1
|
Cheng Z, Wang Z, Tang X, Hu X, Yang F, Yan X. A Multi-View Feature-Based Interpretable Deep Learning Framework for Drug-Drug Interaction Prediction. Interdiscip Sci 2025; 17:437-448. [PMID: 39899225 DOI: 10.1007/s12539-025-00687-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 01/06/2025] [Indexed: 02/04/2025]
Abstract
Drug-drug interactions (DDIs) can result in deleterious consequences when patients take multiple medications simultaneously, emphasizing the critical need for accurate DDI prediction. Computational methods for DDI prediction have garnered recent attention. However, current approaches concentrate solely on single-view features, such as atomic-view or substructure-view features, limiting predictive capacity. The scarcity of research on interpretability studies based on multi-view features is crucial for tracing interactions. Addressing this gap, we present MI-DDI, a multi-view feature-based interpretable deep learning framework for DDI. To fully extract multi-view features, we employ a Message Passing Neural Network (MPNN) to learn atomic features from molecular graphs generated by RDkit, and transformer encoders are used to learn substructure-view embeddings from drug SMILES simultaneously. These atomic-view and substructure-view features are then amalgamated into a holistic drug embedding matrix. Subsequently, an intricately designed interaction module not only establishes a tractable path for understanding interactions but also directly informs the construction of weight matrices, enabling precise and interpretable interaction predictions. Validation on the BIOSNAP dataset and DrugBank dataset demonstrates MI-DDI's superiority. It surpasses the current benchmarks by a substantial average of 3% on BIOSNAP and 1% on DrugBank. Additional experiments underscore the significance of atomic-view information for DDI prediction and confirm that our interaction module indeed learns more effective information for DDI prediction. The source codes are available at https://github.com/ZihuiCheng/MI-DDI .
Collapse
Affiliation(s)
- Zihui Cheng
- School of Computer Science and Artificial Intelligence, Wuhan Textile University, Sunshine Avenue, Wuhan, 430200, China
| | - Zhaojing Wang
- School of Computer Science and Artificial Intelligence, Wuhan Textile University, Sunshine Avenue, Wuhan, 430200, China.
- Textile and Fashion, Hubei Provincial Engineering Research Center for Intelligence, Sunshine Avenue, Wuhan, 430200, China.
| | - Xianfang Tang
- School of Computer Science and Artificial Intelligence, Wuhan Textile University, Sunshine Avenue, Wuhan, 430200, China
- Textile and Fashion, Hubei Provincial Engineering Research Center for Intelligence, Sunshine Avenue, Wuhan, 430200, China
| | - Xinrong Hu
- School of Computer Science and Artificial Intelligence, Wuhan Textile University, Sunshine Avenue, Wuhan, 430200, China
- Textile and Fashion, Hubei Provincial Engineering Research Center for Intelligence, Sunshine Avenue, Wuhan, 430200, China
| | - Fei Yang
- Electronic Information School, Wuhan University, Bayi Road, Wuhan, 430072, China
| | - Xiaoyun Yan
- School of Computer Science and Artificial Intelligence, Wuhan Textile University, Sunshine Avenue, Wuhan, 430200, China
- Textile and Fashion, Hubei Provincial Engineering Research Center for Intelligence, Sunshine Avenue, Wuhan, 430200, China
| |
Collapse
|
2
|
Zhao L, Shang J, Meng X, He X, Zhang Y, Liu JX. Adaptive Multi-Kernel Graph Neural Network for Drug-Drug Interaction Prediction. Interdiscip Sci 2025; 17:409-423. [PMID: 39873945 DOI: 10.1007/s12539-024-00684-1] [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: 05/31/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 01/30/2025]
Abstract
Combination therapy, which synergistically enhances treatment efficacy and inhibits disease progression through the combined effects of multiple drugs, has emerged as a mainstream approach for treating complex diseases and alleviating symptoms. However, drug-drug interactions (DDIs) can sometimes lead to adverse reactions, potentially endangering lives. Therefore, developing efficient and accurate DDI prediction methods is crucial for elucidating drug mechanisms and preventing side effects. Current prediction methods often focus solely on the presence of interactions between drugs when constructing DDI graphs, neglecting the specific types of DDIs. This oversight can result in a decline in predictive performance. To address this issue, we propose an Adaptive Multi-Kernel Graph Neural Network (AMKGNN) for DDI prediction. AMKGNN differentiates DDIs into increase-type and decrease-type interactions, constructing separate increased DDI and decreased DDI graphs as convolutional kernels. AMKGNN employs a graph kernel learning mechanism that adaptively determines the optimal threshold between high-frequency and low-frequency signals in the network to capture node embeddings. Initially, AMKGNN learns drug embedding representations based on these two graph convolutional kernels and various drug features. These representations are then concatenated and input into a deep neural network to predict potential DDIs. The results show that our model achieved AUC and AUPR values above 90% across three sub-tasks on two datasets, significantly outperforming the other five comparison models. Furthermore, ablation experiments and case studies validate the superiority of AMKGNN.
Collapse
Affiliation(s)
- Linqian Zhao
- School of Computer Science, Qufu Normal University, Rizhao, 276826, China
| | - Junliang Shang
- School of Computer Science, Qufu Normal University, Rizhao, 276826, China.
| | - Xianghan Meng
- School of Computer Science, Qufu Normal University, Rizhao, 276826, China
| | - Xin He
- School of Computer Science, Qufu Normal University, Rizhao, 276826, China
| | - Yuanyuan Zhang
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao, 266520, China
| | - Jin-Xing Liu
- School of Computer Science, Qufu Normal University, Rizhao, 276826, China
- School of Health and Life Science, University of Health and Rehabilitation Sciences, Qingdao, 266113, China
| |
Collapse
|
3
|
Kim D, Yu J, Bae SH, Lee J. SF-Rx: A Multioutput Deep Neural Network-Based Framework Predicting Drug-Drug Interaction under Realistic Conditions for Safe Prescription. J Chem Inf Model 2025; 65:4442-4457. [PMID: 40310752 DOI: 10.1021/acs.jcim.5c00075] [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: 05/03/2025]
Abstract
Drug-drug interaction (DDI) can compromise therapeutic efficacy and cause detrimental effects in polypharmacy. Computational prediction of DDI has emerged as an alternative approach to time-consuming clinical experiments for investigating potential drug interactions, yet reliable prediction remains challenging. We present SF-Rx (Safe Prescription), a DDI predictive framework that incorporates structural similarity profiles with pharmacokinetic (PK) and pharmacodynamic (PD) features to predict severity, types, and directionality. Our study employs a scaffold-based cross-validation strategy for paired drugs and enables a realistic evaluation of model performance while quantifying prediction uncertainty. The implementation of federated learning across multiple DDI data sets improves model generalization and overcomes limited chemical diversity in single-source data sets. Our framework provides a promising approach for developing a reliable DDI prediction model under real-world scenarios, potentially improving patient safety in multidrug treatments.
Collapse
Affiliation(s)
- Daeun Kim
- DR. NOAH BIOTECH Inc., 91, Changnyong-daero 256beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do 16229, Republic of Korea
| | - Jaehong Yu
- DR. NOAH BIOTECH Inc., 91, Changnyong-daero 256beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do 16229, Republic of Korea
| | - Sang-Hun Bae
- DR. NOAH BIOTECH Inc., 91, Changnyong-daero 256beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do 16229, Republic of Korea
| | - Jihyun Lee
- DR. NOAH BIOTECH Inc., 91, Changnyong-daero 256beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do 16229, Republic of Korea
| |
Collapse
|
4
|
Lin K, Huang F, Zhuo S, Zhou Q, Pan D, Yu S, Yang F. Asymmetric drug interaction prediction via multi-scale fusion of directed topological relationships and drug features. Comput Biol Chem 2025; 118:108491. [PMID: 40328049 DOI: 10.1016/j.compbiolchem.2025.108491] [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: 12/20/2024] [Revised: 03/18/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025]
Abstract
Drug-drug interactions (DDIs), particularly asymmetric DDIs, pose a critical challenge to patient safety, potentially leading to severe side effects or reduced therapeutic efficacy in combination therapies. However, existing models largely overlook the directionality of interactions and fail to capture the multi-scale topological relationships among drugs, which are crucial in reflecting both local and global interaction patterns within directed drug-drug interaction (DDI) networks. To address these gaps, we propose ADI-MSF, a novel asymmetric DDI prediction model. ADI-MSF integrates directed topological information and drug self-features through a dual-channel multi-scale encoder. The topological encoder utilizes a multi-layer graph attention network to extract first- and second-order directed neighborhood embeddings, whereas the feature encoder employs an autoencoder to generate multi-scale drug representations. These representations were fused using summation and the Hadamard product to form comprehensive drug pair embeddings for downstream predictions. Experiments on two real-world datasets, covering asymmetric DDI prediction (Task 1) and direction prediction (Task 2), demonstrated the superior performance of ADI-MSF, achieving over 95% accuracy and F1 scores for Task 1 and over 92% F1 for Task 2. Ablation studies confirmed the significance of each module, whereas case studies validated the effectiveness of modeling asymmetric DDIs. The source code and data are available at https://github.com/FengxinHuang/ADI-MSF.
Collapse
Affiliation(s)
- Kaibiao Lin
- School of Computer and Information Engineering, Xiamen University of Technology, Xiamen, 361024, Fujian, China
| | - Fengxin Huang
- School of Computer and Information Engineering, Xiamen University of Technology, Xiamen, 361024, Fujian, China
| | - Shuangta Zhuo
- The Second Affiliated Hospital of Xiamen Medical College, Xiamen, 361024, Fujian, China
| | - Qiqi Zhou
- The Second Affiliated Hospital of Xiamen Medical College, Xiamen, 361024, Fujian, China
| | - Dawei Pan
- School of Computer and Information Engineering, Xiamen University of Technology, Xiamen, 361024, Fujian, China.
| | - Shenbao Yu
- College of Computer and Cyber Security, Fujian Normal University, Fuzhou, 350117, Fujian, China
| | - Fan Yang
- Department of Automation, Xiamen University, Xiamen, 361102, Fujian, China.
| |
Collapse
|
5
|
Halder A, Saha B, Roy M, Majumder S. A novel deep sequential learning architecture for drug drug interaction prediction using DDINet. Sci Rep 2025; 15:9337. [PMID: 40102542 PMCID: PMC11920219 DOI: 10.1038/s41598-025-93952-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
Drug drug Interactions (DDI) present considerable challenges in healthcare, often resulting in adverse effects or decreased therapeutic efficacy. This article proposes a novel deep sequential learning architecture called DDINet to predict and classify DDIs between pairs of drugs based on different mechanisms viz., Excretion, Absorption, Metabolism, and Excretion rate (higher serum level) etc. Chemical features such as Hall Smart, Amino Acid count and Carbon types are extracted from each drug (pairs) to apply as an input to the proposed model. Proposed DDINet incorporates attention mechanism and deep sequential learning architectures, such as Long Short-Term Memory and gated recurrent unit. It utilizes the Rcpi toolkit to extract biochemical features of drugs from their chemical composition in Simplified Molecular-Input Line-Entry System format. Experiments are conducted on publicly available DDI datasets from DrugBank and Kaggle. The model's efficacy in predicting and classifying DDIs is evaluated using various performance measures. The experimental results show that DDINet outperformed eight counterpart techniques achieving [Formula: see text] overall accuracy which is also statistically confirmed by Confidence Interval tests and paired t-tests. This architecture may act as an effective computational technique for drug drug interaction with respect to mechanism which may act as a complementary tool to reduce costly wet lab experiments for DDI prediction and classification.
