prediction of drug-target interactions (DTIs) is essential in medicine field, since it benefits the identification of molecular structures potentially interacting with drugs and facilitates the discovery and reposition of drugs. Recently, much attention has been attracted to network representation learning to learn rich information from heterogeneous data. Although network representation learning algorithms have achieved success in predicting DTI, several manually designed meta-graphs limit the capability of extracting complex semantic information. To address the problem, we introduce an adaptive meta-graph-based method, termed AMGDTI, for DTI prediction. In the proposed AMGDTI, the semantic information is automatically aggregated from a heterogeneous network by training an adaptive meta-graph, thereby achieving efficient information integration without requiring domain knowledge. The effectiveness of the proposed AMGDTI is verified on two benchmark datasets. Experimental results demonstrate that the AMGDTI method overall outperforms eight state-of-the-art methods in predicting DTI and achieves the accurate identification of novel DTIs. It is also verified that the adaptive meta-graph exhibits flexibility and effectively captures complex fine-grained semantic information, enabling the learning of intricate heterogeneous network topology and the inference of potential drug-target relationship.
Collapse
Affiliation(s)
- Yansen Su
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei, 230601, China
| | - Zhiyang Hu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei, 230601, China
| | - Fei Wang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei, 230601, China
| | - Yannan Bin
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei, 230601, China
| | - Chunhou Zheng
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei, 230601, China
| | - Haitao Li
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei, 230601, China
| | - Haowen Chen
- College of Computer Science and Electronic Engineering, Hunan University, Hunan, 410082, China
| | - Xiangxiang Zeng
- College of Computer Science and Electronic Engineering, Hunan University, Hunan, 410082, China
| |
Collapse
|
29
|
Samantray D, Tanwar AS, Murali TS, Brand A, Satyamoorthy K, Paul B. A Comprehensive Bioinformatics Resource Guide for Genome-Based Antimicrobial Resistance Studies. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2023; 27:445-460. [PMID: 37861712 DOI: 10.1089/omi.2023.0140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
The use of high-throughput sequencing technologies and bioinformatic tools has greatly transformed microbial genome research. With the help of sophisticated computational tools, it has become easier to perform whole genome assembly, identify and compare different species based on their genomes, and predict the presence of genes responsible for proteins, antimicrobial resistance, and toxins. These bioinformatics resources are likely to continuously improve in quality, become more user-friendly to analyze the multiple genomic data, efficient in generating information and translating it into meaningful knowledge, and enhance our understanding of the genetic mechanism of AMR. In this manuscript, we provide an essential guide for selecting the popular resources for microbial research, such as genome assembly and annotation, antibiotic resistance gene profiling, identification of virulence factors, and drug interaction studies. In addition, we discuss the best practices in computer-oriented microbial genome research, emerging trends in microbial genomic data analysis, integration of multi-omics data, the appropriate use of machine-learning algorithms, and open-source bioinformatics resources for genome data analytics.
Collapse
Affiliation(s)
- Debyani Samantray
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Ankit Singh Tanwar
- United Nations University-Maastricht Economic and Social Research Institute on Innovation and Technology (UNU-MERIT), Maastricht, The Netherlands
- Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, The Netherlands
| | - Thokur Sreepathy Murali
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Angela Brand
- United Nations University-Maastricht Economic and Social Research Institute on Innovation and Technology (UNU-MERIT), Maastricht, The Netherlands
- Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, The Netherlands
- Department of Health Information, Prasanna School of Public Health (PSPH), Manipal Academy of Higher Education, Manipal, India
| | - Kapaettu Satyamoorthy
- SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara (SDM) University, Dharwad, India
| | - Bobby Paul
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
30
|
Wang L, Zhou Y, Chen Q. AMMVF-DTI: A Novel Model Predicting Drug-Target Interactions Based on Attention Mechanism and Multi-View Fusion. Int J Mol Sci 2023; 24:14142. [PMID: 37762445 PMCID: PMC10531525 DOI: 10.3390/ijms241814142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Accurate identification of potential drug-target interactions (DTIs) is a crucial task in drug development and repositioning. Despite the remarkable progress achieved in recent years, improving the performance of DTI prediction still presents significant challenges. In this study, we propose a novel end-to-end deep learning model called AMMVF-DTI (attention mechanism and multi-view fusion), which leverages a multi-head self-attention mechanism to explore varying degrees of interaction between drugs and target proteins. More importantly, AMMVF-DTI extracts interactive features between drugs and proteins from both node-level and graph-level embeddings, enabling a more effective modeling of DTIs. This advantage is generally lacking in existing DTI prediction models. Consequently, when compared to many of the start-of-the-art methods, AMMVF-DTI demonstrated excellent performance on the human, C. elegans, and DrugBank baseline datasets, which can be attributed to its ability to incorporate interactive information and mine features from both local and global structures. The results from additional ablation experiments also confirmed the importance of each module in our AMMVF-DTI model. Finally, a case study is presented utilizing our model for COVID-19-related DTI prediction. We believe the AMMVF-DTI model can not only achieve reasonable accuracy in DTI prediction, but also provide insights into the understanding of potential interactions between drugs and targets.
