1
|
Zong N, Chowdhury S, Zhou S, Rajaganapathy S, Yu Y, Wang L, Dai Q, Li P, Liu X, Bielinski SJ, Chen J, Chen Y, Cerhan JR. Advancing efficacy prediction for electronic health records based emulated trials in repurposing heart failure therapies. NPJ Digit Med 2025; 8:306. [PMID: 40413299 PMCID: PMC12103528 DOI: 10.1038/s41746-025-01705-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/03/2025] [Indexed: 05/27/2025] Open
Abstract
The complexities inherent in EHR data create discrepancies between real-world evidence and RCTs, posing substantial challenges in determining whether a treatment is likely to have a beneficial impact compared to the standard of care in RCTs. The objective of this study is to enhance the prediction of efficacy direction for repurposed drugs tested in RCTs for heart failure (HF). To achieve this, we propose an efficacy direction prediction framework that integrates drug-target predictions with EHR-based Emulation Trials (ET) to derive surrogate endpoints for prediction using HF prognostic markers. Our validation of the proposed novel drug-target prediction model against the BETA benchmark demonstrates superior performance, surpassing existing baseline algorithms. Furthermore, an evaluation of our framework in identifying 17 repurposed drugs-derived from 266 phase 3 HF RCTs-using data from 59,000 patients at the Mayo Clinic highlights its remarkable predictive accuracy.
Collapse
Affiliation(s)
- Nansu Zong
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA.
| | - Shaika Chowdhury
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Shibo Zhou
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Sivaraman Rajaganapathy
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Yue Yu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Qiying Dai
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Pengyang Li
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Xiaoke Liu
- Division of Community Cardiology, Department of Cardiovascular Medicine, La Crosse, Wisconsin, WI, USA
| | | | - Jun Chen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Yongbin Chen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - James R Cerhan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
2
|
Zheng F, Zhao J, Yuan Z, Gao Y, Li Y, Li Y, Geng Y, Qiang Y. Interpretable drug-target affinity prediction based on pre-trained models' output as embeddings and based on structure-aware cross-attention for feature fusion. Mol Divers 2025:10.1007/s11030-025-11194-7. [PMID: 40279085 DOI: 10.1007/s11030-025-11194-7] [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: 03/13/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025]
Abstract
The characteristics of protein pockets can better capture the interaction information between proteins and small molecules, thereby improving the performance of drug-target interaction (DTI) prediction tasks. However, pocket data typically need to be predicted using software such as AlphaFold, which would entail a massive workload for datasets ranging from tens of thousands to hundreds of thousands of samples. Moreover, feature representation networks for 3D pocket data are computationally intensive. To address this, we propose simulating 3D pocket data using sequence data through feature fusion of two different objects based on structure cross-attention (CASD). Additionally, precise feature representation is a prerequisite for accurately identifying pocket information. We introduce a method that leverages the output of the last layer of a pre-trained model as an embedding layer for training a new model from scratch. This approach not only incorporates prior knowledge from the pre-trained model but also expands model capacity, enabling more accurate feature representation. Furthermore, we enhance the multimodal representation of small molecule compounds using feature fusion based on structure cross-attention for the same object (CASS), further improving feature representation capabilities. Our cross-attention mechanisms operate at the token-level or node-level, allowing fine-grained capture of interactions between amino acids and atoms. This enables the identification of the contribution score of each atom or amino acid to the task, making our model interpretable for drug-target prediction. Experimental validation demonstrates that our model achieves state-of-the-art predictive performance.
Collapse
Affiliation(s)
- Fang Zheng
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, 209 University Street, Yuci District, Jinzhong, 030600, China
| | - Juanjuan Zhao
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, 209 University Street, Yuci District, Jinzhong, 030600, China.
- School of Software, Taiyuan University of Technology, 209 University Street, Yuci District, Jinzhong, 030600, China.
| | - Zihang Yuan
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, 209 University Street, Yuci District, Jinzhong, 030600, China
| | - Yuanchen Gao
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, 209 University Street, Yuci District, Jinzhong, 030600, China
| | - Yafeng Li
- Department of Nephrology, The Fifth Hospital of Shanxi Medical University, No. 29 Shuangtasi Street, Taiyuan, 030001, China
- Academy of Microbial Ecology, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District Ct, Taiyuan, 030001, China
| | - Yaheng Li
- Shanxi Provincial Key Laboratory of Kidney Disease, The Fifth Hospital of Shanxi Medical University, No. 29 Shuangtasi Street, Taiyuan, 030001, China
| | - Yan Geng
- Clinical Laboratory, The Fifth Hospital of Shanxi Medical University, No. 29 Shuangtasi Street, Taiyuan, 030001, China
| | - Yan Qiang
- School of Software, North University of China, No. 319, Lane 3, Xueyuan Road, Taiyuan, 030051, Shanxi, China
| |
Collapse
|
3
|
Yu D, Yang X, Shang Y, Yuan S, Liu Y, Liu Y. Drug-target interaction prediction based on metapaths and simplified neighbor aggregation. Methods 2025; 240:154-164. [PMID: 40288620 DOI: 10.1016/j.ymeth.2025.04.012] [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: 01/25/2025] [Revised: 04/02/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025] Open
Abstract
Drug-target interaction (DTI) prediction is critical in drug repositioning and discovery. In current metapath-based prediction methods, attention mechanisms are often used to differentiate the importance of various neighbors, enhancing the model's expressiveness. However, in biological networks with small-scale imbalanced data, attention mechanisms are prone to interference from noise and missing data, leading to instability in weight learning, reduced efficiency, and an increased risk of overfitting. To address these issues, we propose the use of average aggregation to mitigate noise, simplify model complexity, and improve stability. Specifically, we introduce a simplified mean aggregation method for DTI prediction. This approach uses average aggregation, effectively reducing noise interference, lowering model complexity, and preventing overfitting, making it especially suitable for current biological networks. Extensive testing on three heterogeneous biological datasets shows that SNADTI outperforms 12 leading methods across two evaluation metrics, significantly reducing training time and validating its effectiveness in DTI prediction. Complexity analysis reveals that our method offers a substantial computational speed advantage over other methods on the same dataset, highlighting its enhanced efficiency. Experimental results demonstrate that SNADTI excels in prediction accuracy, stability, and reproducibility, confirming its practicality and effectiveness in DTI prediction.
Collapse
Affiliation(s)
- Di Yu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, Hunan, China
| | - Xinyu Yang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, Hunan, China
| | - Yifan Shang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, Hunan, China; Department of Biomedical Engineering, The Chinese University of Hong Kong, 999077, Hong Kong, China.
| | - Sisi Yuan
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, 28223, NC, USA
| | - Yuansheng Liu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, Hunan, China
| | - Yiping Liu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, Hunan, China
| |
Collapse
|
4
|
Li Z, Huang J, Liu X, Xu P, Shen X, Pan C, Zhang W, Liu W, Han H. KRN-DTI: Towards accurate drug-target interaction prediction with Kolmogorov-Arnold and residual networks. Methods 2025; 240:137-144. [PMID: 40287076 DOI: 10.1016/j.ymeth.2025.04.009] [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: 02/21/2025] [Revised: 04/03/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Predicting drug-target interactions (DTIs) accurately is essential in the field of drug discovery. Recently, artificial intelligence (AI) technologies, especially graph convolutional networks (GCNs), have been developed to tackle this challenge. However, as the number of GCN layers increases, models may lose critical information due to excessive smoothing. Moreover, these methods often lack interpretability and are dependent on specific datasets, which limits their generalizability. Consequently, this study introduces a novel method, KRN-DTI, which employs interpretable GCN technology to predict DTIs based on a drug-target heterogeneous network. The method uses GCN technology to identify potential DTIs by leveraging known interactions and dynamically adjusting the weights, thereby enhancing the model's interpretability. Additionally, residual connection technology is employed to integrate GNN outputs, mitigating the over-smoothing issue. Furthermore, the model's interpretability is enhanced by adaptively adjusting weights using Kolmogorov-Arnold Networks (KAN) and attention mechanisms. Experimental results show that KRN-DTI outperforms several advanced computational methods on the benchmark dataset. Case studies further highlight the effectiveness of KRN-DTI in predicting potential DTIs, showcasing its potential for real-world applications in drug discovery. Our code and data are publicly accessible at: https://github.com/lizhen5000/KRN-DTI.git.
Collapse
Affiliation(s)
- Zhen Li
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, Guangdong, 510006, China; School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, 558000, China
| | - Juyuan Huang
- Department of Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Xinxin Liu
- School of Data Science and Artificial Intelligence, Wenzhou University of Technology, Wenzhou, Zhejiang 325035, China.
| | - Peng Xu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, Guangdong, 510006, China.
| | - Xinwen Shen
- School of Data Science and Artificial Intelligence, Wenzhou University of Technology, Wenzhou, Zhejiang 325035, China
| | - Chu Pan
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan, 410000, China
| | - Wei Zhang
- Department of Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Wenbin Liu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, Guangdong, 510006, China.
| | - Henry Han
- School of engineering and computer science, Baylor University, Waco, TX, 76798, USA.
| |
Collapse
|
5
|
Zhang S, Wang X, Li F, Peng S. CPDP: Contrastive Protein-Drug Pre-Training for Novel Drug Discovery. Int J Mol Sci 2025; 26:3761. [PMID: 40332398 PMCID: PMC12028240 DOI: 10.3390/ijms26083761] [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: 03/03/2025] [Revised: 04/01/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
Novel drug discovery and repositioning remain critical challenges in biomedical research, requiring accurate prediction of drug-target interactions (DTIs). We propose the CPDP framework, which builds upon existing biomedical representation models and integrates contrastive learning with multi-dimensional representations of proteins and drugs to predict DTIs. By aligning the representation space, CPDP enables GNN-based methods to achieve zero-shot learning capabilities, allowing for accurate predictions of unseen drug data. This approach enhances DTI prediction performance, particularly for novel drugs not included in the BioHNs dataset. Experimental results demonstrate CPDP's high accuracy and strong generalization ability in predicting novel biological entities while maintaining effectiveness for traditional drug repositioning tasks.