Collapse
Affiliation(s)
- Anindya Halder
- Department of Computer Application, School of Technology, North-Eastern Hill University, Tura Campus, Tura, Meghalaya, 794002, India.
| | - Biswanath Saha
- Department of Computer Application, School of Technology, North-Eastern Hill University, Tura Campus, Tura, Meghalaya, 794002, India.
| | - Moumita Roy
- Department of Computer Science and Engineering, University of Kalyani, Kalyani, West Bengal, 741235, India.
| | - Sukanta Majumder
- Department of Computer Science and Engineering, University of Kalyani, Kalyani, West Bengal, 741235, India.
| |
Collapse
|
6
|
Jung S, Yoo S. Interpretable prediction of drug-drug interactions via text embedding in biomedical literature. Comput Biol Med 2025; 185:109496. [PMID: 39626457 DOI: 10.1016/j.compbiomed.2024.109496] [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: 08/12/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/26/2025]
Abstract
Polypharmacy is a promising approach for treating diseases, especially those with complex symptoms. However, it can lead to unexpected drug-drug interactions (DDIs), potentially reducing efficacy and triggering adverse drug reactions (ADRs). Predicting the risk of DDIs is crucial for ensuring safe drug use, particularly by identifying the types of DDIs and the mechanisms involved. Therefore, this study used biomedical literature to proposed hierarchical attention-based deep learning models to predict DDIs and their types. The proposed model consists of two components: drug embedding and DDI prediction. The drug embedding module extracts representation vectors that effectively capture drug properties using sentence and sequence embedding methods. For sentence embedding, a pre-trained biomedical language model is used to map drug-related sentences into vector space. For sequence embedding, sentence embedding vectors are sequentially fed into bidirectional long short-term memory with a hierarchical attention network, enabling the analysis of sentences relevant to DDI prediction while accounting for the order of the sentences. Finally, DDI prediction is performed using a deep neural network based on the sequence embedding vectors of a drug pair. Our model achieved high performances in the accuracy (0.85-0.90), AUROC (0.98-0.99), and AUPR (0.63-0.95) performance across 164 DDI types. Additionally, the proposed model showed improvements in up to 11 % in AUROC, and 8 % in AUPR. Furthermore, model interprets predictions by leveraging attention mechanisms and drug similarity. The results indicated that the model considered various factors beyond similarity to predict DDIs. These findings may help prevent unforeseen medical accidents and reduce healthcare costs by predicting detailed drug interaction types.
Collapse
Affiliation(s)
- Sunwoo Jung
- Department of Intelligent Electronics and Computer Engineering, Chonnam National University, Gwangju, 61186, South Korea.
| | - Sunyong Yoo
- Department of Intelligent Electronics and Computer Engineering, Chonnam National University, Gwangju, 61186, South Korea.
| |
Collapse
|
7
|
Gao S, Xie J, Zhao Y. A Multi-Source drug combination and Omnidirectional feature fusion approach for predicting Drug-Drug interaction events. J Biomed Inform 2025; 162:104772. [PMID: 39814273 DOI: 10.1016/j.jbi.2025.104772] [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: 10/11/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND In the medical context where polypharmacy is increasingly common, accurately predicting drug-drug interactions (DDIs) is necessary for enhancing clinical medication safety and personalized treatment. Despite progress in identifying potential DDIs, a deep understanding of the underlying mechanisms of DDIs remains limited, constraining the rapid development and clinical application of new drugs. METHODS This study introduces a novel multimodal drug-drug interaction (MMDDI) model based on multi-source drug data and comprehensive feature fusion techniques, aiming to improve the accuracy and depth of DDI prediction. We utilized the real-world DrugBank dataset, which contains rich drug information. Our task was to predict multiple interaction events between drug pairs and analyze the underlying mechanisms of these interactions. The MMDDI model achieves precise predictions through four key stages: feature extraction, drug pairing strategy, fusion network, and multi-source feature integration. We employed advanced data fusion techniques and machine learning algorithms for multidimensional analysis of drug features and interaction events. RESULTS The MMDDI model was comprehensively evaluated on three representative prediction tasks. Experimental results demonstrated that the MMDDI model outperforms existing technologies in terms of predictive accuracy, generalization ability, and interpretability. Specifically, the MMDDI model achieved an accuracy of 93% on the test set, and the area under the AUC-ROC curve reached 0.9505, showing excellent predictive performance. Furthermore, the model's interpretability analysis revealed the complex relationships between drug features and interaction mechanisms, providing new insights for clinical medication decisions. CONCLUSION The MMDDI model not only improves the accuracy of DDI prediction but also provides significant scientific support for clinical medication safety and drug development by deeply analyzing the mechanisms of drug interactions. These findings have the potential to improve patient medication outcomes and contribute to the development of personalized medicine.
Collapse
Affiliation(s)
- Shiwei Gao
- Northwest Normal University, College of Computer Science and Engineering, Lanzhou, China.
| | - Jingjing Xie
- Northwest Normal University, College of Computer Science and Engineering, Lanzhou, China.
| | - Yizhao Zhao
- Northwest Normal University, College of Computer Science and Engineering, Lanzhou, China.
| |
Collapse
|
8
|
Xia Y, Xiong A, Zhang Z, Zou Q, Cui F. A comprehensive review of deep learning-based approaches for drug-drug interaction prediction. Brief Funct Genomics 2025; 24:elae052. [PMID: 39987494 PMCID: PMC11847217 DOI: 10.1093/bfgp/elae052] [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: 04/30/2024] [Revised: 07/29/2024] [Accepted: 02/21/2025] [Indexed: 02/25/2025] Open
Abstract
Deep learning models have made significant progress in the biomedical field, particularly in the prediction of drug-drug interactions (DDIs). DDIs are pharmacodynamic reactions between two or more drugs in the body, which may lead to adverse effects and are of great significance for drug development and clinical research. However, predicting DDI through traditional clinical trials and experiments is not only costly but also time-consuming. When utilizing advanced Artificial Intelligence (AI) and deep learning techniques, both developers and users face multiple challenges, including the problem of acquiring and encoding data, as well as the difficulty of designing computational methods. In this paper, we review a variety of DDI prediction methods, including similarity-based, network-based, and integration-based approaches, to provide an up-to-date and easy-to-understand guide for researchers in different fields. Additionally, we provide an in-depth analysis of widely used molecular representations and a systematic exposition of the theoretical framework of models used to extract features from graph data.
Collapse
Affiliation(s)
- Yan Xia
- School of Computer Science and Technology, Hainan University, No. 58, Renmin Avenue, Haidian Island, Haikou, Hainan Province, 570228, China
| | - An Xiong
- School of Computer Science and Technology, Hainan University, No. 58, Renmin Avenue, Haidian Island, Haikou, Hainan Province, 570228, China
| | - Zilong Zhang
- School of Computer Science and Technology, Hainan University, No. 58, Renmin Avenue, Haidian Island, Haikou, Hainan Province, 570228, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, No. 4, Section 2, Jianshe North Road, Chengdu, Sichuan Province, 610054, China
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, No. 1, Chengdian Road, Kecheng District, Quzhou, Zhejiang Province, 324000, China
| | - Feifei Cui
- School of Computer Science and Technology, Hainan University, No. 58, Renmin Avenue, Haidian Island, Haikou, Hainan Province, 570228, China
| |
Collapse
|
9
|
Zhang L, Sheng Y, Yang J, Hu Z, Peng B. Predicting the toxic side effects of drug interactions using chemical structures and protein sequences. Sci Rep 2024; 14:31503. [PMID: 39733005 PMCID: PMC11682051 DOI: 10.1038/s41598-024-82981-9] [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: 10/03/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
The study aims to address the critical issue of toxic side effects resulting from drug combinations, which can significantly increase health risks, clinical complications, and lead to drug being withdrawn from the market. A model named TSEDDI (toxic side effects of drug-drug interaction) has been developed to improve the identification of drug pairs that may induce toxicity or adverse reactions. By utilizing drug chemical structures and diverse proteins, we employ a convolutional neural network (CNN) to extract features from molecular images, enzyme proteins, transporter proteins, and target proteins. Furthermore, we introduce a weighted binary cross entropy loss function to tackle class imbalance and integrate the multi-head attention mechanism with residual connections to enhance model performance. Our model outperformed advanced baseline models in predicting drug-drug interaction (DDI) side effects, achieving an accuracy of 0.9059 (± 0.0010) and consistently excelling across various evaluation metrics. The case study confirms the potential mechanisms by which four pairs of drugs cause side effects, thus demonstrating the effectiveness of our model in predicting DDI side effects. The TSEDDI model combines multiple attention mechanisms and residual connections, enhancing its ability to detect toxic and adverse effects related to DDIs. As a result, it becomes a valuable resource for promptly identifying adverse reactions in clinical trials. Future research could investigate drug substructures prone to toxic side effects.
Collapse
Affiliation(s)
- Liyuan Zhang
- School of Public Health, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yongxin Sheng
- School of Public Health, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Jinxiang Yang
- School of Public Health, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Zuhai Hu
- School of Public Health, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Bin Peng
- School of Public Health, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
10
|
Jiang Z, Ding P, Shen C, Dai X. Geometric Molecular Graph Representation Learning Model for Drug-Drug Interactions Prediction. IEEE J Biomed Health Inform 2024; 28:7623-7632. [PMID: 39226203 DOI: 10.1109/jbhi.2024.3453956] [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: 09/05/2024]
Abstract
Drug-drug interaction (DDI) can trigger many adverse effects in patients and has emerged as a threat to medicine and public health. Therefore, it is important to predict potential drug interactions since it can provide combination strategies of drugs for systematic and effective treatment. Existing deep learning-based methods often rely on DDI functional networks, or use them as an important part of the model information source. However, it is difficult to discover the interactions of a new drug. To address the above limitations, we propose a geometric molecular graph representation learning model (Mol-DDI) for DDI prediction based on the basic assumption that structure determines function. Mol-DDI only considers the covalent and non-covalent bond information of molecules, then it uses the pre-training idea of large-scale models to learn drug molecular representations and predict drug interactions during the fine-tuning process. Experimental results show that the Mol-DDI model outperforms others on the three datasets and performs better in predicting new drug interaction experiments.
Collapse
|
11
|
Li Y, You ZH, Yuan Y, Mi CG, Huang YA, Yi HC, Hou LX. Integrated Knowledge Graph and Drug Molecular Graph Fusion via Adversarial Networks for Drug-Drug Interaction Prediction. J Chem Inf Model 2024; 64:8361-8372. [PMID: 39475566 DOI: 10.1021/acs.jcim.4c01647] [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: 11/12/2024]
Abstract
The Co-administration of multiple drugs can enhance the efficacy of disease treatment by reducing drug resistance and side effects. However, it also raises the risk of adverse drug interactions, presenting a challenging problem in healthcare. Various approaches have been developed to predict drug-drug interactions (DDIs) by leveraging both knowledge graphs and drug attribute information. While these methods have shown promise, they often fail to effectively capture correlations between biomedical information in the knowledge graph and drug properties. This work introduces a novel end-to-end DDI predictor framework based on generative adversarial networks. This framework utilizes a message-passing neural network to capture molecular structure information while employing the knowledge-aware graph attention network to capture the representation of drugs in the knowledge graph through considering the importance of different multihop neighbor nodes and relationships. The dual generative adversarial networks employ two generators and two discriminators in knowledge graph embedding and molecular topology embedding for adversarial training to capture the interrelations and complementary knowledge between molecular structure information and semantic information from the knowledge graph. comprehensive experiments have demonstrated that the proposed method outperforms state-of-the-art algorithms in binary classification, with improvements of 1.0% in accuracy, 0.45% in area under the receiver operating characteristic curve (AUC), 0.24% in area under the precision-recall curve (AUPR), and 0.98% in F1 score. Furthermore, for multiclass classification tasks, improvements were observed across various evaluation metrics, including 0.9% in accuracy, 0.25% in macro precision, 0.2% in macro recall, and 0.28% in macro F1. Additionally, ablation studies were conducted to showcase the effectiveness and robustness of our method in DDI prediction tasks.
Collapse
Affiliation(s)
- Yu Li
- School of Computer Science, Northwestern Polytechnical University, Xi'an710129, China
| | - Zhu-Hong You
- School of Computer Science, Northwestern Polytechnical University, Xi'an710129, China
| | - Yang Yuan
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou213164, China
| | - Cheng-Gang Mi
- Foreign Language and Literature Institute, Xi'an International Studies University, Xi'an710129, China
| | - Yu-An Huang
- School of Computer Science, Northwestern Polytechnical University, Xi'an710129, China
| | - Hai-Cheng Yi
- School of Computer Science, Northwestern Polytechnical University, Xi'an710129, China
| | - Lin-Xuan Hou
- School of Computer Science, Northwestern Polytechnical University, Xi'an710129, China
| |
Collapse
|
12
|
Kha QH, Nguyen NTK, Le NQK, Kang JH. Development and validation of a machine learning model for predicting drug-drug interactions with oral diabetes medications. Methods 2024; 232:81-88. [PMID: 39489198 DOI: 10.1016/j.ymeth.2024.10.012] [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: 08/31/2024] [Revised: 10/03/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024] Open
Abstract
Diabetes management is often complicated by comorbidities, requiring complex medication regimens that increase the risk of drug-drug interactions (DDIs), potentially compromising treatment outcomes or causing toxicity. Although machine learning (ML) models have made strides in DDI prediction, existing approaches lack specificity for oral diabetes medications and face challenges in interpretability. To address these limitations, we propose a novel ML-based framework utilizing the Simplified Molecular Input Line Entry System (SMILES) to encode structural information of oral diabetes drugs. Using this representation, we developed an XGBoost model, selecting molecular features through LASSO. Our dataset, sourced from DrugBank, included 42 oral diabetes drugs and 1,884 interacting drugs, divided into training, validation, and testing sets. The model identified 606 optimal features, achieving an F1-score of 0.8182. SHAP analysis was employed for feature interpretation, enhancing model transparency and clinical relevance. By predicting adverse DDIs, our model offers a valuable tool for clinical decision-making, aiding safer prescription practices. The 606 critical features provide insights into atomic-level interactions, linking computational predictions with biological experiments. We present a classification model specifically designed for predicting DDIs associated with oral diabetes medications, with an openly accessible web application to support diabetes management in multi-drug regimens and comorbidity settings.