Collapse
|
31
|
Sun J, Xu M, Ru J, James-Bott A, Xiong D, Wang X, Cribbs AP. Small molecule-mediated targeting of microRNAs for drug discovery: Experiments, computational techniques, and disease implications. Eur J Med Chem 2023; 257:115500. [PMID: 37262996 PMCID: PMC11554572 DOI: 10.1016/j.ejmech.2023.115500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/05/2023] [Accepted: 05/15/2023] [Indexed: 06/03/2023]
Abstract
Small molecules have been providing medical breakthroughs for human diseases for more than a century. Recently, identifying small molecule inhibitors that target microRNAs (miRNAs) has gained importance, despite the challenges posed by labour-intensive screening experiments and the significant efforts required for medicinal chemistry optimization. Numerous experimentally-verified cases have demonstrated the potential of miRNA-targeted small molecule inhibitors for disease treatment. This new approach is grounded in their posttranscriptional regulation of the expression of disease-associated genes. Reversing dysregulated gene expression using this mechanism may help control dysfunctional pathways. Furthermore, the ongoing improvement of algorithms has allowed for the integration of computational strategies built on top of laboratory-based data, facilitating a more precise and rational design and discovery of lead compounds. To complement the use of extensive pharmacogenomics data in prioritising potential drugs, our previous work introduced a computational approach based on only molecular sequences. Moreover, various computational tools for predicting molecular interactions in biological networks using similarity-based inference techniques have been accumulated in established studies. However, there are a limited number of comprehensive reviews covering both computational and experimental drug discovery processes. In this review, we outline a cohesive overview of both biological and computational applications in miRNA-targeted drug discovery, along with their disease implications and clinical significance. Finally, utilizing drug-target interaction (DTIs) data from DrugBank, we showcase the effectiveness of deep learning for obtaining the physicochemical characterization of DTIs.
Collapse
Affiliation(s)
- Jianfeng Sun
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK.
| | - Miaoer Xu
- Department of Biology, Emory University, Atlanta, GA, 30322, USA
| | - Jinlong Ru
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, 85354, Germany
| | - Anna James-Bott
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK
| | - Dapeng Xiong
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA; Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Xia Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| | - Adam P Cribbs
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK.
| |
Collapse
|
32
|
El-Sheref EM, Bräse S, Tawfeek HN, Alasmary FA, Youssif BGM. Synthesis, Antioxidant and Antiproliferative Actions of 4-(1,2,3-Triazol-1-yl)quinolin-2(1 H)-ones as Multi-Target Inhibitors. Int J Mol Sci 2023; 24:13300. [PMID: 37686105 PMCID: PMC10488242 DOI: 10.3390/ijms241713300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/19/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
The reaction of 4-azido-quinolin-2(1H)-ones 1a-e with the active methylene compounds pentane-2,4-dione (2a), 1,3-diphenylpropane-1,3-dione (2b), and K2CO3 was investigated in this study. This approach afforded 4-(1,2,3-triazol-1-yl)quinolin-2(1H)-ones 3a-j in high yields and purity. All newly synthesized products' structures were identified. Compounds 3a-j were tested for antiproliferative activity against a panel of four cancer cell lines. In comparison to the reference erlotinib (GI50 = 33), compounds 3f-j were the most potent derivatives, with GI50 values ranging from 22 nM to 31 nM. The most effective antiproliferative derivatives, 3f-j, were subsequently investigated as possible multi-target inhibitors of EGFR, BRAFV600E, and EGFRT790M. Compound 3h was the most potent inhibitor of the studied molecular targets, with IC50 values of 57 nM, 68 nM, and 9.70 nM, respectively. The apoptotic assay results demonstrated that compounds 3g and 3h function as caspase-3, 8, and Bax activators as well as down-regulators of the antiapoptotic Bcl2, and hence can be classified as apoptotic inducers. Finally, compounds 3g and 3h displayed promising antioxidant activity at 10 µM, with DPPH radical scavenging of 70.6% and 73.5%, respectively, compared to Trolox (77.6%).