Collapse
Affiliation(s)
- Shihan Zhang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China;
| | - Xiaoqi Wang
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China;
| | - Fei Li
- Computer Network Information Center, Chinese Academy of Sciences, Beijing 100850, China
| | - Shaoliang Peng
- The State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha 410082, China
| |
Collapse
|
6
|
Huang W, Tian X, Su Y, Zhang S, Chen C, Chen C. Sensing Compound Substructures Combined with Molecular Fingerprinting to Predict Drug-Target Interactions. Interdiscip Sci 2025:10.1007/s12539-025-00698-3. [PMID: 40178777 DOI: 10.1007/s12539-025-00698-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 04/05/2025]
Abstract
Identification of drug-target interactions (DTIs) is critical for drug discovery and drug repositioning. However, most DTI methods that extract features from drug molecules and protein entities neglect specific substructure information of pharmacological responses, which leads to poor predictive performance. Moreover, most existing methods are based on molecular graphs or molecular descriptors to obtain abstract representations of molecules, but combining the two feature learning methods for DTI prediction remains unexplored. Therefore, a new ASCS-DTI framework for DTI prediction is proposed, which utilizes a substructure attention mechanism to flexibly capture substructures of compounds at different grain sizes, allowing the important substructure information of each molecule to be learned. Additionally, the framework combines three different molecular fingerprinting information to comprehensively characterize molecular representations. A stacked convolutional coding module processes the sequence information of target proteins in a multi-scale and multi-level view. Finally, multi-modal fusion of molecular graph features and molecular fingerprint features, along with multi-modal information encoding of DTIs, is performed by the feature fusion module. The method outperforms six advanced baseline models on different benchmark datasets: Biosnap, BindingDB, and Human, with a significant improvement in performance, particularly in maintaining strong results across different experimental settings.
Collapse
Affiliation(s)
- Wanhua Huang
- School of Computer Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Xuecong Tian
- School of Computer Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Ying Su
- School of Computer Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Sizhe Zhang
- School of Software, Xinjiang University, Urumqi, 830046, Xinjiang, China
| | - Chen Chen
- School of Software, Xinjiang University, Urumqi, 830046, Xinjiang, China
| | - Cheng Chen
- School of Software, Xinjiang University, Urumqi, 830046, Xinjiang, China.
| |
Collapse
|
7
|
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
|
8
|
Zhao X, Wang Q, Zhang Y, He C, Yin M, Zhao X. CBKG-DTI: Multi-Level Knowledge Distillation and Biomedical Knowledge Graph for Drug-Target Interaction Prediction. IEEE J Biomed Health Inform 2025; 29:2284-2296. [PMID: 40030432 DOI: 10.1109/jbhi.2024.3500027] [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: 03/05/2025]
Abstract
The prediction of drug-target interactions (DTIs) has emerged as a vital step in drug discovery. Recently, biomedical knowledge graph enables the utilization of multi-omics resources for modelling complex biological systems and further improves overall performance of specific predictive task. However, due to the scale and generalization of biomedical knowledge graph, it is necessary to capture task-specific knowledge from biomedical knowledge graph for DTI prediction. Moreover, although biomedical knowledge graph has rich interactions between biological entities, there still needs to contain unignorable structural information of drugs or targets in the multi-modal fusion manner. To this end, we develop a novel DTI identification framework, CBKG-DTI, which aims to distill task-specific knowledge from the complex knowledge graph to the lightweight DTI prediction model. Specifically, CBKG-DTI first introduces a hierarchy-aware knowledge graph embedding as teacher model to capture semantic hierarchy information of biomedical knowledge graph. Then, to further improve model performance, CBKG-DTI integrates information from multiple aspects such as relational information and structural information by constructing a heterogeneous network and then employs a heterogeneous graph attention network framework as the lightweight student model. Moreover, we design a multi-level distillation mechanism to improve the representation and prediction ability of the lightweight student model via capturing the representation and logit distribution of the teacher model. Finally, we conduct the extensive comparison experiments and can reach the AUC of 0.9751 and the AUPR of 0.6310 under 5-fold cross validation. This not only demonstrates the superiority of CBKG-DTI in DTI prediction, but also, more importantly, validate the effectiveness of the framework capturing task-specific knowledge from biomedical knowledge graph.
Collapse
|
9
|
Quan N, Ma S, Zhao K, Bi X, Zhang L. MFCADTI: improving drug-target interaction prediction by integrating multiple feature through cross attention mechanism. BMC Bioinformatics 2025; 26:57. [PMID: 39966727 PMCID: PMC11834641 DOI: 10.1186/s12859-025-06075-7] [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: 12/23/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
Accurately identifying potential drug-target interactions (DTIs) is a critical step in drug discovery. Multiple heterogeneous biological data provide abundant features for DTI prediction. Many computational methods have been proposed based on these data. However, most of these methods either extract features from sequences or from networks, utilizing only one aspect of the characteristics of drugs and targets, neglecting the complementary information between these two types of features. In fact, integrating different types of features will provide more valuable information for DTI prediction. In this article, we propose a novel method to improve the predictive capability for DTIs, named MFCADTI, by integrating multi-source feature through cross-attention mechanisms. The method extracts network topological features from the heterogeneous network and attribute features from sequences of drugs and targets. Considering the complementarity and heterogeneity between network and attribute features, cross-attention mechanisms are used to integrate the network and attribute features of drugs and targets. To capture the correlations between drugs and targets, cross-attention is used to learn the interaction features of each drug-target pair. We evaluate MFCADTI on two datasets and experimental results demonstrate a significant improvement in the performance of MFCADTI compared to state-of-the-art methods. Finally, case studies illustrate that MFCADTI is an effective DTI prediction way that provides valuable guidance for drug development. The data and source code used in this study are available at: https://github.com/Dejavun/MFCADTI .
Collapse
Affiliation(s)
- Na Quan
- College of Computer Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Shicheng Ma
- College of Software, Xinjiang University, Urumqi, 830046, China
| | - Kai Zhao
- College of Computer Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Xuehua Bi
- Medical Engineering and Technology College, Xinjiang Medical University, Urumqi, 830017, China.
| | - Linlin Zhang
- College of Software, Xinjiang University, Urumqi, 830046, China.
- Center of Network and Information Technology, Xinjiang University, Urumqi, 830046, China.
| |
Collapse
|
10
|
Karimi-Sani I, Sharifi M, Abolpour N, Lotfi M, Atapour A, Takhshid MA, Sahebkar A. Drug repositioning for Parkinson's disease: An emphasis on artificial intelligence approaches. Ageing Res Rev 2025; 104:102651. [PMID: 39755176 DOI: 10.1016/j.arr.2024.102651] [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: 10/08/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
Parkinson's disease (PD) is one of the most incapacitating neurodegenerative diseases (NDDs). PD is the second most common NDD worldwide which affects approximately 1-2 percent of people over 65 years. It is an attractive pursuit for artificial intelligence (AI) to contribute to and evolve PD treatments through drug repositioning by repurposing existing drugs, shelved drugs, or even candidates that do not meet the criteria for clinical trials. A search was conducted in three databases Web of Science, Scopus, and PubMed. We reviewed the data related to the last years (1975-present) to identify those drugs currently being proposed for repositioning in PD. Moreover, we reviewed the present status of the computational approach, including AI/Machine Learning (AI/ML)-powered pharmaceutical discovery efforts and their implementation in PD treatment. It was found that the number of drug repositioning studies for PD has increased recently. Repositioning of drugs in PD is taking off, and scientific communities are increasingly interested in communicating its results and finding effective treatment alternatives for PD. A better chance of success in PD drug discovery has been made possible due to AI/ML algorithm advancements. In addition to the experimentation stage of drug discovery, it is also important to leverage AI in the planning stage of clinical trials to make them more effective. New AI-based models or solutions that increase the success rate of drug development are greatly needed.
Collapse
Affiliation(s)
- Iman Karimi-Sani
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehrdad Sharifi
- Emergency Medicine Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Nahid Abolpour
- Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehrzad Lotfi
- Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran; Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad-Ali Takhshid
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran; Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Palhamkhani F, Alipour M, Dehnad A, Abbasi K, Razzaghi P, Ghasemi JB. DeepCompoundNet: enhancing compound-protein interaction prediction with multimodal convolutional neural networks. J Biomol Struct Dyn 2025; 43:1414-1423. [PMID: 38084744 DOI: 10.1080/07391102.2023.2291829] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/23/2023] [Indexed: 01/16/2025]
Abstract
Virtual screening has emerged as a valuable computational tool for predicting compound-protein interactions, offering a cost-effective and rapid approach to identifying potential candidate drug molecules. Current machine learning-based methods rely on molecular structures and their relationship in the network. The former utilizes information such as amino acid sequences and chemical structures, while the latter leverages interaction network data, such as protein-protein interactions, drug-disease interactions, and protein-disease interactions. However, there has been limited exploration of integrating molecular information with interaction networks. This study presents DeepCompoundNet, a deep learning-based model that integrates protein features, drug properties, and diverse interaction data to predict chemical-protein interactions. DeepCompoundNet outperforms state-of-the-art methods for compound-protein interaction prediction, as demonstrated through performance evaluations. Our findings highlight the complementary nature of multiple interaction data, extending beyond amino acid sequence homology and chemical structure similarity. Moreover, our model's analysis confirms that DeepCompoundNet gets higher performance in predicting interactions between proteins and chemicals not observed in the training samples.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Farnaz Palhamkhani
- Chemistry Department, Faculty of Chemistry, School of Sciences, University of Tehran, Tehran, Iran
| | - Milad Alipour
- Department of Interdisciplinary Technologies, Network Science and Technology, College of Interdisciplinary Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Abbas Dehnad
- Faculty of Mathematics and Computer Science, Allameh Tabatabai University, Tehran, Iran
| | - Karim Abbasi
- Laboratory of System Biology, Bioinformatics & Artificial Intelligence in Medicine (LBB&AI), Faculty of Mathematics and Computer Science, Kharazmi University, Tehran, Iran
| | - Parvin Razzaghi
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Jahan B Ghasemi
- Chemistry Department, Faculty of Chemistry, School of Sciences, University of Tehran, Tehran, Iran
| |
Collapse
|
12
|
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
|
13
|
Sun L, Yin Z, Lu L. ISLRWR: A network diffusion algorithm for drug-target interactions prediction. PLoS One 2025; 20:e0302281. [PMID: 39883675 PMCID: PMC11781719 DOI: 10.1371/journal.pone.0302281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/01/2024] [Indexed: 02/01/2025] Open
Abstract
Machine learning techniques and computer-aided methods are now widely used in the pre-discovery tasks of drug discovery, effectively improving the efficiency of drug development and reducing the workload and cost. In this study, we used multi-source heterogeneous network information to build a network model, learn the network topology through multiple network diffusion algorithms, and obtain compressed low-dimensional feature vectors for predicting drug-target interactions (DTIs). We applied the metropolis-hasting random walk (MHRW) algorithm to improve the performance of the random walk with restart (RWR) algorithm, forming the basis by which the self-loop probability of the current node is removed. Additionally, the propagation efficiency of the MHRW was improved using the improved metropolis-hasting random walk (IMRWR) algorithm, facilitating network deep sampling. Finally, we proposed a correction of the transfer probability of the entire network after increasing the self-loop rate of isolated nodes to form the ISLRWR algorithm. Notably, the ISLRWR algorithm improved the area under the receiver operating characteristic curve (AUROC) by 7.53 and 5.72%, and the area under the precision-recall curve (AUPRC) by 5.95 and 4.19% compared to the RWR and MHRW algorithms, respectively, in predicting DTIs performance. Moreover, after excluding the interference of homologous proteins (popular drugs or targets may lead to inflated prediction results), the ISLRWR algorithm still showed a significant performance improvement.