Collapse
Affiliation(s)
- Quang-Hien Kha
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; AIBioMed Research Group, Taipei Medical University, Taipei 110, Taiwan
| | - Ngan Thi Kim Nguyen
- Programs of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei 106, Taiwan
| | - Nguyen Quoc Khanh Le
- AIBioMed Research Group, Taipei Medical University, Taipei 110, Taiwan; In-Service Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan.
| | - Jiunn-Horng Kang
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Department of Physical Medicine and Rehabilitation, Taipei Medical University Hospital, Taipei 110, Taiwan; Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
13
|
Mir A, Zhu A, Lau R, Barr N, Sheikh Z, Acuna D, Dayal A, Hibino N. Applications, Limitations, and Considerations of Clinical Trials in a Dish. Bioengineering (Basel) 2024; 11:1096. [PMID: 39593756 PMCID: PMC11591410 DOI: 10.3390/bioengineering11111096] [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: 10/07/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Recent advancements in biotechnology forged the path for clinical trials in dish (CTiDs) to advance as a popular method of experimentation in biomedicine. CTiDs play a fundamental role in translational research through technologies such as induced pluripotent stem cells, whole genome sequencing, and organs-on-a-chip. In this review, we explore advancements that enable these CTiD biotechnologies and their applications in animal testing, disease modeling, and space radiation technologies. Furthermore, this review dissects the advantages and disadvantages of CTiDs, as well as their regulatory considerations. Lastly, we evaluate the challenges that CTiDs pose and the role of CTiDs in future experimentation.
Collapse
Affiliation(s)
- Amatullah Mir
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Angie Zhu
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Rico Lau
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Nicolás Barr
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Zyva Sheikh
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Diana Acuna
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Anuhya Dayal
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Narutoshi Hibino
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
- Pediatric Cardiac Surgery, Advocate Children’s Hospital, 4440 W 95th St., Oak Lawn, IL 60453, USA
| |
Collapse
|
14
|
Cingiz MÖ. Ensemble decision of local similarity indices on the biological network for disease related gene prediction. PeerJ 2024; 12:e17975. [PMID: 39247551 PMCID: PMC11380840 DOI: 10.7717/peerj.17975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/05/2024] [Indexed: 09/10/2024] Open
Abstract
Link prediction (LP) is a task for the identification of potential, missing and spurious links in complex networks. Protein-protein interaction (PPI) networks are important for understanding the underlying biological mechanisms of diseases. Many complex networks have been constructed using LP methods; however, there are a limited number of studies that focus on disease-related gene predictions and evaluate these genes using various evaluation criteria. The main objective of the study is to investigate the effect of a simple ensemble method in disease related gene predictions. Local similarity indices (LSIs) based disease related gene predictions were integrated by a simple ensemble decision method, simple majority voting (SMV), on the PPI network to detect accurate disease related genes. Human PPI network was utilized to discover potential disease related genes using four LSIs for the gene prediction. LSIs discovered potential links between disease related genes, which were obtained from OMIM database for gastric, colorectal, breast, prostate and lung cancers. LSIs based disease related genes were ranked due to their LSI scores in descending order for retrieving the top 10, 50 and 100 disease related genes. SMV integrated four LSIs based predictions to obtain SMV based the top 10, 50 and 100 disease related genes. The performance of LSIs based and SMV based genes were evaluated separately by employing overlap analyses, which were performed with GeneCard disease-gene relation dataset and Gene Ontology (GO) terms. The GO-terms were used for biological assessment for the inferred gene lists by LSIs and SMV on all cancer types. Adamic-Adar (AA), Resource Allocation Index (RAI), and SMV based gene lists are generally achieved good performance results on all cancers in both overlap analyses. SMV also outperformed on breast cancer data. The increment in the selection of the number of the top ranked disease related genes also enhanced the performance results of SMV.
Collapse
Affiliation(s)
- Mustafa Özgür Cingiz
- Department of Computer Engineering, Faculty of Engineering and Natural Sciences, Bursa Technical University, Bursa, Turkey
| |
Collapse
|
15
|
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
|
16
|
Nguyen DA, Nguyen CH, Mamitsuka H. Central-Smoothing Hypergraph Neural Networks for Predicting Drug-Drug Interactions. IEEE TRANSACTIONS ON NEURAL NETWORKS AND LEARNING SYSTEMS 2024; 35:11620-11625. [PMID: 37018091 DOI: 10.1109/tnnls.2023.3261860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Predicting drug-drug interactions (DDIs) is the problem of predicting side effects (unwanted outcomes) of a pair of drugs using drug information and known side effects of many pairs. This problem can be formulated as predicting labels (i.e., side effects) for each pair of nodes in a DDI graph, of which nodes are drugs and edges are interacting drugs with known labels. State-of-the-art methods for this problem are graph neural networks (GNNs), which leverage neighborhood information in the graph to learn node representations. For DDI, however, there are many labels with complicated relationships due to the nature of side effects. Usual GNNs often fix labels as one-hot vectors that do not reflect label relationships and potentially do not obtain the highest performance in the difficult cases of infrequent labels. In this brief, we formulate DDI as a hypergraph where each hyperedge is a triple: two nodes for drugs and one node for a label. We then present CentSmoothie, a hypergraph neural network (HGNN) that learns representations of nodes and labels altogether with a novel " central-smoothing " formulation. We empirically demonstrate the performance advantages of CentSmoothie in simulations as well as real datasets.
Collapse
|
17
|
Waqas A, Tripathi A, Ramachandran RP, Stewart PA, Rasool G. Multimodal data integration for oncology in the era of deep neural networks: a review. Front Artif Intell 2024; 7:1408843. [PMID: 39118787 PMCID: PMC11308435 DOI: 10.3389/frai.2024.1408843] [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: 03/28/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer research encompasses data across various scales, modalities, and resolutions, from screening and diagnostic imaging to digitized histopathology slides to various types of molecular data and clinical records. The integration of these diverse data types for personalized cancer care and predictive modeling holds the promise of enhancing the accuracy and reliability of cancer screening, diagnosis, and treatment. Traditional analytical methods, which often focus on isolated or unimodal information, fall short of capturing the complex and heterogeneous nature of cancer data. The advent of deep neural networks has spurred the development of sophisticated multimodal data fusion techniques capable of extracting and synthesizing information from disparate sources. Among these, Graph Neural Networks (GNNs) and Transformers have emerged as powerful tools for multimodal learning, demonstrating significant success. This review presents the foundational principles of multimodal learning including oncology data modalities, taxonomy of multimodal learning, and fusion strategies. We delve into the recent advancements in GNNs and Transformers for the fusion of multimodal data in oncology, spotlighting key studies and their pivotal findings. We discuss the unique challenges of multimodal learning, such as data heterogeneity and integration complexities, alongside the opportunities it presents for a more nuanced and comprehensive understanding of cancer. Finally, we present some of the latest comprehensive multimodal pan-cancer data sources. By surveying the landscape of multimodal data integration in oncology, our goal is to underline the transformative potential of multimodal GNNs and Transformers. Through technological advancements and the methodological innovations presented in this review, we aim to chart a course for future research in this promising field. This review may be the first that highlights the current state of multimodal modeling applications in cancer using GNNs and transformers, presents comprehensive multimodal oncology data sources, and sets the stage for multimodal evolution, encouraging further exploration and development in personalized cancer care.
Collapse
Affiliation(s)
- Asim Waqas
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, United States
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, United States
| | - Aakash Tripathi
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, United States
| | - Ravi P. Ramachandran
- Department of Electrical and Computer Engineering, Rowan University, Glassboro, NJ, United States
| | - Paul A. Stewart
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, United States
| | - Ghulam Rasool
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
18
|
Zhang D, Wang Z, Zhao D, Li J. DRGATAN: Directed relation graph attention aware network for asymmetric drug-drug interaction prediction. iScience 2024; 27:109943. [PMID: 38868194 PMCID: PMC11167430 DOI: 10.1016/j.isci.2024.109943] [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: 12/26/2023] [Revised: 03/21/2024] [Accepted: 05/06/2024] [Indexed: 06/14/2024] Open
Abstract
In scenarios involving the treatment of complex or coexisting diseases with multiple drugs, the potential for severe adverse drug reactions in patients necessitates the identification of potential drug-drug interactions (DDIs). Most existing computational methods have not taken into account the asymmetry and relation types of drug interactions caused by the relation information between drugs, which may lead to missing information in embedded learning. Therefore, this paper proposes a directed relation graph attention aware network (DRGATAN) to predict asymmetric drug interactions. DRGATAN leverages an encoder to learn multi-relational role embeddings of drugs across different types of relations. The experimental results show that DRGATAN's performance is superior to recognized advanced methods. The visualization demonstrates the effect of utilizing asymmetric information, and the case analysis validates the reliability of the proposed method. This study provides guidance for predicting asymmetric drug interactions.
Collapse
Affiliation(s)
- Dehai Zhang
- The Key Laboratory of Software Engineering of Yunnan Province, School of Software, Yunnan University, Kunming 650091, P.R. China
| | - Zhengwu Wang
- The Key Laboratory of Software Engineering of Yunnan Province, School of Software, Yunnan University, Kunming 650091, P.R. China
| | - Di Zhao
- The Key Laboratory of Software Engineering of Yunnan Province, School of Software, Yunnan University, Kunming 650091, P.R. China
| | - Jin Li
- The Key Laboratory of Software Engineering of Yunnan Province, School of Software, Yunnan University, Kunming 650091, P.R. China
| |
Collapse
|
19
|
He SH, Yun L, Yi HC. Accurate prediction of drug combination risk levels based on relational graph convolutional network and multi-head attention. J Transl Med 2024; 22:572. [PMID: 38880914 PMCID: PMC11180398 DOI: 10.1186/s12967-024-05372-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/02/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Accurately identifying the risk level of drug combinations is of great significance in investigating the mechanisms of combination medication and adverse reactions. Most existing methods can only predict whether there is an interaction between two drugs, but cannot directly determine their accurate risk level. METHODS In this study, we propose a multi-class drug combination risk prediction model named AERGCN-DDI, utilizing a relational graph convolutional network with a multi-head attention mechanism. Drug-drug interaction events with varying risk levels are modeled as a heterogeneous information graph. Attribute features of drug nodes and links are learned based on compound chemical structure information. Finally, the AERGCN-DDI model is proposed to predict drug combination risk level based on heterogenous graph neural network and multi-head attention modules. RESULTS To evaluate the effectiveness of the proposed method, five-fold cross-validation and ablation study were conducted. Furthermore, we compared its predictive performance with baseline models and other state-of-the-art methods on two benchmark datasets. Empirical studies demonstrated the superior performances of AERGCN-DDI. CONCLUSIONS AERGCN-DDI emerges as a valuable tool for predicting the risk levels of drug combinations, thereby aiding in clinical medication decision-making, mitigating severe drug side effects, and enhancing patient clinical prognosis.
Collapse
Affiliation(s)
- Shi-Hui He
- School of Information Science and Technology, Yunnan Normal University, Kunming, 650500, China
- Engineering Research Center of Computer Vision and Intelligent Control Technology, Department of Education, Kunming, 650500, China
| | - Lijun Yun
- School of Information Science and Technology, Yunnan Normal University, Kunming, 650500, China.