Collapse
Affiliation(s)
- Essmat M. El-Sheref
- Chemistry Department, Faculty of Science, Minia University, El-Minia 61519, Egypt;
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, IBCS-FMS, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Hendawy N. Tawfeek
- Chemistry Department, Faculty of Science, Minia University, El-Minia 61519, Egypt;
- Unit of Occupational of Safety and Health, Administration Office of Minia University, El-Minia 61519, Egypt
| | - Fatmah Ali Alasmary
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Bahaa G. M. Youssif
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt;
| |
Collapse
|
33
|
Li X, Yang Q, Luo G, Xu L, Dong W, Wang W, Dong S, Wang K, Xuan P, Gao X. SAGDTI: self-attention and graph neural network with multiple information representations for the prediction of drug-target interactions. BIOINFORMATICS ADVANCES 2023; 3:vbad116. [PMID: 38282612 PMCID: PMC10818136 DOI: 10.1093/bioadv/vbad116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/31/2023] [Accepted: 08/24/2023] [Indexed: 01/30/2024]
Abstract
Motivation Accurate identification of target proteins that interact with drugs is a vital step in silico, which can significantly foster the development of drug repurposing and drug discovery. In recent years, numerous deep learning-based methods have been introduced to treat drug-target interaction (DTI) prediction as a classification task. The output of this task is binary identification suggesting the absence or presence of interactions. However, existing studies often (i) neglect the unique molecular attributes when embedding drugs and proteins, and (ii) determine the interaction of drug-target pairs without considering biological interaction information. Results In this study, we propose an end-to-end attention-derived method based on the self-attention mechanism and graph neural network, termed SAGDTI. The aim of this method is to overcome the aforementioned drawbacks in the identification of DTI. SAGDTI is the first method to sufficiently consider the unique molecular attribute representations for both drugs and targets in the input form of the SMILES sequences and three-dimensional structure graphs. In addition, our method aggregates the feature attributes of biological information between drugs and targets through multi-scale topologies and diverse connections. Experimental results illustrate that SAGDTI outperforms existing prediction models, which benefit from the unique molecular attributes embedded by atom-level attention and biological interaction information representation aggregated by node-level attention. Moreover, a case study on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) shows that our model is a powerful tool for identifying DTIs in real life. Availability and implementation The data and codes underlying this article are available in Github at https://github.com/lixiaokun2020/SAGDTI.
Collapse
Affiliation(s)
- Xiaokun Li
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Harbin 150090, China
| | - Qiang Yang
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Harbin 150090, China
| | - Gongning Luo
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Long Xu
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Harbin 150090, China
| | - Weihe Dong
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Harbin 150090, China
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Wei Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Suyu Dong
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Kuanquan Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ping Xuan
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
- Department of Computer Science, School of Engineering, Shantou University, Shantou 515063, China
| | - Xin Gao
- Computer, Electrical and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal 23955, Saudi Arabia
| |
Collapse
|
34
|
Hu X, Yin Z, Zeng Z, Peng Y. Prediction of miRNA-Disease Associations by Cascade Forest Model Based on Stacked Autoencoder. Molecules 2023; 28:5013. [PMID: 37446675 DOI: 10.3390/molecules28135013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/23/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Numerous pieces of evidence have indicated that microRNA (miRNA) plays a crucial role in a series of significant biological processes and is closely related to complex disease. However, the traditional biological experimental methods used to verify disease-related miRNAs are inefficient and expensive. Thus, it is necessary to design some excellent approaches to improve efficiency. In this work, a novel method (CFSAEMDA) is proposed for the prediction of unknown miRNA-disease associations (MDAs). Specifically, we first capture the interactive features of miRNA and disease by integrating multi-source information. Then, the stacked autoencoder is applied for obtaining the underlying feature representation. Finally, the modified cascade forest model is employed to complete the final prediction. The experimental results present that the AUC value obtained by our method is 97.67%. The performance of CFSAEMDA is superior to several of the latest methods. In addition, case studies conducted on lung neoplasms, breast neoplasms and hepatocellular carcinoma further show that the CFSAEMDA method may be regarded as a utility approach to infer unknown disease-miRNA relationships.
Collapse
Affiliation(s)
- Xiang Hu
- Center of Intelligent Computing and Applied Statistics, School of Mathematics, Physics and Statistics, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Zhixiang Yin
- Center of Intelligent Computing and Applied Statistics, School of Mathematics, Physics and Statistics, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Zhiliang Zeng
- Center of Intelligent Computing and Applied Statistics, School of Mathematics, Physics and Statistics, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Yu Peng
- Center of Intelligent Computing and Applied Statistics, School of Mathematics, Physics and Statistics, Shanghai University of Engineering Science, Shanghai 201620, China
| |
Collapse
|
35
|
Amiri Souri E, Chenoweth A, Karagiannis SN, Tsoka S. Drug repurposing and prediction of multiple interaction types via graph embedding. BMC Bioinformatics 2023; 24:202. [PMID: 37193964 DOI: 10.1186/s12859-023-05317-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/30/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND Finding drugs that can interact with a specific target to induce a desired therapeutic outcome is key deliverable in drug discovery for targeted treatment. Therefore, both identifying new drug-target links, as well as delineating the type of drug interaction, are important in drug repurposing studies. RESULTS A computational drug repurposing approach was proposed to predict novel drug-target interactions (DTIs), as well as to predict the type of interaction induced. The methodology is based on mining a heterogeneous graph that integrates drug-drug and protein-protein similarity networks, together with verified drug-disease and protein-disease associations. In order to extract appropriate features, the three-layer heterogeneous graph was mapped to low dimensional vectors using node embedding principles. The DTI prediction problem was formulated as a multi-label, multi-class classification task, aiming to determine drug modes of action. DTIs were defined by concatenating pairs of drug and target vectors extracted from graph embedding, which were used as input to classification via gradient boosted trees, where a model is trained to predict the type of interaction. After validating the prediction ability of DT2Vec+, a comprehensive analysis of all unknown DTIs was conducted to predict the degree and type of interaction. Finally, the model was applied to propose potential approved drugs to target cancer-specific biomarkers. CONCLUSION DT2Vec+ showed promising results in predicting type of DTI, which was achieved via integrating and mapping triplet drug-target-disease association graphs into low-dimensional dense vectors. To our knowledge, this is the first approach that addresses prediction between drugs and targets across six interaction types.