Collapse
Affiliation(s)
- Lu Sun
- School of Mathematics, Physics and Statistics, Institute for Frontier Medical Technology, Center of Intelligent Computing and Applied Statistics, Shanghai University of Engineering Science, Shanghai, 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, China
| | - Lin Lu
- Shanghai Xinhao Information Technology Co., Ltd., Shanghai, China
| |
Collapse
|
14
|
Bushiri Pwesombo D, Beese C, Schmied C, Sun H. Semisupervised Contrastive Learning for Bioactivity Prediction Using Cell Painting Image Data. J Chem Inf Model 2025; 65:528-543. [PMID: 39761993 PMCID: PMC11776044 DOI: 10.1021/acs.jcim.4c00835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 01/28/2025]
Abstract
Morphological profiling has recently demonstrated remarkable potential for identifying the biological activities of small molecules. Alongside the fully supervised and self-supervised machine learning methods recently proposed for bioactivity prediction from Cell Painting image data, we introduce here a semisupervised contrastive (SemiSupCon) learning approach. This approach combines the strengths of using biological annotations in supervised contrastive learning and leveraging large unannotated image data sets with self-supervised contrastive learning. SemiSupCon enhances downstream prediction performance of classifying MeSH pharmacological classifications from PubChem, as well as mode of action and biological target annotations from the Drug Repurposing Hub across two publicly available Cell Painting data sets. Notably, our approach has effectively predicted the biological activities of several unannotated compounds, and these findings were validated through literature searches. This demonstrates that our approach can potentially expedite the exploration of biological activity based on Cell Painting image data with minimal human intervention.
Collapse
Affiliation(s)
- David Bushiri Pwesombo
- Research
Unit Structural Chemistry and Computational Biophysics, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin 13125, Germany
- Institute
of Chemistry, Technische Universität
Berlin, 10623 Berlin, Germany
| | - Carsten Beese
- Research
Unit Structural Chemistry and Computational Biophysics, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin 13125, Germany
| | - Christopher Schmied
- Research
Unit Structural Chemistry and Computational Biophysics, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin 13125, Germany
- EU-OPENSCREEN, Berlin 13125, Germany
| | - Han Sun
- Research
Unit Structural Chemistry and Computational Biophysics, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin 13125, Germany
- Institute
of Chemistry, Technische Universität
Berlin, 10623 Berlin, Germany
| |
Collapse
|
15
|
Zhang B, Quan L, Zhang Z, Cao L, Chen Q, Peng L, Wang J, Jiang Y, Nie L, Li G, Wu T, Lyu Q. MVCL-DTI: Predicting Drug-Target Interactions Using a Multiview Contrastive Learning Model on a Heterogeneous Graph. J Chem Inf Model 2025; 65:1009-1026. [PMID: 39812134 DOI: 10.1021/acs.jcim.4c02073] [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: 01/16/2025]
Abstract
Accurate prediction of drug-target interactions (DTIs) is pivotal for accelerating the processes of drug discovery and drug repurposing. MVCL-DTI, a novel model leveraging heterogeneous graphs for predicting DTIs, tackles the challenge of synthesizing information from varied biological subnetworks. It integrates neighbor view, meta-path view, and diffusion view to capture semantic features and employs an attention-based contrastive learning approach, along with a multiview attention-weighted fusion module, to effectively integrate and adaptively weight the information from the different views. Tested under various conditions on benchmark data sets, including varying positive-to-negative sample ratios, conducting hard negative sampling experiments, and masking known DTIs with different ratios, as well as redundant DTIs with various similarity metrics, MVCL-DTI exhibits strong robust generalization. The model is then employed to predict novel DTIs, with a particular focus on COVID-19-related drugs, highlighting its practical applicability. Ultimately, through features visualization and computational properties analysis, we've pinpointed critical elements, including Gene Ontology and substituent nodes, along with a proper initialization strategy, underscoring their vital role in DTI prediction tasks.
Collapse
Affiliation(s)
- Bei Zhang
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
- China Mobile (Suzhou) Software Technology Company Limited, Suzhou 215163, China
| | - Lijun Quan
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
- Collaborative Innovation Center of Novel Software Technology and Industrialization, Jiangsu 210000, China
| | - Zhijun Zhang
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Lexin Cao
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Qiufeng Chen
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Liangchen Peng
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Junkai Wang
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Yelu Jiang
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Liangpeng Nie
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Geng Li
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Tingfang Wu
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
- Collaborative Innovation Center of Novel Software Technology and Industrialization, Jiangsu 210000, China
| | - Qiang Lyu
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
- Collaborative Innovation Center of Novel Software Technology and Industrialization, Jiangsu 210000, China
| |
Collapse
|
16
|
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
|
17
|
Yang G, Liu Y, Wen S, Chen W, Zhu X, Wang Y. DTI-MHAPR: optimized drug-target interaction prediction via PCA-enhanced features and heterogeneous graph attention networks. BMC Bioinformatics 2025; 26:11. [PMID: 39800678 PMCID: PMC11726937 DOI: 10.1186/s12859-024-06021-z] [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: 08/01/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Drug-target interactions (DTIs) are pivotal in drug discovery and development, and their accurate identification can significantly expedite the process. Numerous DTI prediction methods have emerged, yet many fail to fully harness the feature information of drugs and targets or address the issue of feature redundancy. We aim to refine DTI prediction accuracy by eliminating redundant features and capitalizing on the node topological structure to enhance feature extraction. To achieve this, we introduce a PCA-augmented multi-layer heterogeneous graph-based network that concentrates on key features throughout the encoding-decoding phase. Our approach initiates with the construction of a heterogeneous graph from various similarity metrics, which is then encoded via a graph neural network. We concatenate and integrate the resultant representation vectors to merge multi-level information. Subsequently, principal component analysis is applied to distill the most informative features, with the random forest algorithm employed for the final decoding of the integrated data. Our method outperforms six baseline models in terms of accuracy, as demonstrated by extensive experimentation. Comprehensive ablation studies, visualization of results, and in-depth case analyses further validate our framework's efficacy and interpretability, providing a novel tool for drug discovery that integrates multimodal features.
Collapse
Affiliation(s)
- Guang Yang
- School of Information and Artificial Intelligence, Anhui Agricultural University, Changjiang West Road, Hefei, 230036, Anhui, China
| | - Yinbo Liu
- School of Information and Artificial Intelligence, Anhui Agricultural University, Changjiang West Road, Hefei, 230036, Anhui, China
| | - Sijian Wen
- School of Information and Artificial Intelligence, Anhui Agricultural University, Changjiang West Road, Hefei, 230036, Anhui, China
| | - Wenxi Chen
- School of Information and Artificial Intelligence, Anhui Agricultural University, Changjiang West Road, Hefei, 230036, Anhui, China
| | - Xiaolei Zhu
- School of Information and Artificial Intelligence, Anhui Agricultural University, Changjiang West Road, Hefei, 230036, Anhui, China
| | - Yongmei Wang
- School of Information and Artificial Intelligence, Anhui Agricultural University, Changjiang West Road, Hefei, 230036, Anhui, China.
| |
Collapse
|
18
|
Mishra VP, Singh YN, Khan F, Dutta MK. SeqDPI: A 1D-CNN approach for predicting binding affinity of kinase inhibitors. J Comput Chem 2025; 46:e27518. [PMID: 39644133 DOI: 10.1002/jcc.27518] [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: 03/12/2024] [Revised: 08/26/2024] [Accepted: 10/13/2024] [Indexed: 12/09/2024]
Abstract
Predicting drug target binding affinity has huge relevance in Modern drug discovery and drug repositioning processes which assist doctors to come up with new drugs or even use the existing drugs for new target proteins. In silico models, using advanced deep learning techniques could further assist these prediction tasks by providing most prominent drug target pairs. Considering these factors, a deep learning based algorithmic framework is developed in this study to support drug target interaction prediction. The proposed SeqDPI model extract the relevant drug and protein features from the one dimensional Sequential representation of the dataset considered using optimized CNN networks that deploy convolutions on varying length of amino acid subsequence's to capture hidden pattern, the convolved drug- protein features obtained are then used as an input to L2 penalized feed forward neural network which matches the local residue patterns in protein classes with molecular fingerprints of drugs to predict the binding strength for all drug target pairs. The proposed model reduces the convolution strain typically encountered in existing in silico models that utilize complex 3D structures of drug protein datasets. The result shows that the SeqDPI model achieves a mean square error MSE of (0.167) across cross validation folds, outperforming baseline models such as KronRLS (0.406), Simboost (0.226), and DeepPS (0.214). Additionally, SeqDPI attains a high CI score of 0.9114 on the benchmark KIBA dataset, demonstrating its statistical significance and computational efficiency compared to existing methods. This gives the relevance and effectiveness of SeqDPI model in accurately predicting binding affinities while working with simpler one-dimensional data, making it a robust and computationally cost-effective solution for drug-target interaction prediction.