- Engineering Research Center of Computer Vision and Intelligent Control Technology, Department of Education, Kunming, 650500, China.
| | - Hai-Cheng Yi
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710129, China.
| |
Collapse
|
20
|
Yao R, Shen Z, Xu X, Ling G, Xiang R, Song T, Zhai F, Zhai Y. Knowledge mapping of graph neural networks for drug discovery: a bibliometric and visualized analysis. Front Pharmacol 2024; 15:1393415. [PMID: 38799167 PMCID: PMC11116974 DOI: 10.3389/fphar.2024.1393415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/12/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction In recent years, graph neural network has been extensively applied to drug discovery research. Although researchers have made significant progress in this field, there is less research on bibliometrics. The purpose of this study is to conduct a comprehensive bibliometric analysis of graph neural network applications in drug discovery in order to identify current research hotspots and trends, as well as serve as a reference for future research. Methods Publications from 2017 to 2023 about the application of graph neural network in drug discovery were collected from the Web of Science Core Collection. Bibliometrix, VOSviewer, and Citespace were mainly used for bibliometric studies. Results and Discussion In this paper, a total of 652 papers from 48 countries/regions were included. Research interest in this field is continuously increasing. China and the United States have a significant advantage in terms of funding, the number of publications, and collaborations with other institutions and countries. Although some cooperation networks have been formed in this field, extensive worldwide cooperation still needs to be strengthened. The results of the keyword analysis clarified that graph neural network has primarily been applied to drug-target interaction, drug repurposing, and drug-drug interaction, while graph convolutional neural network and its related optimization methods are currently the core algorithms in this field. Data availability and ethical supervision, balancing computing resources, and developing novel graph neural network models with better interpretability are the key technical issues currently faced. This paper analyzes the current state, hot spots, and trends of graph neural network applications in drug discovery through bibliometric approaches, as well as the current issues and challenges in this field. These findings provide researchers with valuable insights on the current status and future directions of this field.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fei Zhai
- Faculty of Medical Device, Shenyang Pharmaceutical University, Shenyang, China
| | - Yuxuan Zhai
- Faculty of Medical Device, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
21
|
Zhang Y, Deng Z, Xu X, Feng Y, Junliang S. Application of Artificial Intelligence in Drug-Drug Interactions Prediction: A Review. J Chem Inf Model 2024; 64:2158-2173. [PMID: 37458400 DOI: 10.1021/acs.jcim.3c00582] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Drug-drug interactions (DDI) are a critical aspect of drug research that can have adverse effects on patients and can lead to serious consequences. Predicting these events accurately can significantly improve clinicians' ability to make better decisions and establish optimal treatment regimens. However, manually detecting these interactions is time-consuming and labor-intensive. Utilizing the advancements in Artificial Intelligence (AI) is essential for achieving accurate forecasts of DDIs. In this review, DDI prediction tasks are classified into three types according to the type of DDI prediction: undirected DDI prediction, DDI events prediction, and Asymmetric DDI prediction. The paper then reviews the progress of AI for each of these three prediction tasks in DDI and provides a summary of the data sets used as well as the representative methods used in these three prediction directions. In this review, we aim to provide a comprehensive overview of drug interaction prediction. The first section introduces commonly used databases and presents an overview of current research advancements and techniques across three domains of DDI. Additionally, we introduce classical machine learning techniques for predicting undirected drug interactions and provide a timeline for the progression of the predicted drug interaction events. At last, we debate the difficulties and prospects of AI approaches at predicting DDI, emphasizing their potential for improving clinical decision-making and patient outcomes.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao,266000,China
| | - Zengqian Deng
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao,266000,China
| | - Xiaoyu Xu
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao,266000,China
| | - Yinfei Feng
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao,266000,China
| | - Shang Junliang
- School of Information Science and Engineering, Qufu Normal University, Rizhao, 276800, China
| |
Collapse
|
22
|
Luo H, Yin W, Wang J, Zhang G, Liang W, Luo J, Yan C. Drug-drug interactions prediction based on deep learning and knowledge graph: A review. iScience 2024; 27:109148. [PMID: 38405609 PMCID: PMC10884936 DOI: 10.1016/j.isci.2024.109148] [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] [Indexed: 02/27/2024] Open
Abstract
Drug-drug interactions (DDIs) can produce unpredictable pharmacological effects and lead to adverse events that have the potential to cause irreversible damage to the organism. Traditional methods to detect DDIs through biological or pharmacological analysis are time-consuming and expensive, therefore, there is an urgent need to develop computational methods to effectively predict drug-drug interactions. Currently, deep learning and knowledge graph techniques which can effectively extract features of entities have been widely utilized to develop DDI prediction methods. In this research, we aim to systematically review DDI prediction researches applying deep learning and graph knowledge. The available biomedical data and public databases related to drugs are firstly summarized in this review. Then, we discuss the existing drug-drug interactions prediction methods which have utilized deep learning and knowledge graph techniques and group them into three main classes: deep learning-based methods, knowledge graph-based methods, and methods that combine deep learning with knowledge graph. We comprehensively analyze the commonly used drug related data and various DDI prediction methods, and compare these prediction methods on benchmark datasets. Finally, we briefly discuss the challenges related to drug-drug interactions prediction, including asymmetric DDIs prediction and high-order DDI prediction.
Collapse
Affiliation(s)
- Huimin Luo
- School of Computer and Information Engineering, Henan University, Kaifeng, China
- Henan Key Laboratory of Big Data Analysis and Processing, Henan University, Kaifeng, China
| | - Weijie Yin
- School of Computer and Information Engineering, Henan University, Kaifeng, China
| | - Jianlin Wang
- School of Computer and Information Engineering, Henan University, Kaifeng, China
- Academy for Advanced Interdisciplinary Studies, Zhengzhou, China
| | - Ge Zhang
- School of Computer and Information Engineering, Henan University, Kaifeng, China
- Henan Key Laboratory of Big Data Analysis and Processing, Henan University, Kaifeng, China
| | - Wenjuan Liang
- School of Computer and Information Engineering, Henan University, Kaifeng, China
| | - Junwei Luo
- College of Computer Science and Technology, Henan Polytechnic University, Jiaozuo, China
| | - Chaokun Yan
- School of Computer and Information Engineering, Henan University, Kaifeng, China
- Academy for Advanced Interdisciplinary Studies, Zhengzhou, China
| |
Collapse
|
23
|
Yan X, Gu C, Feng Y, Han J. Predicting Drug-drug Interaction with Graph Mutual Interaction Attention Mechanism. Methods 2024; 223:16-25. [PMID: 38262485 DOI: 10.1016/j.ymeth.2024.01.009] [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: 10/26/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 01/25/2024] Open
Abstract
Effective representation of molecules is a crucial step in AI-driven drug design and drug discovery, especially for drug-drug interaction (DDIs) prediction. Previous work usually models the drug information from the drug-related knowledge graph or the single drug molecules, but the interaction information between molecular substructures of drug pair is seldom considered, thus often ignoring the influence of bond information on atom node representation, leading to insufficient drug representation. Moreover, key molecular substructures have significant contribution to the DDIs prediction results. Therefore, in this work, we propose a novel Graph learning framework of Mutual Interaction Attention mechanism (called GMIA) to predict DDIs by effectively representing the drug molecules. Specifically, we build the node-edge message communication encoder to aggregate atom node and the incoming edge information for atom node representation and design the mutual interaction attention decoder to capture the mutual interaction context between molecular graphs of drug pairs. GMIA can bridge the gap between two encoders for the single drug molecules by attention mechanism. We also design a co-attention matrix to analyze the significance of different-size substructures obtained from the encoder-decoder layer and provide interpretability. In comparison with other recent state-of-the-art methods, our GMIA achieves the best results in terms of area under the precision-recall-curve (AUPR), area under the ROC curve (AUC), and F1 score on two different scale datasets. The case study indicates that our GMIA can detect the key substructure for potential DDIs, demonstrating the enhanced performance and interpretation ability of GMIA.
Collapse
Affiliation(s)
- Xiaoying Yan
- College of Computer Science, Xi'an Shiyou University, Xi'an 710065, China.
| | - Chi Gu
- College of Computer Science, Xi'an Shiyou University, Xi'an 710065, China
| | - Yuehua Feng
- College of Computer Science, Xi'an Shiyou University, Xi'an 710065, China
| | - Jiaxin Han
- College of Computer Science, Xi'an Shiyou University, Xi'an 710065, China
| |
Collapse
|
24
|
Asfand-E-Yar M, Hashir Q, Shah AA, Malik HAM, Alourani A, Khalil W. Multimodal CNN-DDI: using multimodal CNN for drug to drug interaction associated events. Sci Rep 2024; 14:4076. [PMID: 38374325 PMCID: PMC10876630 DOI: 10.1038/s41598-024-54409-x] [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] [Accepted: 02/12/2024] [Indexed: 02/21/2024] Open
Abstract
Drug-to-drug interaction (DDIs) occurs when a patient consumes multiple drugs. Therefore, it is possible that any medication can influence other drugs' effectiveness. The drug-to-drug interactions are detected based on the interactions of chemical substructures, targets, pathways, and enzymes; therefore, machine learning (ML) and deep learning (DL) techniques are used to find the associated DDI events. The DL model, i.e., Convolutional Neural Network (CNN), is used to analyze the DDI. DDI is based on the 65 different drug-associated events, which is present in the drug bank database. Our model uses the inputs, which are chemical structures (i.e., smiles of drugs), enzymes, pathways, and the target of the drug. Therefore, for the multi-model CNN, we use several layers, activation functions, and features of drugs to achieve better accuracy as compared to traditional prediction algorithms. We perform different experiments on various hyperparameters. We have also carried out experiments on various iterations of drug features in different sets. Our Multi-Modal Convolutional Neural Network - Drug to Drug Interaction (MCNN-DDI) model achieved an accuracy of 90.00% and an AUPR of 94.78%. The results showed that a combination of the drug's features (i.e., chemical substructure, target, and enzyme) performs better in DDIs-associated events prediction than other features.
Collapse
Affiliation(s)
- Muhammad Asfand-E-Yar
- Department of Computer Science, CoE-AI, Center of Excellence Artificial Intelligence, Bahria University, Islamabad, Pakistan
| | - Qadeer Hashir
- Department of Computer Science, CoE-AI, Center of Excellence Artificial Intelligence, Bahria University, Islamabad, Pakistan
| | - Asghar Ali Shah
- Department of Computer Science, Bahria University, Islamabad , Pakistan
| | | | - Abdullah Alourani
- Department of Management Information Systems and Production Management, College of Business and Economics, Qassim University, Buraydah 51452, Saudi Arabia.
| | - Waqar Khalil
- Department of Computer Science, CoE-AI, Center of Excellence Artificial Intelligence, Bahria University, Islamabad, Pakistan
| |
Collapse
|
25
|
Pan D, Lu P, Wu Y, Kang L, Huang F, Lin K, Yang F. Prediction of multiple types of drug interactions based on multi-scale fusion and dual-view fusion. Front Pharmacol 2024; 15:1354540. [PMID: 38434701 PMCID: PMC10904638 DOI: 10.3389/fphar.2024.1354540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/30/2024] [Indexed: 03/05/2024] Open
Abstract
Potential drug-drug interactions (DDI) can lead to adverse drug reactions (ADR), and DDI prediction can help pharmacy researchers detect harmful DDI early. However, existing DDI prediction methods fall short in fully capturing drug information. They typically employ a single-view input, focusing solely on drug features or drug networks. Moreover, they rely exclusively on the final model layer for predictions, overlooking the nuanced information present across various network layers. To address these limitations, we propose a multi-scale dual-view fusion (MSDF) method for DDI prediction. More specifically, MSDF first constructs two views, topological and feature views of drugs, as model inputs. Then a graph convolutional neural network is used to extract the feature representations from each view. On top of that, a multi-scale fusion module integrates information across different graph convolutional layers to create comprehensive drug embeddings. The embeddings from the two views are summed as the final representation for classification. Experiments on two real-world datasets demonstrate that MSDF achieves higher accuracy than state-of-the-art methods, as the dual-view, multi-scale approach better captures drug characteristics.