Collapse
Affiliation(s)
- E Amiri Souri
- Department of Informatics, Faculty of Natural, Mathematical and Engineering Sciences, King's College London, Bush House, London, WC2B 4BG, UK
| | - A Chenoweth
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, Guy's Hospital, King's College London, London, SE1 9RT, UK
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, Guy's Cancer Centre, King's College London, London, SE1 9RT, UK
| | - S N Karagiannis
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, Guy's Hospital, King's College London, London, SE1 9RT, UK
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, Guy's Cancer Centre, King's College London, London, SE1 9RT, UK
| | - S Tsoka
- Department of Informatics, Faculty of Natural, Mathematical and Engineering Sciences, King's College London, Bush House, London, WC2B 4BG, UK.
| |
Collapse
|
36
|
Li J, Wang Y, Li Z, Lin H, Wu B. LM-DTI: a tool of predicting drug-target interactions using the node2vec and network path score methods. Front Genet 2023; 14:1181592. [PMID: 37229202 PMCID: PMC10203599 DOI: 10.3389/fgene.2023.1181592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/13/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction: Drug-target interaction (DTI) prediction is a key step in drug function discovery and repositioning. The emergence of large-scale heterogeneous biological networks provides an opportunity to identify drug-related target genes, which led to the development of several computational methods for DTI prediction. Methods: Considering the limitations of conventional computational methods, a novel tool named LM-DTI based on integrated information related to lncRNAs and miRNAs was proposed, which adopted the graph embedding (node2vec) and the network path score methods. First, LM-DTI innovatively constructed a heterogeneous information network containing eight networks composed of four types of nodes (drug, target, lncRNA, and miRNA). Next, the node2vec method was used to obtain feature vectors of drug as well as target nodes, and the path score vector of each drug-target pair was calculated using the DASPfind method. Finally, the feature vectors and path score vectors were merged and input into the XGBoost classifier to predict potential drug-target interactions. Results and Discussion: The 10-fold cross validations evaluate the classification accuracies of the LM-DTI. The prediction performance of LM-DTI in AUPR reached 0.96, which showed a significant improvement compared with those of conventional tools. The validity of LM-DTI has also been verified by manually searching literature and various databases. LM-DTI is scalable and computing efficient; thus representing a powerful drug relocation tool that can be accessed for free at http://www.lirmed.com:5038/lm_dti.
Collapse
Affiliation(s)
- Jianwei Li
- School of Artificial Intelligence, Institute of Computational Medicine, Hebei University of Technology, Tianjin, China
- School of Electronic and Information Engineering, Hebei University of Technology, Tianjin, China
| | - Yinfei Wang
- School of Artificial Intelligence, Institute of Computational Medicine, Hebei University of Technology, Tianjin, China
| | - Zhiguang Li
- School of Artificial Intelligence, Institute of Computational Medicine, Hebei University of Technology, Tianjin, China
| | - Hongxin Lin
- School of Artificial Intelligence, Institute of Computational Medicine, Hebei University of Technology, Tianjin, China
| | - Baoqin Wu
- School of Artificial Intelligence, Institute of Computational Medicine, Hebei University of Technology, Tianjin, China
| |
Collapse
|
37
|
Muniyappan S, Rayan AXA, Varrieth GT. DTiGNN: Learning drug-target embedding from a heterogeneous biological network based on a two-level attention-based graph neural network. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:9530-9571. [PMID: 37161255 DOI: 10.3934/mbe.2023419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
MOTIVATION In vitro experiment-based drug-target interaction (DTI) exploration demands more human, financial and data resources. In silico approaches have been recommended for predicting DTIs to reduce time and cost. During the drug development process, one can analyze the therapeutic effect of the drug for a particular disease by identifying how the drug binds to the target for treating that disease. Hence, DTI plays a major role in drug discovery. Many computational methods have been developed for DTI prediction. However, the existing methods have limitations in terms of capturing the interactions via multiple semantics between drug and target nodes in a heterogeneous biological network (HBN). METHODS In this paper, we propose a DTiGNN framework for identifying unknown drug-target pairs. The DTiGNN first calculates the similarity between the drug and target from multiple perspectives. Then, the features of drugs and targets from each perspective are learned separately by using a novel method termed an information entropy-based random walk. Next, all of the learned features from different perspectives are integrated into a single drug and target similarity network by using a multi-view convolutional neural network. Using the integrated similarity networks, drug interactions, drug-disease associations, protein interactions and protein-disease association, the HBN is constructed. Next, a novel embedding algorithm called a meta-graph guided graph neural network is used to learn the embedding of drugs and targets. Then, a convolutional neural network is employed to infer new DTIs after balancing the sample using oversampling techniques. RESULTS The DTiGNN is applied to various datasets, and the result shows better performance in terms of the area under receiver operating characteristic curve (AUC) and area under precision-recall curve (AUPR), with scores of 0.98 and 0.99, respectively. There are 23,739 newly predicted DTI pairs in total.