Collapse
Affiliation(s)
- Vinay Priy Mishra
- Centre for Advanced Studies, Dr. A.P.J. Abdul Kalam Technical University, Lucknow, India
| | - Yogendra Narain Singh
- Department of Computer Science & Engineering, Institute of Engineering and Technology, Lucknow, India
| | - Feroz Khan
- Technology Dissemination & Computational Biology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | | |
Collapse
|
19
|
Xie Y, Wang X, Wang P, Bi X. A pseudo-label supervised graph fusion attention network for drug–target interaction prediction. EXPERT SYSTEMS WITH APPLICATIONS 2025; 259:125264. [DOI: 10.1016/j.eswa.2024.125264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
20
|
Bhatia T, Sharma S. Drug Repurposing: Insights into Current Advances and Future Applications. Curr Med Chem 2025; 32:468-510. [PMID: 37946344 DOI: 10.2174/0109298673266470231023110841] [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: 08/06/2023] [Revised: 09/04/2023] [Accepted: 09/11/2023] [Indexed: 11/12/2023]
Abstract
Drug development is a complex and expensive process that involves extensive research and testing before a new drug can be approved for use. This has led to a limited availability of potential therapeutics for many diseases. Despite significant advances in biomedical science, the process of drug development remains a bottleneck, as all hypotheses must be tested through experiments and observations, which can be timeconsuming and costly. To address this challenge, drug repurposing has emerged as an innovative strategy for finding new uses for existing medications that go beyond their original intended use. This approach has the potential to speed up the drug development process and reduce costs, making it an attractive option for pharmaceutical companies and researchers alike. It involves the identification of existing drugs or compounds that have the potential to be used for the treatment of a different disease or condition. This can be done through a variety of approaches, including screening existing drugs against new disease targets, investigating the biological mechanisms of existing drugs, and analyzing data from clinical trials and electronic health records. Additionally, repurposing drugs can lead to the identification of new therapeutic targets and mechanisms of action, which can enhance our understanding of disease biology and lead to the development of more effective treatments. Overall, drug repurposing is an exciting and promising area of research that has the potential to revolutionize the drug development process and improve the lives of millions of people around the world. The present review provides insights on types of interaction, approaches, availability of databases, applications and limitations of drug repurposing.
Collapse
Affiliation(s)
- Trisha Bhatia
- School of Pharmacy, National Forensic Sciences University, Gandhinagar, Gujarat, 382007, India
| | - Shweta Sharma
- School of Pharmacy, National Forensic Sciences University, Gandhinagar, Gujarat, 382007, India
| |
Collapse
|
21
|
Ning Q, Wang Y, Zhao Y, Sun J, Jiang L, Wang K, Yin M. DMHGNN: Double multi-view heterogeneous graph neural network framework for drug-target interaction prediction. Artif Intell Med 2025; 159:103023. [PMID: 39579417 DOI: 10.1016/j.artmed.2024.103023] [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: 12/03/2023] [Revised: 10/09/2024] [Accepted: 11/15/2024] [Indexed: 11/25/2024]
Abstract
Accurate identification of drug-target interactions (DTIs) plays a crucial role in drug discovery. Compared with traditional experimental methods that are labor-intensive and time-consuming, computational methods for drug-target interactions prediction are more popular in recent years. Conventional computational methods almost simply view heterogeneous network constructed by the drug-related and protein-related dataset instead of comprehensively exploring drug-protein pair (DPP) information. To address this limitation, we proposed a Double Multi-view Heterogeneous Graph Neural Network framework for drug-target interaction prediction (DMHGNN). In DMHGNN, one multi-view heterogeneous graph neural network is based on meta-paths and denoising autoencoder for protein-, drug-related heterogeneous network learning, and another multi-view heterogeneous graph neural network is based on multi-channel graph convolutional network for drug-protein pair similarity network learning. First, a meta-path-based graph encoder with the attention mechanism is used for substructure learning of complex relationships from heterogeneous network constructed by proteins, drugs, side-effects and diseases, obtaining key information that is easy to be ignored in global learning of heterogeneous networks, and multi-source neighbouring features for drugs and proteins are learned from heterogeneous network via denoising auto-encoder model. Then, multi-view graphs of drug-protein pairs (DPPs) including the topology graph, semantics graph and collaborative graph with shared weights are constructed, and the multi-channel graph convolutional network (GCN) is utilized to learn the deep representation of DPPs. Finally, a multi-layer fully connection network is trained to predict drug-target interactions. Experiments have demonstrated its effectiveness and better performance than state-of-the-art methods.
Collapse
Affiliation(s)
- Qiao Ning
- The School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214122, Jiangsu, China; Information Science and Technology, Dalian Maritime University, Dalian 116026, Liaoning, China; Neusoft Education Technology Group, Dalian 116026, Liaoning, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun 130015, Jilin, China.
| | - Yue Wang
- Information Science and Technology, Dalian Maritime University, Dalian 116026, Liaoning, China
| | - Yaomiao Zhao
- Information Science and Technology, Dalian Maritime University, Dalian 116026, Liaoning, China
| | - Jiahao Sun
- Computer Science and Technology, the Northeast Normal University, Changchun 999078, Jilin, China
| | - Lu Jiang
- Information Science and Technology, Dalian Maritime University, Dalian 116026, Liaoning, China; Computer Science and Technology, the Northeast Normal University, Changchun 999078, Jilin, China.
| | - Kaidi Wang
- Computer Science and Technology, the Northeast Normal University, Changchun 999078, Jilin, China
| | - Minghao Yin
- Computer Science and Technology, the Northeast Normal University, Changchun 999078, Jilin, China.
| |
Collapse
|
22
|
Wang M, Wang J, Ji J, Ma C, Wang H, He J, Song Y, Zhang X, Cao Y, Dai Y, Hua M, Qin R, Li K, Cao L. Improving compound-protein interaction prediction by focusing on intra-modality and inter-modality dynamics with a multimodal tensor fusion strategy. Comput Struct Biotechnol J 2024; 23:3714-3729. [PMID: 39525082 PMCID: PMC11544084 DOI: 10.1016/j.csbj.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 11/16/2024] Open
Abstract
Identifying novel compound-protein interactions (CPIs) plays a pivotal role in target identification and drug discovery. Although the recent multimodal methods have achieved outstanding advances in CPI prediction, they fail to effectively learn both intra-modality and inter-modality dynamics, which limits their prediction performance. To address the limitation, we propose a novel multimodal tensor fusion CPI prediction framework, named MMTF-CPI, which contains three unimodal learning modules for structure, heterogeneous network and transcriptional profiling modalities, a tensor fusion module and a prediction module. MMTF-CPI is capable of focusing on both intra-modality and inter-modality dynamics with the tensor fusion module. We demonstrated that MMTF-CPI is superior to multiple state-of-the-art multimodal methods across seven datasets. The prediction performance of MMTF-CPI is significantly improved with the tensor fusion module compared to other fusion methods. Moreover, our case studies confirmed the practical value of MMTF-CPI in target identification. Via MMTF-CPI, we also discovered several candidate compounds for the therapy of breast cancer and non-small cell lung cancer.
Collapse
Affiliation(s)
- Meng Wang
- Department of Biostatistics, Harbin Medical University, Harbin 150081, China
| | - Jianmin Wang
- Department of Integrative Biotechnology, Yonsei University, Incheon 21983, South Korea
| | - Jianxin Ji
- Department of Biostatistics, Harbin Medical University, Harbin 150081, China
| | - Chenjing Ma
- Department of Biostatistics, Harbin Medical University, Harbin 150081, China
| | - Hesong Wang
- Department of Biostatistics, Harbin Medical University, Harbin 150081, China
| | - Jia He
- Department of Biostatistics, Harbin Medical University, Harbin 150081, China
| | - Yongzhen Song
- Department of Biostatistics, Harbin Medical University, Harbin 150081, China
| | - Xuan Zhang
- Department of Biostatistics, Harbin Medical University, Harbin 150081, China
| | - Yong Cao
- Department of Biostatistics, Harbin Medical University, Harbin 150081, China
| | - Yanyan Dai
- Department of Biostatistics, Harbin Medical University, Harbin 150081, China
| | - Menglei Hua
- Department of Biostatistics, Harbin Medical University, Harbin 150081, China
| | - Ruihao Qin
- Department of Biostatistics, Harbin Medical University, Harbin 150081, China
| | - Kang Li
- Department of Biostatistics, Harbin Medical University, Harbin 150081, China
| | - Lei Cao
- Department of Biostatistics, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
23
|
Li M, Liu H, Kong F, Lv P. DTRE: A model for predicting drug-target interactions of endometrial cancer based on heterogeneous graph. FUTURE GENERATION COMPUTER SYSTEMS 2024; 161:478-486. [DOI: 10.1016/j.future.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
|
24
|
Mishra S, Chinthala A, Bhattacharya M. Drug-target prediction through self supervised learning with dual task ensemble approach. Comput Biol Chem 2024; 113:108244. [PMID: 39454455 DOI: 10.1016/j.compbiolchem.2024.108244] [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: 06/16/2024] [Revised: 09/15/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024]
Abstract
Drug-Target interaction (DTI) prediction, a transformative approach in pharmaceutical research, seeks novel therapeutic applications for computational method based virtual screening, existing drugs to address untreated diseases and discovery of existing drugs side effects. The proposed model predict DTI through Heterogeneous biological network by combining drug, genes and disease related knowledge. For the purpose of embedding extraction Self-supervised learning (SSL) has been used which, trains models through pretext tasks, eliminating the need for manual annotations. The pretext tasks are related to either structural based information or similarity based information. To mitigate GNN vulnerability to non-robustness, ensemble learning can be incorporated into GNNs, harnessing multiple models to enhance robustness. This paper introduces a Graph neural network based architecture consisting of task based module and ensemble module for link prediction of DTI. The ensemble module of dual task combinations, both in cold start and warm start scenarios achieve very good performance as it provide 0.960 in cold start and 0.970 in warm start mean AUCROC score with less deviation.