Collapse
Affiliation(s)
- Dawei Pan
- School of Computer and Information Engineering, Xiamen University of Technology, Xiamen, China
| | - Ping Lu
- School of Economics and Management, Xiamen University of Technology, Xiamen, China
| | - Yunbing Wu
- College of Computer and Big Data, Fuzhou University, Fuzhou, China
| | - Liping Kang
- Pasteur Institute, Soochow University, Suzhou, China
| | - Fengxin Huang
- School of Computer and Information Engineering, Xiamen University of Technology, Xiamen, China
| | - Kaibiao Lin
- School of Computer and Information Engineering, Xiamen University of Technology, Xiamen, China
| | - Fan Yang
- Shenzhen Research Institute of Xiamen University, Shenzhen, China
- Department of Automation, Xiamen University, Xiamen, China
| |
Collapse
|
26
|
Jin Q, Xie J, Huang D, Zhao C, He H. MSFF-MA-DDI: Multi-Source Feature Fusion with Multiple Attention blocks for predicting Drug-Drug Interaction events. Comput Biol Chem 2024; 108:108001. [PMID: 38154317 DOI: 10.1016/j.compbiolchem.2023.108001] [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: 07/23/2023] [Revised: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 12/30/2023]
Abstract
The interaction of multiple drugs could lead to severe events, which cause medical injuries and expenses. Accurate prediction of drug-drug interaction (DDI) events can help clinicians make effective decisions and establish appropriate therapy programs. However, there exist two issues worthy of further consideration. (i) The global features of drug molecules should be paid attention to, rather than just their local characteristics. (ii) The fusion of multi-source features should also be studied to capture the comprehensive features of the drug. This study designs a Multi-Source Feature Fusion framework with Multiple Attention blocks named MSFF-MA-DDI that utilizes multimodal data for DDI event prediction. MSFF-MA-DDI can (i) encode global correlations between long-distance atoms in drug molecular sequences by a self-attention layer based on a position embedding block and (ii) fuse drug sequence features and heterogeneous features (chemical substructure, target, and enzyme) through a multi-head attention block to better represent the features of drugs. Experiments on real-world datasets show that MSFF-MA-DDI can achieve performance that is close to or even better than state-of-the-art models. Especially in cold start scenarios, the model can achieve the best performance. The effectiveness of the model is also supported by the case study on nervous system drugs. The source codes and data are available at https://github.com/BioCenter-SHU/MSFF-MA-DDI.
Collapse
Affiliation(s)
- Qi Jin
- School of Computer Engineering and Science, Shanghai University, Shanghai, 200444, China
| | - Jiang Xie
- School of Computer Engineering and Science, Shanghai University, Shanghai, 200444, China.
| | - Dingkai Huang
- School of Computer Engineering and Science, Shanghai University, Shanghai, 200444, China
| | - Chang Zhao
- School of Computer Engineering and Science, Shanghai University, Shanghai, 200444, China
| | - Hongjian He
- School of Computer Engineering and Science, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
27
|
Chen S, Semenov I, Zhang F, Yang Y, Geng J, Feng X, Meng Q, Lei K. An effective framework for predicting drug-drug interactions based on molecular substructures and knowledge graph neural network. Comput Biol Med 2024; 169:107900. [PMID: 38199213 DOI: 10.1016/j.compbiomed.2023.107900] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/27/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024]
Abstract
Drug-drug interactions (DDIs) play a central role in drug research, as the simultaneous administration of multiple drugs can have harmful or beneficial effects. Harmful interactions lead to adverse reactions, some of which can be life-threatening, while beneficial interactions can promote efficacy. Therefore, it is crucial for physicians, patients, and the research community to identify potential DDIs. Although many AI-based techniques have been proposed for predicting DDIs, most existing computational models primarily focus on integrating multiple data sources or combining popular embedding methods. Researchers often overlook the valuable information within the molecular structure of drugs or only consider the structural information of drugs, neglecting the relationship or topological information between drugs and other biological objects. In this study, we propose MSKG-DDI - a two-component framework that incorporates the Drug Chemical Structure Graph-based component and the Drug Knowledge Graph-based component to capture multimodal characteristics of drugs. Subsequently, a multimodal fusion neural layer is utilized to explore the complementarity between multimodal representations of drugs. Extensive experiments were conducted using two real-world datasets, and the results demonstrate that MSKG-DDI outperforms other state-of-the-art models in binary-class, multi-class, and multi-label prediction tasks under both transductive and inductive settings. Furthermore, the ablation analysis further confirms the practical usefulness of MSKG-DDI.
Collapse
Affiliation(s)
- Siqi Chen
- School of Information Science and Engineering, Chongqing Jiaotong University, Chongqing, 400074, China
| | - Ivan Semenov
- College of Intelligence and Computing, Tianjin University, Tianjin, 300072, China
| | - Fengyun Zhang
- College of Intelligence and Computing, Tianjin University, Tianjin, 300072, China
| | - Yang Yang
- College of Intelligence and Computing, Tianjin University, Tianjin, 300072, China
| | - Jie Geng
- TianJin Chest Hospital, Tianjin University, Tianjin, 300222, China
| | - Xuequan Feng
- Tianjin First Central Hospital, Tianjin, 300192, China.
| | - Qinghua Meng
- Tianjin Key Laboratory of Sports Physiology and Sports Medicine, Tianjin University of Sport, Tianjin, 301617, China
| | - Kaiyou Lei
- College of Computer and Information Science, Southwest University, Chongqing, 400715, China
| |
Collapse
|
28
|
Kpanou R, Dallaire P, Rousseau E, Corbeil J. Learning self-supervised molecular representations for drug-drug interaction prediction. BMC Bioinformatics 2024; 25:47. [PMID: 38291362 PMCID: PMC10829170 DOI: 10.1186/s12859-024-05643-7] [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: 09/20/2023] [Accepted: 01/05/2024] [Indexed: 02/01/2024] Open
Abstract
Drug-drug interactions (DDI) are a critical concern in healthcare due to their potential to cause adverse effects and compromise patient safety. Supervised machine learning models for DDI prediction need to be optimized to learn abstract, transferable features, and generalize to larger chemical spaces, primarily due to the scarcity of high-quality labeled DDI data. Inspired by recent advances in computer vision, we present SMR-DDI, a self-supervised framework that leverages contrastive learning to embed drugs into a scaffold-based feature space. Molecular scaffolds represent the core structural motifs that drive pharmacological activities, making them valuable for learning informative representations. Specifically, we pre-trained SMR-DDI on a large-scale unlabeled molecular dataset. We generated augmented views for each molecule via SMILES enumeration and optimized the embedding process through contrastive loss minimization between views. This enables the model to capture relevant and robust molecular features while reducing noise. We then transfer the learned representations for the downstream prediction of DDI. Experiments show that the new feature space has comparable expressivity to state-of-the-art molecular representations and achieved competitive DDI prediction results while training on less data. Additional investigations also revealed that pre-training on more extensive and diverse unlabeled molecular datasets improved the model's capability to embed molecules more effectively. Our results highlight contrastive learning as a promising approach for DDI prediction that can identify potentially hazardous drug combinations using only structural information.
Collapse
Affiliation(s)
- Rogia Kpanou
- Département d'informatique et Génie Logiciel, Université Laval, Québec City, QC, Canada.
| | - Patrick Dallaire
- Département d'informatique et Génie Logiciel, Université Laval, Québec City, QC, Canada
| | - Elsa Rousseau
- Département d'informatique et Génie Logiciel, Université Laval, Québec City, QC, Canada
- Centre de Recherche en Données Massives de l'Université Laval, Québec City, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec City, QC, Canada
| | - Jacques Corbeil
- Centre de Recherche en Données Massives de l'Université Laval, Québec City, QC, Canada.
- Centre de Recherche en Infectiologie de l'Université Laval, Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.
- Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec City, QC, Canada.
| |
Collapse
|
29
|
Zhou B, Ran B, Chen L. A GraphSAGE-based model with fingerprints only to predict drug-drug interactions. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:2922-2942. [PMID: 38454713 DOI: 10.3934/mbe.2024130] [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: 03/09/2024]
Abstract
Drugs are an effective way to treat various diseases. Some diseases are so complicated that the effect of a single drug for such diseases is limited, which has led to the emergence of combination drug therapy. The use multiple drugs to treat these diseases can improve the drug efficacy, but it can also bring adverse effects. Thus, it is essential to determine drug-drug interactions (DDIs). Recently, deep learning algorithms have become popular to design DDI prediction models. However, most deep learning-based models need several types of drug properties, inducing the application problems for drugs without these properties. In this study, a new deep learning-based model was designed to predict DDIs. For wide applications, drugs were first represented by commonly used properties, referred to as fingerprint features. Then, these features were perfectly fused with the drug interaction network by a type of graph convolutional network method, GraphSAGE, yielding high-level drug features. The inner product was adopted to score the strength of drug pairs. The model was evaluated by 10-fold cross-validation, resulting in an AUROC of 0.9704 and AUPR of 0.9727. Such performance was better than the previous model which directly used drug fingerprint features and was competitive compared with some other previous models that used more drug properties. Furthermore, the ablation tests indicated the importance of the main parts of the model, and we analyzed the strengths and limitations of a model for drugs with different degrees in the network. This model identified some novel DDIs that may bring expected benefits, such as the combination of PEA and cannabinol that may produce better effects. DDIs that may cause unexpected side effects have also been discovered, such as the combined use of WIN 55,212-2 and cannabinol. These DDIs can provide novel insights for treating complex diseases or avoiding adverse drug events.
Collapse
Affiliation(s)
- Bo Zhou
- Institute of Wound Prevention and Treatment, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
- School of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Bing Ran
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| |
Collapse
|
30
|
Wang NN, Zhu B, Li XL, Liu S, Shi JY, Cao DS. Comprehensive Review of Drug-Drug Interaction Prediction Based on Machine Learning: Current Status, Challenges, and Opportunities. J Chem Inf Model 2024; 64:96-109. [PMID: 38132638 DOI: 10.1021/acs.jcim.3c01304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Detecting drug-drug interactions (DDIs) is an essential step in drug development and drug administration. Given the shortcomings of current experimental methods, the machine learning (ML) approach has become a reliable alternative, attracting extensive attention from the academic and industrial fields. With the rapid development of computational science and the growing popularity of cross-disciplinary research, a large number of DDI prediction studies based on ML methods have been published in recent years. To give an insight into the current situation and future direction of DDI prediction research, we systemically review these studies from three aspects: (1) the classic DDI databases, mainly including databases of drugs, side effects, and DDI information; (2) commonly used drug attributes, which focus on chemical, biological, and phenotypic attributes for representing drugs; (3) popular ML approaches, such as shallow learning-based, deep learning-based, recommender system-based, and knowledge graph-based methods for DDI detection. For each section, related studies are described, summarized, and compared, respectively. In the end, we conclude the research status of DDI prediction based on ML methods and point out the existing issues, future challenges, potential opportunities, and subsequent research direction.
Collapse
Affiliation(s)
- Ning-Ning Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
| | - Bei Zhu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, P.R. China
| | - Xin-Liang Li
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
| | - Shao Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
| | - Jian-Yu Shi
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, P.R. China
| | - Dong-Sheng Cao
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P.R. China
| |
Collapse
|
31
|
Yang X, Huang K, Yang D, Zhao W, Zhou X. Biomedical Big Data Technologies, Applications, and Challenges for Precision Medicine: A Review. GLOBAL CHALLENGES (HOBOKEN, NJ) 2024; 8:2300163. [PMID: 38223896 PMCID: PMC10784210 DOI: 10.1002/gch2.202300163] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/20/2023] [Indexed: 01/16/2024]
Abstract
The explosive growth of biomedical Big Data presents both significant opportunities and challenges in the realm of knowledge discovery and translational applications within precision medicine. Efficient management, analysis, and interpretation of big data can pave the way for groundbreaking advancements in precision medicine. However, the unprecedented strides in the automated collection of large-scale molecular and clinical data have also introduced formidable challenges in terms of data analysis and interpretation, necessitating the development of novel computational approaches. Some potential challenges include the curse of dimensionality, data heterogeneity, missing data, class imbalance, and scalability issues. This overview article focuses on the recent progress and breakthroughs in the application of big data within precision medicine. Key aspects are summarized, including content, data sources, technologies, tools, challenges, and existing gaps. Nine fields-Datawarehouse and data management, electronic medical record, biomedical imaging informatics, Artificial intelligence-aided surgical design and surgery optimization, omics data, health monitoring data, knowledge graph, public health informatics, and security and privacy-are discussed.