Collapse
Affiliation(s)
- Saranya Muniyappan
- Computer Science and Engineering, CEG Campus, Anna University, Tamil Nadu, India
| | | | | |
Collapse
|
38
|
Wang L, Song Y, Wang H, Zhang X, Wang M, He J, Li S, Zhang L, Li K, Cao L. Advances of Artificial Intelligence in Anti-Cancer Drug Design: A Review of the Past Decade. Pharmaceuticals (Basel) 2023; 16:253. [PMID: 37259400 PMCID: PMC9963982 DOI: 10.3390/ph16020253] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/25/2023] [Accepted: 02/06/2023] [Indexed: 10/13/2023] Open
Abstract
Anti-cancer drug design has been acknowledged as a complicated, expensive, time-consuming, and challenging task. How to reduce the research costs and speed up the development process of anti-cancer drug designs has become a challenging and urgent question for the pharmaceutical industry. Computer-aided drug design methods have played a major role in the development of cancer treatments for over three decades. Recently, artificial intelligence has emerged as a powerful and promising technology for faster, cheaper, and more effective anti-cancer drug designs. This study is a narrative review that reviews a wide range of applications of artificial intelligence-based methods in anti-cancer drug design. We further clarify the fundamental principles of these methods, along with their advantages and disadvantages. Furthermore, we collate a large number of databases, including the omics database, the epigenomics database, the chemical compound database, and drug databases. Other researchers can consider them and adapt them to their own requirements.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Kang Li
- Department of Biostatistics, School of Public Health, Harbin Medical University, Harbin 150081, China
| | - Lei Cao
- Department of Biostatistics, School of Public Health, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
39
|
Vemula D, Jayasurya P, Sushmitha V, Kumar YN, Bhandari V. CADD, AI and ML in drug discovery: A comprehensive review. Eur J Pharm Sci 2023; 181:106324. [PMID: 36347444 DOI: 10.1016/j.ejps.2022.106324] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 11/06/2022]
Abstract
Computer-aided drug design (CADD) is an emerging field that has drawn a lot of interest because of its potential to expedite and lower the cost of the drug development process. Drug discovery research is expensive and time-consuming, and it frequently took 10-15 years for a drug to be commercially available. CADD has significantly impacted this area of research. Further, the combination of CADD with Artificial Intelligence (AI), Machine Learning (ML), and Deep Learning (DL) technologies to handle enormous amounts of biological data has reduced the time and cost associated with the drug development process. This review will discuss how CADD, AI, ML, and DL approaches help identify drug candidates and various other steps of the drug discovery process. It will also provide a detailed overview of the different in silico tools used and how these approaches interact.
Collapse
Affiliation(s)
- Divya Vemula
- National Institute of Pharmaceutical Education and Research- Hyderabad, India
| | - Perka Jayasurya
- National Institute of Pharmaceutical Education and Research- Hyderabad, India
| | - Varthiya Sushmitha
- National Institute of Pharmaceutical Education and Research- Hyderabad, India
| | | | - Vasundhra Bhandari
- National Institute of Pharmaceutical Education and Research- Hyderabad, India.
| |
Collapse
|
40
|
Peng Y, Zhao S, Zeng Z, Hu X, Yin Z. LGBMDF: A cascade forest framework with LightGBM for predicting drug-target interactions. Front Microbiol 2023; 13:1092467. [PMID: 36687573 PMCID: PMC9849804 DOI: 10.3389/fmicb.2022.1092467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/07/2022] [Indexed: 01/07/2023] Open
Abstract
Prediction of drug-target interactions (DTIs) plays an important role in drug development. However, traditional laboratory methods to determine DTIs require a lot of time and capital costs. In recent years, many studies have shown that using machine learning methods to predict DTIs can speed up the drug development process and reduce capital costs. An excellent DTI prediction method should have both high prediction accuracy and low computational cost. In this study, we noticed that the previous research based on deep forests used XGBoost as the estimator in the cascade, we applied LightGBM instead of XGBoost to the cascade forest as the estimator, then the estimator group was determined experimentally as three LightGBMs and three ExtraTrees, this new model is called LGBMDF. We conducted 5-fold cross-validation on LGBMDF and other state-of-the-art methods using the same dataset, and compared their Sn, Sp, MCC, AUC and AUPR. Finally, we found that our method has better performance and faster calculation speed.
Collapse
|
41
|
Jamali AA, Kusalik A, Wu FX. NMTF-DTI: A Nonnegative Matrix Tri-factorization Approach With Multiple Kernel Fusion for Drug-Target Interaction Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:586-594. [PMID: 34914594 DOI: 10.1109/tcbb.2021.3135978] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Prediction of drug-target interactions (DTIs) plays a significant role in drug development and drug discovery. Although this task requires a large investment in terms of time and cost, especially when it is performed experimentally, the results are not necessarily significant. Computational DTI prediction is a shortcut to reduce the risks of experimental methods. In this study, we propose an effective approach of nonnegative matrix tri-factorization, referred to as NMTF-DTI, to predict the interaction scores between drugs and targets. NMTF-DTI utilizes multiple kernels (similarity measures) for drugs and targets and Laplacian regularization to boost the prediction performance. The performance of NMTF-DTI is evaluated via cross-validation and is compared with existing DTI prediction methods in terms of the area under the receiver operating characteristic (ROC) curve (AUC) and the area under the precision and recall curve (AUPR). We evaluate our method on four gold standard datasets, comparing to other state-of-the-art methods. Cross-validation and a separate, manually created dataset are used to set parameters. The results show that NMTF-DTI outperforms other competing methods. Moreover, the results of a case study also confirm the superiority of NMTF-DTI.