Collapse
Affiliation(s)
- Surabhi Mishra
- ABV- Indian Institute of Information Technology and Management., Morena Road, Gwalior, 474015, India.
| | - Ashish Chinthala
- ABV- Indian Institute of Information Technology and Management., Morena Road, Gwalior, 474015, India.
| | - Mahua Bhattacharya
- ABV- Indian Institute of Information Technology and Management., Morena Road, Gwalior, 474015, India.
| |
Collapse
|
25
|
Zong N, Chowdhury S, Zhou S, Rajaganapathy S, Yu Y, Wang L, Dai Q, Li P, Liu X, Bielinski SJ, Chen J, Chen Y, Cerhan JR. Advancing Efficacy Prediction for EHR-based Emulated Trials in Repurposing Heart Failure Therapies. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.05.25.23290531. [PMID: 37398384 PMCID: PMC10312819 DOI: 10.1101/2023.05.25.23290531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Introduction The High mortality rates associated with heart failure (HF) have propelled the strategy of drug repurposing, which seeks new therapeutic uses for existing, approved drugs to enhance the management of HF symptoms effectively. An emerging trend focuses on utilizing real-world data, like EHR, to mimic randomized controlled trials (RCTs) for evaluating treatment outcomes through what are known as emulated trials (ET). Nonetheless, the intricacies inherent in EHR data-comprising detailed patient histories in databases, the omission of certain biomarkers or specific diagnostic tests, and partial records of symptoms-introduce notable discrepancies between EHR data and the stringent standards of RCTs. This gap poses a substantial challenge in conducting an ET to accurately predict treatment efficacy. Objective The objective of this research is to predict the efficacy of drugs repurposed for HF in randomized trials by leveraging EHR in ET. Methods We proposed an ET framework to predict drug efficacy, integrating target prediction based on biomedical databases with statistical analysis using EHR data. Specifically, we developed a novel target prediction model that learns low-dimensional representations of drug molecules, protein sequences, and diverse biomedical associations from a knowledge graph. Additionally, we crafted strategies to improve the prediction by considering the interactions between HF drugs and biological factors in the context of HF prognostic markers. Results Our validation of the drug-target prediction model against the BETA benchmark demonstrated superior performance, with an average AUCROC of 97.7%, PRAUC of 97.4%, F1 score of 93.1%, and a General Score of 96.1%, surpassing existing baseline algorithms. Further analysis of our ET framework on identifying 17 repurposed drugs-derived from 266 phase 3 HF RCTs-using data from 59,000 patients at the Mayo Clinic highlighted the framework's remarkable predictive accuracy. This analysis took into account various factors such as biological variables (e.g., gender, age, ethnicity), HF medications (e.g., ACE inhibitors, Beta-blockers, ARBs, Loop Diuretics), types of HF (HFpEF and HFrEF), confounders, and prognostic markers (e.g., NT-proBNP, bUn, creatinine, and hemoglobin). The ET framework significantly improved the accuracy compared to the baseline efficacy analysis that utilized EHR data. Notably, the best results were improved in AUC-ROC from 75.71% to 93.57% and in PRAUC from 78.66% to 90.34%, compared to the baseline models. Conclusion Our study presents an ET framework that significantly enhances drug efficacy emulation by integrating EHR-based analysis with target prediction. We demonstrated substantial success in predicting the efficacy of 17 HF drugs repurposed for phase 3 RCTs, showcasing the framework's potential in advancing HF treatment strategies.
Collapse
Affiliation(s)
- Nansu Zong
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Shaika Chowdhury
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Shibo Zhou
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Sivaraman Rajaganapathy
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Yue Yu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Qiying Dai
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Pengyang Li
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, VA, USA
| | - Xiaoke Liu
- Division of Community Cardiology, Department of Cardiovascular Medicine, La Crosse, Wisconsin, WI, USA
| | | | - Jun Chen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Yongbin Chen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - James R. Cerhan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
26
|
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
|
27
|
Zhang M, Hong Y, Shen L, Xu S, Xu Y, Zhang X, Liu J, Liu X. A heterogeneous graph neural network with automatic discovery of effective metapaths for drug–target interaction prediction. FUTURE GENERATION COMPUTER SYSTEMS 2024; 160:283-294. [DOI: 10.1016/j.future.2024.05.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
28
|
Xu B, Chen J, Wang Y, Fu Q, Lu Y. Reinforced Metapath Optimization in Heterogeneous Information Networks for Drug-Target Interaction Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:2315-2329. [PMID: 39316496 DOI: 10.1109/tcbb.2024.3467135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Graph neural networks offer an effective avenue for predicting drug-target interactions. In this domain, researchers have found that constructing heterogeneous information networks based on metapaths using diverse biological datasets enhances prediction performance. However, the performance of such methods is closely tied to the selection of metapaths and the compatibility between metapath subgraphs and graph neural networks. Most existing approaches still rely on fixed strategies for selecting metapaths and often fail to fully exploit node information along the metapaths, limiting the improvement in model performance. This paper introduces a novel method for predicting drug-target interactions by optimizing metapaths in heterogeneous information networks. On one hand, the method formulates the metapath optimization problem as a Markov decision process, using the enhancement of downstream network performance as a reward signal. Through iterative training of a reinforcement learning agent, a high-quality set of metapaths is learned. On the other hand, to fully leverage node information along the metapaths, the paper constructs subgraphs based on nodes along the metapaths. Different depths of subgraphs are processed using different graph convolutional neural network. The proposed method is validated using standard heterogeneous biological benchmark datasets. Experimental results on standard datasets show significant advantages over traditional methods.
Collapse
|
29
|
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
|
30
|
Li Y, Zhang X, Chen Z, Yang H, Liu Y, Wang H, Yan T, Xiang J, Wang B. Accurate prediction of drug-target interactions in Chinese and western medicine by the CWI-DTI model. Sci Rep 2024; 14:25054. [PMID: 39443630 PMCID: PMC11499656 DOI: 10.1038/s41598-024-76367-0] [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: 07/10/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Accurate prediction of drug-target interactions (DTIs) is crucial for advancing drug discovery and repurposing. Computational methods have significantly improved the efficiency of experimental predictions for drug-target interactions in Western medicine. However, accurately predicting the complex relationships between Chinese medicine ingredients and targets remains a formidable challenge due to the vast number and high heterogeneity of these ingredients. In this study, we introduce the CWI-DTI method, which achieves high-accuracy prediction of DTIs using a large dataset of interactive relationships of drug ingredients or candidate targets. Moreover, we present a novel dataset to evaluate the prediction accuracy of both Chinese and Western medicine. Through meticulous collection and preprocessing of data on ingredients and targets, we employ an innovative autoencoder framework to fuse multiple drug (target) topological similarity matrices. Additionally, we employ denoising blocks, sparse blocks, and stacked blocks to extract crucial features from the similarity matrix, reducing noise and enhancing accuracy across diverse datasets. Our results indicate that the CWI-DTI model shows improved performance compared to several existing state-of-the-art methods on the datasets tested in both Western and Chinese medicine databases. The findings of this study hold immense promise for advancing DTI prediction in Chinese and Western medicine, thus fostering more efficient drug discovery and repurposing endeavors. Our model is available at https://github.com/WANG-BIN-LAB/CWIDTI .
Collapse
Affiliation(s)
- Ying Li
- Department of Computer Science and Technology, Taiyuan University of Technology, Taiyuan, China
| | - Xingyu Zhang
- Department of Computer Science and Technology, Taiyuan University of Technology, Taiyuan, China
| | - Zhuo Chen
- Department of Computer Science and Technology, Taiyuan University of Technology, Taiyuan, China
| | - Hongye Yang
- Department of Computer Science and Technology, Taiyuan University of Technology, Taiyuan, China
| | - Yuhui Liu
- Department of Computer Science and Technology, Taiyuan University of Technology, Taiyuan, China
| | - Huiqing Wang
- Department of Computer Science and Technology, Taiyuan University of Technology, Taiyuan, China
| | - Ting Yan
- Department of Pathology, Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, China
| | - Jie Xiang
- Department of Computer Science and Technology, Taiyuan University of Technology, Taiyuan, China
| | - Bin Wang
- Department of Computer Science and Technology, Taiyuan University of Technology, Taiyuan, China.
| |
Collapse
|
31
|
Wang C, Yang Y, Song J, Nan X. Research Progresses and Applications of Knowledge Graph Embedding Technique in Chemistry. J Chem Inf Model 2024; 64:7189-7213. [PMID: 39302256 DOI: 10.1021/acs.jcim.4c00791] [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: 09/22/2024]
Abstract
A knowledge graph (KG) is a technique for modeling entities and their interrelations. Knowledge graph embedding (KGE) translates these entities and relationships into a continuous vector space to facilitate dense and efficient representations. In the domain of chemistry, applying KG and KGE techniques integrates heterogeneous chemical information into a coherent and user-friendly framework, enhances the representation of chemical data features, and is beneficial for downstream tasks, such as chemical property prediction. This paper begins with a comprehensive review of classical and contemporary KGE methodologies, including distance-based models, semantic matching models, and neural network-based approaches. We then catalogue the primary databases employed in chemistry and biochemistry that furnish the KGs with essential chemical data. Subsequently, we explore the latest applications of KG and KGE in chemistry, focusing on risk assessment, property prediction, and drug discovery. Finally, we discuss the current challenges to KG and KGE techniques and provide a perspective on their potential future developments.
Collapse
Affiliation(s)
- Chuanghui Wang
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450001, China
| | - Yunqing Yang
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450001, China
| | - Jinshuai Song
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaofei Nan
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
32
|
Zhang P, Lin P, Li D, Wang W, Qi X, Li J, Xiong J. MGACL: Prediction Drug-Protein Interaction Based on Meta-Graph Association-Aware Contrastive Learning. Biomolecules 2024; 14:1267. [PMID: 39456200 PMCID: PMC11505808 DOI: 10.3390/biom14101267] [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: 08/03/2024] [Revised: 09/20/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
The identification of drug-target interaction (DTI) is crucial for drug discovery. However, how to reduce the graph neural network's false positives due to its bias and negative transfer in the original bipartite graph remains to be clarified. Considering that the impact of heterogeneous auxiliary information on DTI varies depending on the drug and target, we established an adaptive enhanced personalized meta-knowledge transfer network named Meta Graph Association-Aware Contrastive Learning (MGACL), which can transfer personalized heterogeneous auxiliary information from different nodes and reduce data bias. Meanwhile, we propose a novel DTI association-aware contrastive learning strategy that aligns high-frequency drug representations with learned auxiliary graph representations to prevent negative transfer. Our study improves the DTI prediction performance by about 3%, evaluated by analyzing the area under the curve (AUC) and area under the precision-recall curve (AUPRC) compared with existing methods, which is more conducive to accurately identifying drug targets for the development of new drugs.
Collapse
Affiliation(s)
- Pinglu Zhang
- Faculty of Information Science and Engineering, Ocean University of China, Qingdao 266003, China; (P.Z.); (W.W.)
| | - Peng Lin
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (P.L.); (D.L.); (X.Q.)
| | - Dehai Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (P.L.); (D.L.); (X.Q.)
| | - Wanchun Wang
- Faculty of Information Science and Engineering, Ocean University of China, Qingdao 266003, China; (P.Z.); (W.W.)
| | - Xin Qi
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (P.L.); (D.L.); (X.Q.)
| | - Jing Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (P.L.); (D.L.); (X.Q.)
| | - Jianshe Xiong
- Faculty of Information Science and Engineering, Ocean University of China, Qingdao 266003, China; (P.Z.); (W.W.)
| |
Collapse
|
33
|
Majidifar S, Zabihian A, Hooshmand M. Combination therapy synergism prediction for virus treatment using machine learning models. PLoS One 2024; 19:e0309733. [PMID: 39231124 PMCID: PMC11373828 DOI: 10.1371/journal.pone.0309733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/16/2024] [Indexed: 09/06/2024] Open
Abstract
Combining different drugs synergistically is an essential aspect of developing effective treatments. Although there is a plethora of research on computational prediction for new combination therapies, there is limited to no research on combination therapies in the treatment of viral diseases. This paper proposes AI-based models for predicting novel antiviral combinations to treat virus diseases synergistically. To do this, we assembled a comprehensive dataset comprising information on viral strains, drug compounds, and their known interactions. As far as we know, this is the first dataset and learning model on combination therapy for viruses. Our proposal includes using a random forest model, an SVM model, and a deep model to train viral combination therapy. The machine learning models showed the highest performance, and the predicted values were validated by a t-test, indicating the effectiveness of the proposed methods. One of the predicted combinations of acyclovir and ribavirin has been experimentally confirmed to have a synergistic antiviral effect against herpes simplex type-1 virus, as described in the literature.