Collapse
Affiliation(s)
- Xue Yang
- Department of Pancreatic Surgery and West China Biomedical Big Data CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Kexin Huang
- Department of Pancreatic Surgery and West China Biomedical Big Data CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Dewei Yang
- College of Advanced Manufacturing EngineeringChongqing University of Posts and TelecommunicationsChongqingChongqing400000China
| | - Weiling Zhao
- Center for Systems MedicineSchool of Biomedical InformaticsUTHealth at HoustonHoustonTX77030USA
| | - Xiaobo Zhou
- Center for Systems MedicineSchool of Biomedical InformaticsUTHealth at HoustonHoustonTX77030USA
| |
Collapse
|
32
|
Lin S, Mao X, Hong L, Lin S, Wei DQ, Xiong Y. MATT-DDI: Predicting multi-type drug-drug interactions via heterogeneous attention mechanisms. Methods 2023; 220:1-10. [PMID: 37858611 DOI: 10.1016/j.ymeth.2023.10.007] [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: 09/22/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 10/21/2023] Open
Abstract
The joint use of multiple drugs can result in adverse drug-drug interactions (DDIs) and side effects that harm the body. Accurate identification of DDIs is crucial for avoiding accidental drug side effects and understanding potential mechanisms underlying DDIs. Several computational methods have been proposed for multi-type DDI prediction, but most rely on the similarity profiles of drugs as the drug feature vectors, which may result in information leakage and overoptimistic performance when predicting interactions between new drugs. To address this issue, we propose a novel method, MATT-DDI, for predicting multi-type DDIs based on the original feature vectors of drugs and multiple attention mechanisms. MATT-DDI consists of three main modules: the top k most similar drug pair selection module, heterogeneous attention mechanism module and multi‑type DDI prediction module. Firstly, based on the feature vector of the input drug pair (IDP), k drug pairs that are most similar to the input drug pair from the training dataset are selected according to cosine similarity between drug pairs. Then, the vectors of k selected drug pairs are averaged to obtain a new drug pair (NDP). Next, IDP and NDP are fed into heterogeneous attention modules, including scaled dot product attention and bilinear attention, to extract latent feature vectors. Finally, these latent feature vectors are taken as input of the classification module to predict DDI types. We evaluated MATT-DDI on three different tasks. The experimental results show that MATT-DDI provides better or comparable performance compared to several state-of-the-art methods, and its feasibility is supported by case studies. MATT-DDI is a robust model for predicting multi-type DDIs with excellent performance and no information leakage.
Collapse
Affiliation(s)
- Shenggeng Lin
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xueying Mao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Liang Hong
- Shanghai Artificial Intelligence Laboratory, Shanghai 200232, China; School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shuangjun Lin
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; Zhongjing Research and Industrialization Institute of Chinese Medicine, Nanyang 473006, China; Peng Cheng National Laboratory, Shenzhen 518055, China
| | - Yi Xiong
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; Shanghai Artificial Intelligence Laboratory, Shanghai 200232, China.
| |
Collapse
|
33
|
Feng J, Liang Y, Yu T. MM-GANN-DDI: Multimodal Graph-Agnostic Neural Networks for Predicting Drug-Drug Interaction Events. Comput Biol Med 2023; 166:107492. [PMID: 37820558 DOI: 10.1016/j.compbiomed.2023.107492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/15/2023] [Accepted: 09/15/2023] [Indexed: 10/13/2023]
Abstract
Personalized treatment of complex diseases relies on combined medication. However, the occurrence of unexpected drug-drug interactions (DDIs) in these combinations can lead to adverse effects or even fatalities. Although recent computational methods exhibit promising performance in DDI screening, their practical implementation faces two significant challenges: (i) the availability of comprehensive datasets to support clinical application, and (ii) the ability to infer DDI types for new drugs beyond the existing dataset coverage. To mitigate these challenges, we propose MM-GANN-DDI: a Multimodal Graph-Agnostic Neural Network for Predicting Drug-Drug Interaction Events. We first mine six drug modalities and incorporate a graph attention (GAT) mechanism to fuse these modalities with the topological features of the DDI graph. We further propose a novel graph neural network training mechanism called graph-agnostic meta-training (GAMT), which effectively leverages topological information from the DDI graph and efficiently predicts DDI types for new drugs beyond the available dataset. Specifically, GAMT samples meta-graphs from the original DDI graph, splitting them into support and query sets to simulate seen and unseen drugs. Two-level optimizations are applied to enhance the model's generalization capability. We evaluate our model on two datasets (DB-v1 and DB-v2) across three tasks. Our MM-GANN-DDI demonstrates competitive performance on all three tasks. Notably, in Task 2, which focuses on predicting DDI types for drugs outside the dataset, our proposed model outperforms other methods, exhibiting an improvement of 4.6 percentage points in AUPR on DB-v1 and 5.9 percentage points on DB-v2. Additionally, our model surpasses state-of-the-art methods and classic approaches in terms of accuracy, F1 score, precision, and recall. Ablation experiments provide further validation of the effectiveness of the proposed model design. Importantly, our model exhibits the potential to discover unobserved DDIs, demonstrating its practical application in clinical medication.
Collapse
Affiliation(s)
- Junning Feng
- Faculty of Innovation Engineering, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China; School of Data Science, The Chinese University of Hong Kong-Shenzhen, Shenzhen, 518055, China
| | - Yong Liang
- Peng Cheng Laboratory, Shenzhen, 518055, China.
| | - Tianwei Yu
- School of Data Science, The Chinese University of Hong Kong-Shenzhen, Shenzhen, 518055, China
| |
Collapse
|
34
|
Ning G, Sun Y, Ling J, Chen J, He J. BDN-DDI: A bilinear dual-view representation learning framework for drug-drug interaction prediction. Comput Biol Med 2023; 165:107340. [PMID: 37603959 DOI: 10.1016/j.compbiomed.2023.107340] [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: 06/02/2023] [Revised: 07/23/2023] [Accepted: 08/07/2023] [Indexed: 08/23/2023]
Abstract
Drug-drug interactions (DDIs) refer to the potential effects of two or more drugs interacting with each other when used simultaneously, which may lead to adverse reactions or reduced drug efficacy. Accurate prediction of DDIs is a significant concern in recent years. Currently, the drug chemical substructure-based learning method has substantially improved DDIs prediction. However, we notice that most related works ignore the detailed interaction among atoms when extracting the substructure information of drugs. This problem results in incomplete information extraction and may limit the model's predictive ability. In this work, we proposed a novel framework named BDN-DDI (a bilinear dual-view representation learning framework for drug-drug interaction prediction) to infer potential DDIs. In the proposed framework, the encoder consists of six stacked BDN blocks, each of which extracts the feature representation of drug molecules through a bilinear representation extraction layer. The extracted feature is then used to learn embeddings of drug substructures from the single drug learning layer (intra-layer) and the drug-pair learning layer (inter-layer). Finally, the learned embeddings are fed into a decoder to predict DDI events. Based on our experiments, BDN-DDI has an AUROC value of over 99% for the warm-start task. Additionally, it outperformed the state-of-the-art methods by an average of 3.4% for the cold-start tasks. Finally, our method's effectiveness is further validated by visualizing several case studies.
Collapse
Affiliation(s)
- Guoquan Ning
- School of Computer Science and Technology, Guangdong University of Technology, Guangzhou 510006, China
| | - Yuping Sun
- School of Computer Science and Technology, Guangdong University of Technology, Guangzhou 510006, China.
| | - Jie Ling
- School of Computer Science and Technology, Guangdong University of Technology, Guangzhou 510006, China
| | - Jijia Chen
- School of Computer Science and Technology, Guangdong University of Technology, Guangzhou 510006, China
| | - Jiaxi He
- School of Computer Science and Technology, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
35
|
Li Z, Tu X, Chen Y, Lin W. HetDDI: a pre-trained heterogeneous graph neural network model for drug-drug interaction prediction. Brief Bioinform 2023; 24:bbad385. [PMID: 37903412 DOI: 10.1093/bib/bbad385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/12/2023] [Accepted: 09/13/2023] [Indexed: 11/01/2023] Open
Abstract
The simultaneous use of two or more drugs due to multi-disease comorbidity continues to increase, which may cause adverse reactions between drugs that seriously threaten public health. Therefore, the prediction of drug-drug interaction (DDI) has become a hot topic not only in clinics but also in bioinformatics. In this study, we propose a novel pre-trained heterogeneous graph neural network (HGNN) model named HetDDI, which aggregates the structural information in drug molecule graphs and rich semantic information in biomedical knowledge graph to predict DDIs. In HetDDI, we first initialize the parameters of the model with different pre-training methods. Then we apply the pre-trained HGNN to learn the feature representation of drugs from multi-source heterogeneous information, which can more effectively utilize drugs' internal structure and abundant external biomedical knowledge, thus leading to better DDI prediction. We evaluate our model on three DDI prediction tasks (binary-class, multi-class and multi-label) with three datasets and further assess its performance on three scenarios (S1, S2 and S3). The results show that the accuracy of HetDDI can achieve 98.82% in the binary-class task, 98.13% in the multi-class task and 96.66% in the multi-label one on S1, which outperforms the state-of-the-art methods by at least 2%. On S2 and S3, our method also achieves exciting performance. Furthermore, the case studies confirm that our model performs well in predicting unknown DDIs. Source codes are available at https://github.com/LinsLab/HetDDI.
Collapse
Affiliation(s)
- Zhe Li
- School of Computer Science, University of South China, Hengyang, 421001 Hunan, China
| | - Xinyi Tu
- School of Computer Science, University of South China, Hengyang, 421001 Hunan, China
| | - Yuping Chen
- School of Pharmacy, University of South China, Hengyang 421001, China
| | - Wenbin Lin
- School of Mathematics and Physics, University of South China, Hengyang 421001, China
| |
Collapse
|
36
|
Gan Y, Liu W, Xu G, Yan C, Zou G. DMFDDI: deep multimodal fusion for drug-drug interaction prediction. Brief Bioinform 2023; 24:bbad397. [PMID: 37930025 DOI: 10.1093/bib/bbad397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 09/28/2023] [Accepted: 10/13/2023] [Indexed: 11/07/2023] Open
Abstract
Drug combination therapy has gradually become a promising treatment strategy for complex or co-existing diseases. As drug-drug interactions (DDIs) may cause unexpected adverse drug reactions, DDI prediction is an important task in pharmacology and clinical applications. Recently, researchers have proposed several deep learning methods to predict DDIs. However, these methods mainly exploit the chemical or biological features of drugs, which is insufficient and limits the performances of DDI prediction. Here, we propose a new deep multimodal feature fusion framework for DDI prediction, DMFDDI, which fuses drug molecular graph, DDI network and the biochemical similarity features of drugs to predict DDIs. To fully extract drug molecular structure, we introduce an attention-gated graph neural network for capturing the global features of the molecular graph and the local features of each atom. A sparse graph convolution network is introduced to learn the topological structure information of the DDI network. In the multimodal feature fusion module, an attention mechanism is used to efficiently fuse different features. To validate the performance of DMFDDI, we compare it with 10 state-of-the-art methods. The comparison results demonstrate that DMFDDI achieves better performance in DDI prediction. Our method DMFDDI is implemented in Python using the Pytorch machine-learning library, and it is freely available at https://github.com/DHUDEBLab/DMFDDI.git.
Collapse
Affiliation(s)
- Yanglan Gan
- School of Computer Science and Technology, Donghua University, 2999 North Renmin Road, 201600, Shanghai, China
| | - Wenxiao Liu
- School of Computer Science and Technology, Donghua University, 2999 North Renmin Road, 201600, Shanghai, China
| | - Guangwei Xu
- School of Computer Science and Technology, Donghua University, 2999 North Renmin Road, 201600, Shanghai, China
| | - Cairong Yan
- School of Computer Science and Technology, Donghua University, 2999 North Renmin Road, 201600, Shanghai, China
| | - Guobing Zou
- School of Computer Engineering and Science, Shanghai University, 99 Shangda Road, 200444, Shanghai, China
| |
Collapse
|
37
|
Zhang R, Wang X, Wang P, Meng Z, Cui W, Zhou Y. HTCL-DDI: a hierarchical triple-view contrastive learning framework for drug-drug interaction prediction. Brief Bioinform 2023; 24:bbad324. [PMID: 37742052 DOI: 10.1093/bib/bbad324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/26/2023] [Accepted: 08/24/2023] [Indexed: 09/25/2023] Open
Abstract
Drug-drug interaction (DDI) prediction can discover potential risks of drug combinations in advance by detecting drug pairs that are likely to interact with each other, sparking an increasing demand for computational methods of DDI prediction. However, existing computational DDI methods mostly rely on the single-view paradigm, failing to handle the complex features and intricate patterns of DDIs due to the limited expressiveness of the single view. To this end, we propose a Hierarchical Triple-view Contrastive Learning framework for Drug-Drug Interaction prediction (HTCL-DDI), leveraging the molecular, structural and semantic views to model the complicated information involved in DDI prediction. To aggregate the intra-molecular compositional and structural information, we present a dual attention-aware network in the molecular view. Based on the molecular view, to further capture inter-molecular information, we utilize the one-hop neighboring information and high-order semantic relations in the structural view and semantic view, respectively. Then, we introduce contrastive learning to enhance drug representation learning from multifaceted aspects and improve the robustness of HTCL-DDI. Finally, we conduct extensive experiments on three real-world datasets. All the experimental results show the significant improvement of HTCL-DDI over the state-of-the-art methods, which also demonstrates that HTCL-DDI opens new avenues for ensuring medication safety and identifying synergistic drug combinations.