Collapse
|
42
|
Li Y, Sun C, Wei JM, Liu J. Drug-Protein interaction prediction by correcting the effect of incomplete information in heterogeneous information. Bioinformatics 2022; 38:5073-5080. [PMID: 36111859 DOI: 10.1093/bioinformatics/btac629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/30/2022] [Accepted: 09/15/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Large-scale heterogeneous data provide diverse perspectives for predicting drug-protein interactions (DPIs). However, the available information on molecular interactions and clinical associations related to drugs or proteins is incomplete because there may be unproven interactions and associations. This incomplete information in the available data is presented in the form of non-interaction and non-correlation, which may mislead the prediction model. Existing methods fuse incomplete and complete information without considering their integrity, so the negative effects of incomplete information still exist. RESULTS We develop a network-based DPI prediction method named BRWCP, which uses the complete information network to correct the prediction results acquired by the incomplete information network. By integrating relevant heterogeneous information that may be incomplete, the feature similarities of drugs and proteins are obtained. Combining the feature similarities and known DPIs, an incomplete information-based drug-protein heterogeneous network is constructed. Then, a bidirectional random walk with pruning algorithm is adopted in this heterogeneous network to predict potential DPIs. Next, the predicted DPIs are combined with the chemical fingerprint similarity of drugs and amino acid sequence similarity of proteins to construct the complete information network. The bidirectional random walk with pruning algorithm is applied in the new network to obtain the final prediction results until it converges. Experimental results show that BRWCP is superior to several state-of-the-art DPI prediction methods, and case studies further confirm its ability to tap potential DPIs. AVAILABILITY AND IMPLEMENTATION The code and data used in BRWCP are available at https://github.com/lyfdomain/BRWCP. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Yanfei Li
- College of Computer Science, Nankai University, Tianjin 300071, China.,Institute of Big Data, Nankai University, Tianjin 300071, China
| | - Chang Sun
- College of Computer Science, Nankai University, Tianjin 300071, China.,Institute of Big Data, Nankai University, Tianjin 300071, China
| | - Jin-Mao Wei
- College of Computer Science, Nankai University, Tianjin 300071, China.,Institute of Big Data, Nankai University, Tianjin 300071, China
| | - Jian Liu
- College of Computer Science, Nankai University, Tianjin 300071, China.,Institute of Big Data, Nankai University, Tianjin 300071, China
| |
Collapse
|
43
|
Maghsoudi S, Taghavi Shahraki B, Rameh F, Nazarabi M, Fatahi Y, Akhavan O, Rabiee M, Mostafavi E, Lima EC, Saeb MR, Rabiee N. A review on computer-aided chemogenomics and drug repositioning for rational COVID-19 drug discovery. Chem Biol Drug Des 2022; 100:699-721. [PMID: 36002440 PMCID: PMC9539342 DOI: 10.1111/cbdd.14136] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/07/2022] [Accepted: 08/21/2022] [Indexed: 11/29/2022]
Abstract
Application of materials capable of energy harvesting to increase the efficiency and environmental adaptability is sometimes reflected in the ability of discovery of some traces in an environment-either experimentally or computationally-to enlarge practical application window. The emergence of computational methods, particularly computer-aided drug discovery (CADD), provides ample opportunities for the rapid discovery and development of unprecedented drugs. The expensive and time-consuming process of traditional drug discovery is no longer feasible, for nowadays the identification of potential drug candidates is much easier for therapeutic targets through elaborate in silico approaches, allowing the prediction of the toxicity of drugs, such as drug repositioning (DR) and chemical genomics (chemogenomics). Coronaviruses (CoVs) are cross-species viruses that are able to spread expeditiously from the into new host species, which in turn cause epidemic diseases. In this sense, this review furnishes an outline of computational strategies and their applications in drug discovery. A special focus is placed on chemogenomics and DR as unique and emerging system-based disciplines on CoV drug and target discovery to model protein networks against a library of compounds. Furthermore, to demonstrate the special advantages of CADD methods in rapidly finding a drug for this deadly virus, numerous examples of the recent achievements grounded on molecular docking, chemogenomics, and DR are reported, analyzed, and interpreted in detail. It is believed that the outcome of this review assists developers of energy harvesting materials and systems for detection of future unexpected kinds of CoVs or other variants.