Collapse
Affiliation(s)
- Shayan Majidifar
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Arash Zabihian
- Department of QA, Kimia Zist Parsian Pharmaceutical Company, Zanjan, Iran
| | - Mohsen Hooshmand
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| |
Collapse
|
34
|
Ahmed KT, Ansari MI, Zhang W. DTI-LM: language model powered drug-target interaction prediction. Bioinformatics 2024; 40:btae533. [PMID: 39221997 PMCID: PMC11520403 DOI: 10.1093/bioinformatics/btae533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 08/05/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024] Open
Abstract
MOTIVATION The identification and understanding of drug-target interactions (DTIs) play a pivotal role in the drug discovery and development process. Sequence representations of drugs and proteins in computational model offer advantages such as their widespread availability, easier input quality control, and reduced computational resource requirements. These make them an efficient and accessible tools for various computational biology and drug discovery applications. Many sequence-based DTI prediction methods have been developed over the years. Despite the advancement in methodology, cold start DTI prediction involving unknown drug or protein remains a challenging task, particularly for sequence-based models. Introducing DTI-LM, a novel framework leveraging advanced pretrained language models, we harness their exceptional context-capturing abilities along with neighborhood information to predict DTIs. DTI-LM is specifically designed to rely solely on sequence representations for drugs and proteins, aiming to bridge the gap between warm start and cold start predictions. RESULTS Large-scale experiments on four datasets show that DTI-LM can achieve state-of-the-art performance on DTI predictions. Notably, it excels in overcoming the common challenges faced by sequence-based models in cold start predictions for proteins, yielding impressive results. The incorporation of neighborhood information through a graph attention network further enhances prediction accuracy. Nevertheless, a disparity persists between cold start predictions for proteins and drugs. A detailed examination of DTI-LM reveals that language models exhibit contrasting capabilities in capturing similarities between drugs and proteins. AVAILABILITY AND IMPLEMENTATION Source code is available at: https://github.com/compbiolabucf/DTI-LM.
Collapse
Affiliation(s)
- Khandakar Tanvir Ahmed
- Department of Computer Science, University of Central Florida, Orlando, FL 32816, United States
- Genomics and Bioinformatics Cluster, University of Central Florida, Orlando, FL 32816, United States
| | - Md Istiaq Ansari
- Department of Computer Science, University of Central Florida, Orlando, FL 32816, United States
- Genomics and Bioinformatics Cluster, University of Central Florida, Orlando, FL 32816, United States
| | - Wei Zhang
- Department of Computer Science, University of Central Florida, Orlando, FL 32816, United States
- Genomics and Bioinformatics Cluster, University of Central Florida, Orlando, FL 32816, United States
| |
Collapse
|
35
|
Wang S, Liu Y, Zhang Y, Zhang K, Song X, Zhang Y, Pang S. CHL-DTI: A Novel High-Low Order Information Convergence Framework for Effective Drug-Target Interaction Prediction. Interdiscip Sci 2024; 16:568-578. [PMID: 38483753 DOI: 10.1007/s12539-024-00608-z] [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: 07/22/2023] [Revised: 01/05/2024] [Accepted: 01/07/2024] [Indexed: 09/19/2024]
Abstract
Recognizing drug-target interactions (DTI) stands as a pivotal element in the expansive field of drug discovery. Traditional biological wet experiments, although valuable, are time-consuming and costly as methods. Recently, computational methods grounded in network learning have demonstrated great advantages by effective topological feature extraction and attracted extensive research attention. However, most existing network-based learning methods only consider the low-order binary correlation between individual drug and target, neglecting the potential higher-order correlation information derived from multiple drugs and targets. High-order information, as an essential component, exhibits complementarity with low-order information. Hence, the incorporation of higher-order associations between drugs and targets, while adequately integrating them with the existing lower-order information, could potentially yield substantial breakthroughs in predicting drug-target interactions. We propose a novel dual channels network-based learning model CHL-DTI that converges high-order information from hypergraphs and low-order information from ordinary graph for drug-target interaction prediction. The convergence of high-low order information in CHL-DTI is manifested in two key aspects. First, during the feature extraction stage, the model integrates both high-level semantic information and low-level topological information by combining hypergraphs and ordinary graph. Second, CHL-DTI fully fuse the innovative introduced drug-protein pairs (DPP) hypergraph network structure with ordinary topological network structure information. Extensive experimentation conducted on three public datasets showcases the superior performance of CHL-DTI in DTI prediction tasks when compared to SOTA methods. The source code of CHL-DTI is available at https://github.com/UPCLyy/CHL-DTI .
Collapse
Affiliation(s)
- Shudong Wang
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Yingye Liu
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Yuanyuan Zhang
- College of Information and Control Engineering, Qingdao University of Technology, Qingdao, 266520, China.
| | - Kuijie Zhang
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Xuanmo Song
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Yu Zhang
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Shanchen Pang
- College of Computer Science and Technology, China University of Petroleum (East China), Qingdao, 266580, China
| |
Collapse
|
36
|
Pang H, Wei S, Du Z, Zhao Y, Cai S, Zhao Y. Graph Representation Learning Based on Specific Subgraphs for Biomedical Interaction Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:1552-1564. [PMID: 38767994 DOI: 10.1109/tcbb.2024.3402741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Discovering the novel associations of biomedical entities is of great significance and can facilitate not only the identification of network biomarkers of disease but also the search for putative drug targets.Graph representation learning (GRL) has incredible potential to efficiently predict the interactions from biomedical networks by modeling the robust representation for each node.> However, the current GRL-based methods learn the representation of nodes by aggregating the features of their neighbors with equal weights. Furthermore, they also fail to identify which features of higher-order neighbors are integrated into the representation of the central node. In this work, we propose a novel graph representation learning framework: a multi-order graph neural network based on reconstructed specific subgraphs (MGRS) for biomedical interaction prediction. In the MGRS, we apply the multi-order graph aggregation module (MOGA) to learn the wide-view representation by integrating the multi-hop neighbor features. Besides, we propose a subgraph selection module (SGSM) to reconstruct the specific subgraph with adaptive edge weights for each node. SGSM can clearly explore the dependency of the node representation on the neighbor features and learn the subgraph-based representation based on the reconstructed weighted subgraphs. Extensive experimental results on four public biomedical networks demonstrate that the MGRS performs better and is more robust than the latest baselines.
Collapse
|
37
|
Wang H, Cui Z, Yang Y, Wang B, Zhu L, Zhang W. A Network Enhancement Method to Identify Spurious Drug-Drug Interactions. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:1335-1347. [PMID: 38635380 DOI: 10.1109/tcbb.2024.3385796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
As medical safety and drug regulation gain heightened attention, the detection of spurious drug-drug interactions (DDI) has become key in healthcare. Although current research using graph neural networks (GNNs) to predict DDI has shown impressive results, it often fails to account for false DDI in the constructed DDI networks. Such inaccuracies caused by data errors, false alarms, or incorrect drug details can skew the network's structure and hinder the accuracy of GNN-based predictions. To tackle this challenge, we propose ANSM, a network-enhancement method specifically designed to identify and attenuate spurious links between drugs for ensuring the accuracy of DDI networks. ANSM integrates three key components: the feature extractor, the network optimizer, and the discriminative classifier. The feature extractor captures local structural features from drug node pairs, while the network optimizer leverages network information to improve feature extraction and reduce the impact of spurious DDI links. The discriminative classifier then identifies potential spurious links. Experimental results demonstrate that ANSM outperforms state-of-the-art methods in identifying spurious DDI.
Collapse
|
38
|
Ohnuki Y, Akiyama M, Sakakibara Y. Deep learning of multimodal networks with topological regularization for drug repositioning. J Cheminform 2024; 16:103. [PMID: 39180095 PMCID: PMC11342530 DOI: 10.1186/s13321-024-00897-y] [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: 01/14/2024] [Accepted: 08/12/2024] [Indexed: 08/26/2024] Open
Abstract
MOTIVATION Computational techniques for drug-disease prediction are essential in enhancing drug discovery and repositioning. While many methods utilize multimodal networks from various biological databases, few integrate comprehensive multi-omics data, including transcriptomes, proteomes, and metabolomes. We introduce STRGNN, a novel graph deep learning approach that predicts drug-disease relationships using extensive multimodal networks comprising proteins, RNAs, metabolites, and compounds. We have constructed a detailed dataset incorporating multi-omics data and developed a learning algorithm with topological regularization. This algorithm selectively leverages informative modalities while filtering out redundancies. RESULTS STRGNN demonstrates superior accuracy compared to existing methods and has identified several novel drug effects, corroborating existing literature. STRGNN emerges as a powerful tool for drug prediction and discovery. The source code for STRGNN, along with the dataset for performance evaluation, is available at https://github.com/yuto-ohnuki/STRGNN.git .
Collapse
Affiliation(s)
- Yuto Ohnuki
- Department of Biosciences and Informatics, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Manato Akiyama
- Department of Biosciences and Informatics, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Yasubumi Sakakibara
- Department of Biosciences and Informatics, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan.
| |
Collapse
|
39
|
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
|
40
|
Zabihian A, Asghari J, Hooshmand M, Gharaghani S. A comparative analysis of computational drug repurposing approaches: proposing a novel tensor-matrix-tensor factorization method. Mol Divers 2024; 28:2177-2196. [PMID: 38683487 DOI: 10.1007/s11030-024-10851-7] [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: 12/28/2023] [Accepted: 03/18/2024] [Indexed: 05/01/2024]
Abstract
Efficient drug discovery relies on drug repurposing, an important and open research field. This work presents a novel factorization method and a practical comparison of different approaches for drug repurposing. First, we propose a novel tensor-matrix-tensor (TMT) formulation as a new data array method with a gradient-based factorization procedure. Additionally, this paper examines and contrasts four computational drug repurposing approaches-factorization-based methods, machine learning methods, deep learning methods, and graph neural networks-to fulfill the second purpose. We test the strategies on two datasets and assess each approach's performance, drawbacks, problems, and benefits based on results. The results demonstrate that deep learning techniques work better than other strategies and that their results might be more reliable. Ultimately, graph neural methods need to be in an inductive manner to have a reliable prediction.