Collapse
Affiliation(s)
- Ran Zhang
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuezhi Wang
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Pengfei Wang
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhen Meng
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wenjuan Cui
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuanchun Zhou
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
38
|
Yang E, Zhang H, Zang Z, Zhou Z, Wang S, Liu Z, Liu Y. GCNfold: A novel lightweight model with valid extractors for RNA secondary structure prediction. Comput Biol Med 2023; 164:107246. [PMID: 37487383 DOI: 10.1016/j.compbiomed.2023.107246] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/23/2023] [Accepted: 07/07/2023] [Indexed: 07/26/2023]
Abstract
RNA secondary structure is essential for predicting the tertiary structure and understanding RNA function. Recent research tends to stack numerous modules to design large deep-learning models. This can increase the accuracy to more than 70%, as well as significant training costs and prediction efficiency. We proposed a model with three feature extractors called GCNfold. Structure Extractor utilizes a three-layer Graph Convolutional Network (GCN) to mine the structural information of RNA, such as stems, hairpin, and internal loops. Structure and Sequence Fusion embeds structural information into sequences with Transformer Encoders. Long-distance Dependency Extractor captures long-range pairwise relationships by UNet. The experiments indicate that GCNfold has a small number of parameters, a fast inference speed, and a high accuracy among all models with over 80% accuracy. Additionally, GCNfold-Small takes only 90ms to infer an RNA secondary structure and can achieve close to 90% accuracy on average. The GCNfold code is available on Github https://github.com/EnbinYang/GCNfold.
Collapse
Affiliation(s)
- Enbin Yang
- College of Computer Science and Technology, Jilin University, Changchun, 130012, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, 130012, China
| | - Hao Zhang
- College of Computer Science and Technology, Jilin University, Changchun, 130012, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, 130012, China; College of Software, Jilin University, Changchun, 130012, China
| | - Zinan Zang
- College of Computer Science and Technology, Jilin University, Changchun, 130012, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, 130012, China
| | - Zhiyong Zhou
- College of Computer Science and Technology, Jilin University, Changchun, 130012, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, 130012, China
| | - Shuo Wang
- College of Computer Science and Technology, Jilin University, Changchun, 130012, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, 130012, China
| | - Zhen Liu
- College of Computer Science and Technology, Jilin University, Changchun, 130012, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, 130012, China; Graduate School of Engineering, Nagasaki Institute of Applied Science, 536 Aba-machi, Nagasaki 851-0193, Japan
| | - Yuanning Liu
- College of Computer Science and Technology, Jilin University, Changchun, 130012, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, 130012, China; College of Software, Jilin University, Changchun, 130012, China.
| |
Collapse
|
39
|
Pan L, Xiao X, Liu S, Peng S. An Integration Framework of Secure Multiparty Computation and Deep Neural Network for Improving Drug-Drug Interaction Predictions. J Comput Biol 2023; 30:1034-1045. [PMID: 37707993 DOI: 10.1089/cmb.2023.0076] [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: 09/16/2023] Open
Abstract
Drug-drug interaction (DDI) is a key concern in drug development and pharmacovigilance. It is important to improve DDI predictions by integrating multisource data from various pharmaceutical companies. Unfortunately, the data privacy and financial interest issues seriously influence the interinstitutional collaborations for DDI predictions. We propose multiparty computation DDI (MPCDDI), a secure MPC-based deep learning framework for DDI predictions. MPCDDI leverages the secret sharing technologies to incorporate the drug-related feature data from multiple institutions and develops a deep learning model for DDI predictions. In MPCDDI, all data transmission and deep learning operations are integrated into secure MPC frameworks to enable high-quality collaboration among pharmaceutical institutions without divulging private drug-related information. The results suggest that MPCDDI is superior to other eight baselines and achieves the similar performance to that of the corresponding plaintext collaborations. More interestingly, MPCDDI significantly outperforms methods that use private data from the single institution. In summary, MPCDDI is an effective framework for promoting collaborative and privacy-preserving drug discovery.
Collapse
Affiliation(s)
- Liang Pan
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Xia Xiao
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | | | - Shaoliang Peng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
- The State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| |
Collapse
|
40
|
Masumshah R, Eslahchi C. DPSP: a multimodal deep learning framework for polypharmacy side effects prediction. BIOINFORMATICS ADVANCES 2023; 3:vbad110. [PMID: 37701676 PMCID: PMC10493180 DOI: 10.1093/bioadv/vbad110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/01/2023] [Accepted: 08/15/2023] [Indexed: 09/14/2023]
Abstract
Motivation Because unanticipated drug-drug interactions (DDIs) can result in severe bodily harm, identifying the adverse effects of polypharmacy is one of the most important tasks in human health. Over the past few decades, computational methods for predicting the adverse effects of polypharmacy have been developed. Results This article presents DPSP, a framework for predicting polypharmacy side effects based on the construction of novel drug features and the application of a deep neural network to predict DDIs. In the first step, a variety of drug information is evaluated, and a feature extraction method and the Jaccard similarity are used to determine similarities between two drugs. By combining these similarities, a novel feature vector is generated for each drug. In the second step, the method predicts DDIs for specific DDI events using a multimodal framework and drug feature vectors. On three benchmark datasets, the performance of DPSP is measured by comparing its results to those of several well-known methods, such as GNN-DDI, MSTE, MDF-SA-DDI, NNPS, DDIMDL, DNN, DeepDDI, KNN, LR, and RF. DPSP outperforms these classification methods based on a variety of classification metrics. The results indicate that the use of diverse drug information is effective and efficient for identifying DDI adverse effects. Availability and implementation The source code and datasets are available at https://github.com/raziyehmasumshah/DPSP.
Collapse
Affiliation(s)
- Raziyeh Masumshah
- Department of Computer and Data Sciences, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran 1983969411, Iran
| | - Changiz Eslahchi
- Department of Computer and Data Sciences, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran 1983969411, Iran
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran 193955746, Iran
| |
Collapse
|
41
|
Han CD, Wang CC, Huang L, Chen X. MCFF-MTDDI: multi-channel feature fusion for multi-typed drug-drug interaction prediction. Brief Bioinform 2023; 24:bbad215. [PMID: 37291761 DOI: 10.1093/bib/bbad215] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/11/2023] [Accepted: 05/21/2023] [Indexed: 06/10/2023] Open
Abstract
Adverse drug-drug interactions (DDIs) have become an increasingly serious problem in the medical and health system. Recently, the effective application of deep learning and biomedical knowledge graphs (KGs) have improved the DDI prediction performance of computational models. However, the problems of feature redundancy and KG noise also arise, bringing new challenges for researchers. To overcome these challenges, we proposed a Multi-Channel Feature Fusion model for multi-typed DDI prediction (MCFF-MTDDI). Specifically, we first extracted drug chemical structure features, drug pairs' extra label features, and KG features of drugs. Then, these different features were effectively fused by a multi-channel feature fusion module. Finally, multi-typed DDIs were predicted through the fully connected neural network. To our knowledge, we are the first to integrate the extra label information into KG-based multi-typed DDI prediction; besides, we innovatively proposed a novel KG feature learning method and a State Encoder to obtain target drug pairs' KG-based features which contained more abundant and more key drug-related KG information with less noise; furthermore, a Gated Recurrent Unit-based multi-channel feature fusion module was proposed in an innovative way to yield more comprehensive feature information about drug pairs, effectively alleviating the problem of feature redundancy. We experimented with four datasets in the multi-class and the multi-label prediction tasks to comprehensively evaluate the performance of MCFF-MTDDI for predicting interactions of known-known drugs, known-new drugs and new-new drugs. In addition, we further conducted ablation studies and case studies. All the results fully demonstrated the effectiveness of MCFF-MTDDI.
Collapse
Affiliation(s)
- Chen-Di Han
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou, 221116, China
| | - Chun-Chun Wang
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou, 221116, China
- School of Science, Jiangnan University, Wuxi, 214122, China
| | - Li Huang
- The Future Laboratory, Tsinghua University, Beijing, 100084, China
| | - Xing Chen
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou, 221116, China
- School of Science, Jiangnan University, Wuxi, 214122, China
- Artificial Intelligence Research Institute, China University of Mining and Technology, Xuzhou, 221116, China
| |
Collapse
|
42
|
Deng H, Li Q, Liu Y, Zhu J. MTMG: A multi-task model with multi-granularity information for drug-drug interaction extraction. Heliyon 2023; 9:e16819. [PMID: 37484258 PMCID: PMC10360954 DOI: 10.1016/j.heliyon.2023.e16819] [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: 05/08/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 07/25/2023] Open
Abstract
Drug-drug interactions (DDIs) extraction includes identifying drug entities and interactions between drug pairs from the biomedical corpus. The discovery of potential DDIs aids in our understanding of the mechanisms underlying adverse reactions or combination therapy to improve patient safety. The manual extraction of DDIs is very time-consuming and expensive; therefore, computer-aided extraction of DDIs is vital. Many neural network-based methods have been proposed and achieved good efficiency in the extraction of DDIs over the years. However, most studies improved the performance of DDIs extraction with various external drug features while directly using golden drug entities, leading to error propagation and low universality in practical application. In this paper, we propose a new multi-task framework called MTMG, which changes DDIs extraction from a sentence-level classification task to a sequence labeling task named Drug-Specified Token Classification (DSTC). The proposed approach, MTMG, jointly trains DSTC with drug named entity recognition (DNER) and two sentence-level auxiliary tasks we designed. We aim to improve the performance of the entire DDIs extraction pipeline by better using the correlation between entities and relationships and, to the extent possible, using the information of varying granularity implied in the dataset. Experimental results show that MTMG can both improve the accuracy of DNER and DDIs extraction and outperforms state-of-the-art technique.
Collapse
|
43
|
Zhang M, Gao H, Liao X, Ning B, Gu H, Yu B. DBGRU-SE: predicting drug-drug interactions based on double BiGRU and squeeze-and-excitation attention mechanism. Brief Bioinform 2023:7176312. [PMID: 37225428 DOI: 10.1093/bib/bbad184] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/03/2023] [Accepted: 04/23/2023] [Indexed: 05/26/2023] Open
Abstract
The prediction of drug-drug interactions (DDIs) is essential for the development and repositioning of new drugs. Meanwhile, they play a vital role in the fields of biopharmaceuticals, disease diagnosis and pharmacological treatment. This article proposes a new method called DBGRU-SE for predicting DDIs. Firstly, FP3 fingerprints, MACCS fingerprints, Pubchem fingerprints and 1D and 2D molecular descriptors are used to extract the feature information of the drugs. Secondly, Group Lasso is used to remove redundant features. Then, SMOTE-ENN is applied to balance the data to obtain the best feature vectors. Finally, the best feature vectors are fed into the classifier combining BiGRU and squeeze-and-excitation (SE) attention mechanisms to predict DDIs. After applying five-fold cross-validation, The ACC values of DBGRU-SE model on the two datasets are 97.51 and 94.98%, and the AUC are 99.60 and 98.85%, respectively. The results showed that DBGRU-SE had good predictive performance for drug-drug interactions.
Collapse
Affiliation(s)
| | - Hongli Gao
- Qingdao University of Science and Technology, China
| | - Xin Liao
- Qingdao University of Science and Technology, China
| | - Baoxing Ning
- Qingdao University of Science and Technology, China
| | - Haiming Gu
- Qingdao University of Science and Technology, China
| | - Bin Yu
- Qingdao University of Science and Technology, China
| |
Collapse
|
44
|
Zhao W, Yuan X, Shen X, Jiang X, Shi C, He T, Hu X. Improving drug-drug interactions prediction with interpretability via meta-path-based information fusion. Brief Bioinform 2023; 24:7030845. [PMID: 36750041 DOI: 10.1093/bib/bbad041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/01/2023] [Accepted: 01/18/2023] [Indexed: 02/09/2023] Open
Abstract
Drug-drug interactions (DDIs) are compound effects when patients take two or more drugs at the same time, which may weaken the efficacy of drugs or cause unexpected side effects. Thus, accurately predicting DDIs is of great significance for the drug development and the drug safety surveillance. Although many methods have been proposed for the task, the biological knowledge related to DDIs is not fully utilized and the complex semantics among drug-related biological entities are not effectively captured in existing methods, leading to suboptimal performance. Moreover, the lack of interpretability for the predicted results also limits the wide application of existing methods for DDIs prediction. In this study, we propose a novel framework for predicting DDIs with interpretability. Specifically, we construct a heterogeneous information network (HIN) by explicitly utilizing the biological knowledge related to the procedure of inducing DDIs. To capture the complex semantics in HIN, a meta-path-based information fusion mechanism is proposed to learn high-quality representations of drugs. In addition, an attention mechanism is designed to combine semantic information obtained from meta-paths with different lengths to obtain final representations of drugs for DDIs prediction. Comprehensive experiments are conducted on 2410 approved drugs, and the results of predictive performance comparison show that our proposed framework outperforms selected representative baselines on the task of DDIs prediction. The results of ablation study and cold-start scenario indicate that the meta-path-based information fusion mechanism red is beneficial for capturing the complex semantics among drug-related biological entities. Moreover, the results of case study demonstrate that the designed attention mechanism is able to provide partial interpretability for the predicted DDIs. Therefore, the proposed method will be a feasible solution to the task of predicting DDIs.