Collapse
Affiliation(s)
- Saeid Maghsoudi
- Faculty of Medicine, Department of Physiology and PathophysiologyUniversity of ManitobaWinnipegManitobaCanada
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba (CHRIM), University of ManitobaWinnipegManitobaCanada
| | | | | | - Masoomeh Nazarabi
- Faculty of Organic Chemistry, Department of ChemistryUniversity of KashanKashanIran
| | - Yousef Fatahi
- Department of Pharmaceutical Nanotechnology, Faculty of PharmacyTehran University of Medical SciencesTehranIran
- Nanotechnology Research Center, Faculty of PharmacyTehran University of Medical SciencesTehranIran
| | - Omid Akhavan
- Department of PhysicsSharif University of TechnologyTehranIran
| | - Mohammad Rabiee
- Biomaterials Group, Department of Biomedical EngineeringAmirkabir University of TechnologyTehranIran
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of MedicineStanfordCaliforniaUSA
- Department of MedicineStanford University School of MedicineStanfordCaliforniaUSA
| | - Eder C. Lima
- Institute of Chemistry, Federal University of Rio Grande Do Sul (UFRGS)Porto AlegreBrazil
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of ChemistryGdańsk University of TechnologyGdańskPoland
| | - Navid Rabiee
- Department of PhysicsSharif University of TechnologyTehranIran
- School of EngineeringMacquarie UniversitySydneyNew South WalesAustralia
- Department of Materials Science and EngineeringPohang University of Science and Technology (POSTECH)PohangSouth Korea
| |
Collapse
|
44
|
An explainable framework for drug repositioning from disease information network. Neurocomputing 2022. [DOI: 10.1016/j.neucom.2022.09.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
45
|
Sun G, Dong D, Dong Z, Zhang Q, Fang H, Wang C, Zhang S, Wu S, Dong Y, Wan Y. Drug repositioning: A bibliometric analysis. Front Pharmacol 2022; 13:974849. [PMID: 36225586 PMCID: PMC9549161 DOI: 10.3389/fphar.2022.974849] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/12/2022] [Indexed: 11/14/2022] Open
Abstract
Drug repurposing has become an effective approach to drug discovery, as it offers a new way to explore drugs. Based on the Science Citation Index Expanded (SCI-E) and Social Sciences Citation Index (SSCI) databases of the Web of Science core collection, this study presents a bibliometric analysis of drug repurposing publications from 2010 to 2020. Data were cleaned, mined, and visualized using Derwent Data Analyzer (DDA) software. An overview of the history and development trend of the number of publications, major journals, major countries, major institutions, author keywords, major contributors, and major research fields is provided. There were 2,978 publications included in the study. The findings show that the United States leads in this area of research, followed by China, the United Kingdom, and India. The Chinese Academy of Science published the most research studies, and NIH ranked first on the h-index. The Icahn School of Medicine at Mt Sinai leads in the average number of citations per study. Sci Rep, Drug Discov. Today, and Brief. Bioinform. are the three most productive journals evaluated from three separate perspectives, and pharmacology and pharmacy are unquestionably the most commonly used subject categories. Cheng, FX; Mucke, HAM; and Butte, AJ are the top 20 most prolific and influential authors. Keyword analysis shows that in recent years, most research has focused on drug discovery/drug development, COVID-19/SARS-CoV-2/coronavirus, molecular docking, virtual screening, cancer, and other research areas. The hotspots have changed in recent years, with COVID-19/SARS-CoV-2/coronavirus being the most popular topic for current drug repurposing research.
Collapse
Affiliation(s)
- Guojun Sun
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Dashun Dong
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Zuojun Dong
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Qian Zhang
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Hui Fang
- Institute of Information Resource, Zhejiang University of Technology, Hangzhou, China
| | - Chaojun Wang
- Hangzhou Aeronautical Sanatorium for Special Service of Chinese Air Force, Hangzhou, China
| | - Shaoya Zhang
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Shuaijun Wu
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Yichen Dong
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yuehua Wan
- Institute of Information Resource, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
46
|
Zhang Y, Wu M, Wang S, Chen W. EFMSDTI: Drug-target interaction prediction based on an efficient fusion of multi-source data. Front Pharmacol 2022; 13:1009996. [PMID: 36210804 PMCID: PMC9538487 DOI: 10.3389/fphar.2022.1009996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Accurate identification of Drug Target Interactions (DTIs) is of great significance for understanding the mechanism of drug treatment and discovering new drugs for disease treatment. Currently, computational methods of DTIs prediction that combine drug and target multi-source data can effectively reduce the cost and time of drug development. However, in multi-source data processing, the contribution of different source data to DTIs is often not considered. Therefore, how to make full use of the contribution of different source data to predict DTIs for efficient fusion is the key to improving the prediction accuracy of DTIs. In this paper, considering the contribution of different source data to DTIs prediction, a DTIs prediction approach based on an effective fusion of drug and target multi-source data is proposed, named EFMSDTI. EFMSDTI first builds 15 similarity networks based on multi-source information networks classified as topological and semantic graphs of drugs and targets according to their biological characteristics. Then, the multi-networks are fused by selective and entropy weighting based on similarity network fusion (SNF) according to their contribution to DTIs prediction. The deep neural networks model learns the embedding of low-dimensional vectors of drugs and targets. Finally, the LightGBM algorithm based on Gradient Boosting Decision Tree (GBDT) is used to complete DTIs prediction. Experimental results show that EFMSDTI has better performance (AUROC and AUPR are 0.982) than several state-of-the-art algorithms. Also, it has a good effect on analyzing the top 1000 prediction results, while 990 of the first 1000DTIs were confirmed. Code and data are available at https://github.com/meng-jie/EFMSDTI.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao, Shandong, China