Collapse
Affiliation(s)
- Arash Zabihian
- Department of Bioinformatics, Kish International Campus, University of Tehran, Kish, Iran
| | - Javad Asghari
- Department of Computer Science and Information Technology, Institute of Advanced Studies in Basic Sciences, Zanjan, Iran
| | - Mohsen Hooshmand
- Department of Computer Science and Information Technology, Institute of Advanced Studies in Basic Sciences, Zanjan, Iran.
| | - Sajjad Gharaghani
- Laboratory of Bioinformatics and Drug Design, University of Tehran, Tehran, Iran
| |
Collapse
|
41
|
Zhou X, Yang J, Luo Y, Shen X. HNCGAT: a method for predicting plant metabolite-protein interaction using heterogeneous neighbor contrastive graph attention network. Brief Bioinform 2024; 25:bbae397. [PMID: 39162311 PMCID: PMC11730448 DOI: 10.1093/bib/bbae397] [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: 04/07/2024] [Revised: 07/15/2024] [Accepted: 07/27/2024] [Indexed: 08/21/2024] Open
Abstract
The prediction of metabolite-protein interactions (MPIs) plays an important role in plant basic life functions. Compared with the traditional experimental methods and the high-throughput genomics methods using statistical correlation, applying heterogeneous graph neural networks to the prediction of MPIs in plants can reduce the cost of manpower, resources, and time. However, to the best of our knowledge, applying heterogeneous graph neural networks to the prediction of MPIs in plants still remains under-explored. In this work, we propose a novel model named heterogeneous neighbor contrastive graph attention network (HNCGAT), for the prediction of MPIs in Arabidopsis. The HNCGAT employs the type-specific attention-based neighborhood aggregation mechanism to learn node embeddings of proteins, metabolites, and functional-annotations, and designs a novel heterogeneous neighbor contrastive learning framework to preserve heterogeneous network topological structures. Extensive experimental results and ablation study demonstrate the effectiveness of the HNCGAT model for MPI prediction. In addition, a case study on our MPI prediction results supports that the HNCGAT model can effectively predict the potential MPIs in plant.
Collapse
Affiliation(s)
- Xi Zhou
- School of Tropical Agriculture and Forestry, Hainan University, 58 Renmin Avenue, Haikou 570228, Hainan, China
| | - Jing Yang
- School of Tropical Agriculture and Forestry, Hainan University, 58 Renmin Avenue, Haikou 570228, Hainan, China
| | - Yin Luo
- School of Tropical Agriculture and Forestry, Hainan University, 58 Renmin Avenue, Haikou 570228, Hainan, China
| | - Xiao Shen
- School of Computer Science and Technology, Hainan University, 58 Renmin Avenue, Haikou 570228, Hainan, China
| |
Collapse
|
42
|
Yuan Y, Hu R, Chen S, Zhang X, Liu Z, Zhou G. CKG-IMC: An inductive matrix completion method enhanced by CKG and GNN for Alzheimer's disease compound-protein interactions prediction. Comput Biol Med 2024; 177:108612. [PMID: 38838556 DOI: 10.1016/j.compbiomed.2024.108612] [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: 12/05/2023] [Revised: 04/17/2024] [Accepted: 05/11/2024] [Indexed: 06/07/2024]
Abstract
Alzheimer's disease (AD) is one of the most prevalent chronic neurodegenerative disorders globally, with a rapidly growing population of AD patients and currently no effective therapeutic interventions available. Consequently, the development of therapeutic anti-AD drugs and the identification of AD targets represent one of the most urgent tasks. In this study, in addition to considering known drugs and targets, we explore compound-protein interactions (CPIs) between compounds and proteins relevant to AD. We propose a deep learning model called CKG-IMC to predict Alzheimer's disease compound-protein interaction relationships. CKG-IMC comprises three modules: a collaborative knowledge graph (CKG), a principal neighborhood aggregation graph neural network (PNA), and an inductive matrix completion (IMC). The collaborative knowledge graph is used to learn semantic associations between entities, PNA is employed to extract structural features of the relationship network, and IMC is utilized for CPIs prediction. Compared with a total of 16 baseline models based on similarities, knowledge graphs, and graph neural networks, our model achieves state-of-the-art performance in experiments of 10-fold cross-validation and independent test. Furthermore, we use CKG-IMC to predict compounds interacting with two confirmed AD targets, 42-amino-acid β-amyloid (Aβ42) protein and microtubule-associated protein tau (tau protein), as well as proteins interacting with five FDA-approved anti-AD drugs. The results indicate that the majority of predictions are supported by literature, and molecular docking experiments demonstrate a strong affinity between the predicted compounds and targets.
Collapse
Affiliation(s)
- Yongna Yuan
- School of Information Science & Engineering, Lanzhou University, South Tianshui Road, Lanzhou, 730000, Gansu, China.
| | - Rizhen Hu
- School of Information Science & Engineering, Lanzhou University, South Tianshui Road, Lanzhou, 730000, Gansu, China
| | - Siming Chen
- School of Information Science & Engineering, Lanzhou University, South Tianshui Road, Lanzhou, 730000, Gansu, China
| | - Xiaopeng Zhang
- School of Information Science & Engineering, Lanzhou University, South Tianshui Road, Lanzhou, 730000, Gansu, China
| | - Zhenyu Liu
- School of Information Science & Engineering, Lanzhou University, South Tianshui Road, Lanzhou, 730000, Gansu, China; School of Cyberspace Security, Gansu University of Political Science and Law, Anning West Road, Lanzhou, 730070, Gansu, China
| | - Gonghai Zhou
- School of Information Science & Engineering, Lanzhou University, South Tianshui Road, Lanzhou, 730000, Gansu, China
| |
Collapse
|
43
|
Li Y, Liang W, Peng L, Zhang D, Yang C, Li KC. Predicting Drug-Target Interactions Via Dual-Stream Graph Neural Network. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:948-958. [PMID: 36074878 DOI: 10.1109/tcbb.2022.3204188] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Drug target interaction prediction is a crucial stage in drug discovery. However, brute-force search over a compound database is financially infeasible. We have witnessed the increasing measured drug-target interactions records in recent years, and the rich drug/protein-related information allows the usage of graph machine learning. Despite the advances in deep learning-enabled drug-target interaction, there are still open challenges: (1) rich and complex relationship between drugs and proteins can be explored; (2) the intermediate node is not calibrated in the heterogeneous graph. To tackle with above issues, this paper proposed a framework named DSG-DTI. Specifically, DSG-DTI has the heterogeneous graph autoencoder and heterogeneous attention network-based Matrix Completion. Our framework ensures that the known types of nodes (e.g., drug, target, side effects, diseases) are precisely embedded into high-dimensional space with our pretraining skills. Also, the attention-based heterogeneous graph-based matrix completion achieves highly competitive results via effective long-range dependencies extraction. We verify our model on two public benchmarks. The result of two publicly available benchmark application programs show that the proposed scheme effectively predicts drug-target interactions and can generalize to newly registered drugs and targets with slight performance degradation, outperforming the best accuracy compared with other baselines.
Collapse
|
44
|
Luo Y, Duan G, Zhao Q, Bi X, Wang J. DTKGIN: Predicting drug-target interactions based on knowledge graph and intent graph. Methods 2024; 226:21-27. [PMID: 38608849 DOI: 10.1016/j.ymeth.2024.04.010] [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: 07/14/2023] [Revised: 01/16/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024] Open
Abstract
Knowledge graph intent graph attention mechanism Predicting drug-target interactions (DTIs) plays a crucial role in drug discovery and drug development. Considering the high cost and risk of biological experiments, developing computational approaches to explore the interactions between drugs and targets can effectively reduce the time and cost of drug development. Recently, many methods have made significant progress in predicting DTIs. However, existing approaches still suffer from the high sparsity of DTI datasets and the cold start problem. In this paper, we develop a new model to predict drug-target interactions via a knowledge graph and intent graph named DTKGIN. Our method can effectively capture biological environment information for targets and drugs by mining their associated relations in the knowledge graph and considering drug-target interactions at a fine-grained level in the intent graph. DTKGIN learns the representation of drugs and targets from the knowledge graph and the intent graph. Then the probabilities of interactions between drugs and targets are obtained through the inner product of the representation of drugs and targets. Experimental results show that our proposed method outperforms other state-of-the-art methods in 10-fold cross-validation, especially in cold-start experimental settings. Furthermore, the case studies demonstrate the effectiveness of DTKGIN in predicting potential drug-target interactions. The code is available on GitHub: https://github.com/Royluoyi123/DTKGIN.
Collapse
Affiliation(s)
- Yi Luo
- School of Computer Science and Engineering, Central South University, Changsha 410083, China; Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
| | - Guihua Duan
- School of Computer Science and Engineering, Central South University, Changsha 410083, China; Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China.
| | - Qichang Zhao
- School of Computer Science and Engineering, Central South University, Changsha 410083, China; Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
| | - Xuehua Bi
- School of Computer Science and Engineering, Central South University, Changsha 410083, China; Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
| | - Jianxin Wang
- School of Computer Science and Engineering, Central South University, Changsha 410083, China; Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
| |
Collapse
|
45
|
Cao J, Chen Q, Qiu J, Wang Y, Lan W, Du X, Tan K. NGCN: Drug-target interaction prediction by integrating information and feature learning from heterogeneous network. J Cell Mol Med 2024; 28:e18224. [PMID: 38509739 PMCID: PMC10955156 DOI: 10.1111/jcmm.18224] [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: 12/07/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Drug-target interaction (DTI) prediction is essential for new drug design and development. Constructing heterogeneous network based on diverse information about drugs, proteins and diseases provides new opportunities for DTI prediction. However, the inherent complexity, high dimensionality and noise of such a network prevent us from taking full advantage of these network characteristics. This article proposes a novel method, NGCN, to predict drug-target interactions from an integrated heterogeneous network, from which to extract relevant biological properties and association information while maintaining the topology information. It focuses on learning the topology representation of drugs and targets to improve the performance of DTI prediction. Unlike traditional methods, it focuses on learning the low-dimensional topology representation of drugs and targets via graph-based convolutional neural network. NGCN achieves substantial performance improvements over other state-of-the-art methods, such as a nearly 1.0% increase in AUPR value. Moreover, we verify the robustness of NGCN through benchmark tests, and the experimental results demonstrate it is an extensible framework capable of combining heterogeneous information for DTI prediction.