Collapse
Affiliation(s)
- Weizhong Zhao
- Hubei Provincial Key Laboratory of Artificial Intelligence and Smart Learning, Central China Normal University, Wuhan, Hubei 430079, PR China
- School of Computer Science, Beijing University of Posts and Telecommunications, Beijing, 100876, PR China
- National Language Resources Monitoring & Research Center for Network Media, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Xueling Yuan
- Hubei Provincial Key Laboratory of Artificial Intelligence and Smart Learning, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Xianjun Shen
- Hubei Provincial Key Laboratory of Artificial Intelligence and Smart Learning, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Xingpeng Jiang
- Hubei Provincial Key Laboratory of Artificial Intelligence and Smart Learning, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Chuan Shi
- School of Computer Science, Beijing University of Posts and Telecommunications, Beijing 100876, PR China
| | - Tingting He
- Hubei Provincial Key Laboratory of Artificial Intelligence and Smart Learning, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Xiaohua Hu
- College of Computing & Informatics, Drexel University, Philadelphia, PA 19104, USA
| |
Collapse
|
45
|
Zhang J, Chen M, Liu J, Peng D, Dai Z, Zou X, Li Z. A Knowledge-Graph-Based Multimodal Deep Learning Framework for Identifying Drug-Drug Interactions. Molecules 2023; 28:molecules28031490. [PMID: 36771157 PMCID: PMC9919258 DOI: 10.3390/molecules28031490] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The identification of drug-drug interactions (DDIs) plays a crucial role in various areas of drug development. In this study, a deep learning framework (KGCN_NFM) is presented to recognize DDIs using coupling knowledge graph convolutional networks (KGCNs) with neural factorization machines (NFMs). A KGCN is used to learn the embedding representation containing high-order structural information and semantic information in the knowledge graph (KG). The embedding and the Morgan molecular fingerprint of drugs are then used as input of NFMs to predict DDIs. The performance and effectiveness of the current method have been evaluated and confirmed based on the two real-world datasets with different sizes, and the results demonstrate that KGCN_NFM outperforms the state-of-the-art algorithms. Moreover, the identified interactions between topotecan and dantron by KGCN_NFM were validated through MTT assays, apoptosis experiments, cell cycle analysis, and molecular docking. Our study shows that the combination therapy of the two drugs exerts a synergistic anticancer effect, which provides an effective treatment strategy against lung carcinoma. These results reveal that KGCN_NFM is a valuable tool for integrating heterogeneous information to identify potential DDIs.
Collapse
Affiliation(s)
- Jing Zhang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Meng Chen
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Jie Liu
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Dongdong Peng
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zong Dai
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Xiaoyong Zou
- School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
- Correspondence: (X.Z.); (Z.L.)
| | - Zhanchao Li
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Correspondence: (X.Z.); (Z.L.)
| |
Collapse
|
46
|
Hong E, Jeon J, Kim HU. Recent development of machine learning models for the prediction of drug-drug interactions. KOREAN J CHEM ENG 2023; 40:276-285. [PMID: 36748027 PMCID: PMC9894510 DOI: 10.1007/s11814-023-1377-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/09/2022] [Accepted: 12/16/2022] [Indexed: 02/05/2023]
Abstract
Polypharmacy, the co-administration of multiple drugs, has become an area of concern as the elderly population grows and an unexpected infection, such as COVID-19 pandemic, keeps emerging. However, it is very costly and time-consuming to experimentally examine the pharmacological effects of polypharmacy. To address this challenge, machine learning models that predict drug-drug interactions (DDIs) have actively been developed in recent years. In particular, the growing volume of drug datasets and the advances in machine learning have facilitated the model development. In this regard, this review discusses the DDI-predicting machine learning models that have been developed since 2018. Our discussion focuses on dataset sources used to develop the models, featurization approaches of molecular structures and biological information, and types of DDI prediction outcomes from the models. Finally, we make suggestions for research opportunities in this field.
Collapse
Affiliation(s)
- Eujin Hong
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141 Korea
| | - Junhyeok Jeon
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141 Korea
| | - Hyun Uk Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141 Korea
- BioProcess Engineering Research Center and BioInformatics Research Center, KAIST, Daejeon, 34141 Korea
| |
Collapse
|
47
|
Artificial Intelligence and Data Mining for the Pharmacovigilance of Drug-Drug Interactions. Clin Ther 2023; 45:117-133. [PMID: 36732152 DOI: 10.1016/j.clinthera.2023.01.002] [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: 10/02/2022] [Revised: 12/15/2022] [Accepted: 01/09/2023] [Indexed: 02/01/2023]
Abstract
Despite increasing mechanistic understanding, undetected and underrecognized drug-drug interactions (DDIs) persist. This elusiveness relates to an interwoven complexity of increasing polypharmacy, multiplex mechanistic pathways, and human biological individuality. This persistent elusiveness motivates development of artificial intelligence (AI)-based approaches to enhancing DDI detection and prediction capabilities. The literature is vast and roughly divided into "prediction" and "detection." The former relatively emphasizes biological and chemical knowledge bases, drug development, new drugs, and beneficial interactions, whereas the latter utilizes more traditional sources such as spontaneous reports, claims data, and electronic health records to detect novel adverse DDIs with authorized drugs. However, it is not a bright line, either nominally or in practice, and both are in scope for pharmacovigilance supporting signal detection but also signal refinement and evaluation, by providing data-based mechanistic arguments for/against DDI signals. The wide array of intricate and elegant methods has expanded the pharmacovigilance tool kit. How much they add to real prospective pharmacovigilance, reduce the public health impact of DDIs, and at what cost in terms of false alarms amplified by automation bias and its sequelae are open questions. (Clin Ther. 2023;45:XXX-XXX) © 2023 Elsevier HS Journals, Inc.
Collapse
|
48
|
Ma M, Lei X. A dual graph neural network for drug-drug interactions prediction based on molecular structure and interactions. PLoS Comput Biol 2023; 19:e1010812. [PMID: 36701288 PMCID: PMC9879511 DOI: 10.1371/journal.pcbi.1010812] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/12/2022] [Indexed: 01/27/2023] Open
Abstract
Expressive molecular representation plays critical roles in researching drug design, while effective methods are beneficial to learning molecular representations and solving related problems in drug discovery, especially for drug-drug interactions (DDIs) prediction. Recently, a lot of work has been put forward using graph neural networks (GNNs) to forecast DDIs and learn molecular representations. However, under the current GNNs structure, the majority of approaches learn drug molecular representation from one-dimensional string or two-dimensional molecular graph structure, while the interaction information between chemical substructure remains rarely explored, and it is neglected to identify key substructures that contribute significantly to the DDIs prediction. Therefore, we proposed a dual graph neural network named DGNN-DDI to learn drug molecular features by using molecular structure and interactions. Specifically, we first designed a directed message passing neural network with substructure attention mechanism (SA-DMPNN) to adaptively extract substructures. Second, in order to improve the final features, we separated the drug-drug interactions into pairwise interactions between each drug's unique substructures. Then, the features are adopted to predict interaction probability of a DDI tuple. We evaluated DGNN-DDI on real-world dataset. Compared to state-of-the-art methods, the model improved DDIs prediction performance. We also conducted case study on existing drugs aiming to predict drug combinations that may be effective for the novel coronavirus disease 2019 (COVID-19). Moreover, the visual interpretation results proved that the DGNN-DDI was sensitive to the structure information of drugs and able to detect the key substructures for DDIs. These advantages demonstrated that the proposed method enhanced the performance and interpretation capability of DDI prediction modeling.
Collapse
Affiliation(s)
- Mei Ma
- School of Computer Science, Shaanxi Normal University, Xi’an, China
- School of Mathematics and Statistics, Qinghai Normal University, Qinghai, China
| | - Xiujuan Lei
- School of Computer Science, Shaanxi Normal University, Xi’an, China
- * E-mail:
| |
Collapse
|
49
|
Lin J, Wu L, Zhu J, Liang X, Xia Y, Xie S, Qin T, Liu TY. R2-DDI: relation-aware feature refinement for drug-drug interaction prediction. Brief Bioinform 2023; 24:6961471. [PMID: 36573491 DOI: 10.1093/bib/bbac576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/14/2022] [Accepted: 11/25/2022] [Indexed: 12/28/2022] Open
Abstract
Precisely predicting the drug-drug interaction (DDI) is an important application and host research topic in drug discovery, especially for avoiding the adverse effect when using drug combination treatment for patients. Nowadays, machine learning and deep learning methods have achieved great success in DDI prediction. However, we notice that most of the works ignore the importance of the relation type when building the DDI prediction models. In this work, we propose a novel R$^2$-DDI framework, which introduces a relation-aware feature refinement module for drug representation learning. The relation feature is integrated into drug representation and refined in the framework. With the refinement features, we also incorporate the consistency training method to regularize the multi-branch predictions for better generalization. Through extensive experiments and studies, we demonstrate our R$^2$-DDI approach can significantly improve the DDI prediction performance over multiple real-world datasets and settings, and our method shows better generalization ability with the help of the feature refinement design.
Collapse
Affiliation(s)
- Jiacheng Lin
- Department of Automation, Tsinghua University, 30 Shuangqing Rd, Haidian District, 100084 Beijing, China
| | - Lijun Wu
- Microsoft Research AI4Science, No. 5 Dan Ling Street, Haidian District, 100080 Beijing, China
| | - Jinhua Zhu
- CAS Key Laboratory of GIPAS, EEIS Department, University of Science and Technology of China, No. 96, JinZhai Road Baohe District, 230026 Hefei, Anhui Province, China
| | - Xiaobo Liang
- Institute of Artificial Intelligence, Soochow University, No. 178, Yucai Rd, Gusu District, 215006 Soochow, Jaingsu Province, China
| | - Yingce Xia
- Microsoft Research AI4Science, No. 5 Dan Ling Street, Haidian District, 100080 Beijing, China
| | - Shufang Xie
- Microsoft Research AI4Science, No. 5 Dan Ling Street, Haidian District, 100080 Beijing, China
| | - Tao Qin
- Microsoft Research AI4Science, No. 5 Dan Ling Street, Haidian District, 100080 Beijing, China
| | - Tie-Yan Liu
- Microsoft Research AI4Science, No. 5 Dan Ling Street, Haidian District, 100080 Beijing, China
| |
Collapse
|
50
|
MSResG: Using GAE and Residual GCN to Predict Drug-Drug Interactions Based on Multi-source Drug Features. Interdiscip Sci 2023; 15:171-188. [PMID: 36646843 DOI: 10.1007/s12539-023-00550-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/18/2023]
Abstract
Drug-drug interaction refers to taking the two drugs may produce certain reaction which may be a threat to patients' health, or enhance the efficacy helpful for medical work. Therefore, it is necessary to study and predict it. In fact, traditional experimental methods can be used for drug-drug interaction prediction, but they are time-consuming and costly, so we prefer to use more accurate and convenient calculation methods to predict the unknown drug-drug interaction. In this paper, we proposed a deep learning framework called MSResG that considers multi-sources features of drugs and combines them with Graph Auto-Encoder to predicting. Firstly, the model obtains four feature representations of drugs from the database, namely, chemical substructure, target, pathway and enzyme, and then calculates the Jaccard similarity of the drugs. To balance different drug features, we perform similarity integration by finding the mean value. Then we will be comprehensive similarity network combined with drug interaction network, and encodes and decodes it using the graph auto-encoder based on residual graph convolution network. Encoding is to learn the potential feature vectors of drugs, which contain similar information and interaction information. Decoding is to reconstruct the network to predict unknown drug-drug interaction. The experimental results show that our model has advanced performance and is superior to other existing advanced methods. Case study also shows that MSResG has practical significance.
Collapse
|