- College of Computer science and Technology, China University of Petroleum (East China), Qingdao, Shandong, China
- *Correspondence: Yuanyuan Zhang,
| | - Mengjie Wu
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao, Shandong, China
| | - Shudong Wang
- College of Computer science and Technology, China University of Petroleum (East China), Qingdao, Shandong, China
| | - Wei Chen
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao, Shandong, China
| |
Collapse
|
47
|
Liu B, Papadopoulos D, Malliaros FD, Tsoumakas G, Papadopoulos AN. Multiple similarity drug-target interaction prediction with random walks and matrix factorization. Brief Bioinform 2022; 23:6692553. [PMID: 36070659 DOI: 10.1093/bib/bbac353] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/11/2022] [Accepted: 07/27/2022] [Indexed: 11/14/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Bin Liu
- Key Laboratory of Data Engineering and Visual Computing, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
- School of Informatics, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | | | - Fragkiskos D Malliaros
- Paris-Saclay University, CentraleSupélec, Inria, Centre for Visual Computing (CVN), 91190 Gif-Sur-Yvette, France
| | - Grigorios Tsoumakas
- School of Informatics, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | | |
Collapse
|
48
|
Cheng Z, Zhao Q, Li Y, Wang J. IIFDTI: predicting drug-target interactions through interactive and independent features based on attention mechanism. Bioinformatics 2022; 38:4153-4161. [PMID: 35801934 DOI: 10.1093/bioinformatics/btac485] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/02/2022] [Accepted: 07/07/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Identifying drug-target interactions is a crucial step for drug discovery and design. Traditional biochemical experiments are credible to accurately validate drug-target interactions. However, they are also extremely laborious, time-consuming and expensive. With the collection of more validated biomedical data and the advancement of computing technology, the computational methods based on chemogenomics gradually attract more attention, which guide the experimental verifications. RESULTS In this study, we propose an end-to-end deep learning-based method named IIFDTI to predict drug-target interactions (DTIs) based on independent features of drug-target pairs and interactive features of their substructures. First, the interactive features of substructures between drugs and targets are extracted by the bidirectional encoder-decoder architecture. The independent features of drugs and targets are extracted by the graph neural networks and convolutional neural networks, respectively. Then, all extracted features are fused and inputted into fully connected dense layers in downstream tasks for predicting DTIs. IIFDTI takes into account the independent features of drugs/targets and simulates the interactive features of the substructures from the biological perspective. Multiple experiments show that IIFDTI outperforms the state-of-the-art methods in terms of the area under the receiver operating characteristics curve (AUC), the area under the precision-recall curve (AUPR), precision, and recall on benchmark datasets. In addition, the mapped visualizations of attention weights indicate that IIFDTI has learned the biological knowledge insights, and two case studies illustrate the capabilities of IIFDTI in practical applications. AVAILABILITY AND IMPLEMENTATION The data and codes underlying this article are available in Github at https://github.com/czjczj/IIFDTI. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Zhongjian Cheng
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Qichang Zhao
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Yaohang Li
- Department of Computer Science, Old Dominion University, Norfolk, VA 23529, USA
| | - Jianxin Wang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
49
|
GCHN-DTI: Predicting drug-target interactions by graph convolution on heterogeneous networks. Methods 2022; 206:101-107. [PMID: 36058415 DOI: 10.1016/j.ymeth.2022.08.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/17/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022] Open
Abstract
Determining the interaction of drug and target plays a key role in the process of drug development and discovery. The calculation methods can predict new interactions and speed up the process of drug development. In recent studies, the network-based approaches have been proposed to predict drug-target interactions. However, these methods cannot fully utilize the node information from heterogeneous networks. Therefore, we propose a method based on heterogeneous graph convolutional neural network for drug-target interaction prediction, GCHN-DTI (Predicting drug-target interactions by graph convolution on heterogeneous net-works), to predict potential DTIs. GCHN-DTI integrates network information from drug-target interactions, drug-drug interactions, drug-similarities, target-target interactions, and target-similarities. Then, the graph convolution operation is used in the heterogeneous network to obtain the node embedding of the drugs and the targets. Furthermore, we incorporate an attention mechanism between graph convolutional layers to combine node embedding from each layer. Finally, the drug-target interaction score is predicted based on the node embedding of the drugs and the targets. Our model uses fewer network types and achieves higher prediction performance. In addition, the prediction performance of the model will be significantly improved on the dataset with a higher proportion of positive samples. The experimental evaluations show that GCHN-DTI outperforms several state-of-the-art prediction methods.
Collapse
|
50
|
Lian M, Wang X, Du W. Integrated multi-similarity fusion and heterogeneous graph inference for drug-target interaction prediction. Neurocomputing 2022. [DOI: 10.1016/j.neucom.2022.04.104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|