Collapse
Affiliation(s)
- Junyue Cao
- College of Life Science and TechnologyGuangxi UniversityNanningChina
| | - Qingfeng Chen
- School of Computer, Electronics and InformationGuangxi UniversityNanningChina
| | - Junlai Qiu
- School of Computer, Electronics and InformationGuangxi UniversityNanningChina
| | - Yiming Wang
- School of Computer, Electronics and InformationGuangxi UniversityNanningChina
| | - Wei Lan
- School of Computer, Electronics and InformationGuangxi UniversityNanningChina
| | - Xiaojing Du
- School of Computer, Electronics and InformationGuangxi UniversityNanningChina
| | - Kai Tan
- School of Computer, Electronics and InformationGuangxi UniversityNanningChina
| |
Collapse
|
46
|
Liu C, Xiao K, Yu C, Lei Y, Lyu K, Tian T, Zhao D, Zhou F, Tang H, Zeng J. A probabilistic knowledge graph for target identification. PLoS Comput Biol 2024; 20:e1011945. [PMID: 38578805 PMCID: PMC11034645 DOI: 10.1371/journal.pcbi.1011945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 04/22/2024] [Accepted: 02/24/2024] [Indexed: 04/07/2024] Open
Abstract
Early identification of safe and efficacious disease targets is crucial to alleviating the tremendous cost of drug discovery projects. However, existing experimental methods for identifying new targets are generally labor-intensive and failure-prone. On the other hand, computational approaches, especially machine learning-based frameworks, have shown remarkable application potential in drug discovery. In this work, we propose Progeni, a novel machine learning-based framework for target identification. In addition to fully exploiting the known heterogeneous biological networks from various sources, Progeni integrates literature evidence about the relations between biological entities to construct a probabilistic knowledge graph. Graph neural networks are then employed in Progeni to learn the feature embeddings of biological entities to facilitate the identification of biologically relevant target candidates. A comprehensive evaluation of Progeni demonstrated its superior predictive power over the baseline methods on the target identification task. In addition, our extensive tests showed that Progeni exhibited high robustness to the negative effect of exposure bias, a common phenomenon in recommendation systems, and effectively identified new targets that can be strongly supported by the literature. Moreover, our wet lab experiments successfully validated the biological significance of the top target candidates predicted by Progeni for melanoma and colorectal cancer. All these results suggested that Progeni can identify biologically effective targets and thus provide a powerful and useful tool for advancing the drug discovery process.
Collapse
Affiliation(s)
- Chang Liu
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
| | - Kaimin Xiao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, Beijing, China
| | - Cuinan Yu
- Machine Learning Department, Silexon AI Technology Co., Ltd., Nanjing, Jiangsu Province, China
| | - Yipin Lei
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
| | - Kangbo Lyu
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
| | - Tingzhong Tian
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
| | - Dan Zhao
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
| | - Fengfeng Zhou
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, Jilin Province, China
| | - Haidong Tang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Jianyang Zeng
- School of Engineering, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Research Center for Industries of the Future and School of Engineering, Westlake University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
47
|
Ghandikota SK, Jegga AG. Application of artificial intelligence and machine learning in drug repurposing. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 205:171-211. [PMID: 38789178 DOI: 10.1016/bs.pmbts.2024.03.030] [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: 05/26/2024]
Abstract
The purpose of drug repurposing is to leverage previously approved drugs for a particular disease indication and apply them to another disease. It can be seen as a faster and more cost-effective approach to drug discovery and a powerful tool for achieving precision medicine. In addition, drug repurposing can be used to identify therapeutic candidates for rare diseases and phenotypic conditions with limited information on disease biology. Machine learning and artificial intelligence (AI) methodologies have enabled the construction of effective, data-driven repurposing pipelines by integrating and analyzing large-scale biomedical data. Recent technological advances, especially in heterogeneous network mining and natural language processing, have opened up exciting new opportunities and analytical strategies for drug repurposing. In this review, we first introduce the challenges in repurposing approaches and highlight some success stories, including those during the COVID-19 pandemic. Next, we review some existing computational frameworks in the literature, organized on the basis of the type of biomedical input data analyzed and the computational algorithms involved. In conclusion, we outline some exciting new directions that drug repurposing research may take, as pioneered by the generative AI revolution.
Collapse
Affiliation(s)
- Sudhir K Ghandikota
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Anil G Jegga
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.
| |
Collapse
|
48
|
Tian Z, Han C, Xu L, Teng Z, Song W. MGCNSS: miRNA-disease association prediction with multi-layer graph convolution and distance-based negative sample selection strategy. Brief Bioinform 2024; 25:bbae168. [PMID: 38622356 PMCID: PMC11018511 DOI: 10.1093/bib/bbae168] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/14/2024] [Accepted: 03/31/2024] [Indexed: 04/17/2024] Open
Abstract
Identifying disease-associated microRNAs (miRNAs) could help understand the deep mechanism of diseases, which promotes the development of new medicine. Recently, network-based approaches have been widely proposed for inferring the potential associations between miRNAs and diseases. However, these approaches ignore the importance of different relations in meta-paths when learning the embeddings of miRNAs and diseases. Besides, they pay little attention to screening out reliable negative samples which is crucial for improving the prediction accuracy. In this study, we propose a novel approach named MGCNSS with the multi-layer graph convolution and high-quality negative sample selection strategy. Specifically, MGCNSS first constructs a comprehensive heterogeneous network by integrating miRNA and disease similarity networks coupled with their known association relationships. Then, we employ the multi-layer graph convolution to automatically capture the meta-path relations with different lengths in the heterogeneous network and learn the discriminative representations of miRNAs and diseases. After that, MGCNSS establishes a highly reliable negative sample set from the unlabeled sample set with the negative distance-based sample selection strategy. Finally, we train MGCNSS under an unsupervised learning manner and predict the potential associations between miRNAs and diseases. The experimental results fully demonstrate that MGCNSS outperforms all baseline methods on both balanced and imbalanced datasets. More importantly, we conduct case studies on colon neoplasms and esophageal neoplasms, further confirming the ability of MGCNSS to detect potential candidate miRNAs. The source code is publicly available on GitHub https://github.com/15136943622/MGCNSS/tree/master.
Collapse
Affiliation(s)
- Zhen Tian
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450000, China
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China
| | - Chenguang Han
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450000, China
| | - Lewen Xu
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450000, China
| | - Zhixia Teng
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Wei Song
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
49
|
Zhang Y, Li S, Meng K, Sun S. Machine Learning for Sequence and Structure-Based Protein-Ligand Interaction Prediction. J Chem Inf Model 2024; 64:1456-1472. [PMID: 38385768 DOI: 10.1021/acs.jcim.3c01841] [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: 02/23/2024]
Abstract
Developing new drugs is too expensive and time -consuming. Accurately predicting the interaction between drugs and targets will likely change how the drug is discovered. Machine learning-based protein-ligand interaction prediction has demonstrated significant potential. In this paper, computational methods, focusing on sequence and structure to study protein-ligand interactions, are examined. Therefore, this paper starts by presenting an overview of the data sets applied in this area, as well as the various approaches applied for representing proteins and ligands. Then, sequence-based and structure-based classification criteria are subsequently utilized to categorize and summarize both the classical machine learning models and deep learning models employed in protein-ligand interaction studies. Moreover, the evaluation methods and interpretability of these models are proposed. Furthermore, delving into the diverse applications of protein-ligand interaction models in drug research is presented. Lastly, the current challenges and future directions in this field are addressed.
Collapse
Affiliation(s)
- Yunjiang Zhang
- Beijing Key Laboratory for Green Catalysis and Separation, The Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P. R. China
| | - Shuyuan Li
- Beijing Key Laboratory for Green Catalysis and Separation, The Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P. R. China
| | - Kong Meng
- Beijing Key Laboratory for Green Catalysis and Separation, The Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P. R. China
| | - Shaorui Sun
- Beijing Key Laboratory for Green Catalysis and Separation, The Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P. R. China
| |
Collapse
|
50
|
E Z, Qiao G, Wang G, Li Y. GSL-DTI: Graph structure learning network for Drug-Target interaction prediction. Methods 2024; 223:136-145. [PMID: 38360082 DOI: 10.1016/j.ymeth.2024.01.018] [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: 07/22/2023] [Revised: 12/23/2023] [Accepted: 01/29/2024] [Indexed: 02/17/2024] Open
Abstract
MOTIVATION Drug-target interaction prediction is an important area of research to predict whether there is an interaction between a drug molecule and its target protein. It plays a critical role in drug discovery and development by facilitating the identification of potential drug candidates and expediting the overall process. Given the time-consuming, expensive, and high-risk nature of traditional drug discovery methods, the prediction of drug-target interactions has become an indispensable tool. Using machine learning and deep learning to tackle this class of problems has become a mainstream approach, and graph-based models have recently received much attention in this field. However, many current graph-based Drug-Target Interaction (DTI) prediction methods rely on manually defined rules to construct the Drug-Protein Pair (DPP) network during the DPP representation learning process. However, these methods fail to capture the true underlying relationships between drug molecules and target proteins. RESULTS We propose GSL-DTI, an automatic graph structure learning model used for predicting drug-target interactions (DTIs). Initially, we integrate large-scale heterogeneous networks using a graph convolution network based on meta-paths, effectively learning the representations of drugs and target proteins. Subsequently, we construct drug-protein pairs based on these representations. In contrast to previous studies that construct DPP networks based on manual rules, our method introduces an automatic graph structure learning approach. This approach utilizes a filter gate on the affinity scores of DPPs and relies on the classification loss of downstream tasks to guide the learning of the underlying DPP network structure. Based on the learned DPP network, we transform the prediction of drug-target interactions into a node classification problem. The comprehensive experiments conducted on three public datasets have shown the superiority of GSL-DTI in the tasks of DTI prediction. Additionally, GSL-DTI provides a fresh perspective for advancing research in graph structure learning for DTI prediction.
Collapse
Affiliation(s)
- Zixuan E
- College of Computer and Control Engineering, Northeast Forestry University,Harbin 150006, China
| | - Guanyu Qiao
- College of Computer and Control Engineering, Northeast Forestry University,Harbin 150006, China
| | - Guohua Wang
- College of Computer and Control Engineering, Northeast Forestry University,Harbin 150006, China.
| | - Yang Li
- College of Computer and Control Engineering, Northeast Forestry University,Harbin 150006, China.
| |
Collapse
|