1
|
Carpenter KA, Altman RB. Databases of ligand-binding pockets and protein-ligand interactions. Comput Struct Biotechnol J 2024; 23:1320-1338. [PMID: 38585646 PMCID: PMC10997877 DOI: 10.1016/j.csbj.2024.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/16/2024] [Accepted: 03/17/2024] [Indexed: 04/09/2024] Open
Abstract
Many research groups and institutions have created a variety of databases curating experimental and predicted data related to protein-ligand binding. The landscape of available databases is dynamic, with new databases emerging and established databases becoming defunct. Here, we review the current state of databases that contain binding pockets and protein-ligand binding interactions. We have compiled a list of such databases, fifty-three of which are currently available for use. We discuss variation in how binding pockets are defined and summarize pocket-finding methods. We organize the fifty-three databases into subgroups based on goals and contents, and describe standard use cases. We also illustrate that pockets within the same protein are characterized differently across different databases. Finally, we assess critical issues of sustainability, accessibility and redundancy.
Collapse
Affiliation(s)
- Kristy A. Carpenter
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Russ B. Altman
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
2
|
Yi Z, Xie M. Polypharmacy side effect prediction based on semi-implicit graph variational auto-encoder. J Bioinform Comput Biol 2024; 22:2450020. [PMID: 39262053 DOI: 10.1142/s0219720024500203] [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: 09/13/2024]
Abstract
Polypharmacy, the use of drug combinations, is an effective approach for treating complex diseases, but it increases the risk of adverse effects. To predict novel polypharmacy side effects based on known ones, many computational methods have been proposed. However, most of them generate deterministic low-dimensional embeddings when modeling the latent space of drugs, which cannot effectively capture potential side effect associations between drugs. In this study, we present SIPSE, a novel approach for predicting polypharmacy side effects. SIPSE integrates single-drug side effect information and drug-target protein data to construct novel drug feature vectors. Leveraging a semi-implicit graph variational auto-encoder, SIPSE models known polypharmacy side effects and generates flexible latent distributions for drug nodes. SIPSE infers the current node distribution by combining the distributions of neighboring nodes with embedding noise. By sampling node embeddings from these distributions, SIPSE effectively predicts polypharmacy side effects between drugs. One key innovation of SIPSE is its incorporation of uncertainty propagation through noise embedding and neighborhood sharing, enhancing its graph analysis capabilities. Extensive experiments on a benchmark dataset of polypharmacy side effects demonstrated that SIPSE significantly outperformed five state-of-the-art methods in predicting polypharmacy side effects.
Collapse
Affiliation(s)
- Zhou Yi
- College of Information Science and Engineering, Hunan Normal University, Changsha 410081, P. R. China
| | - Minzhu Xie
- College of Information Science and Engineering, Hunan Normal University, Changsha 410081, P. R. China
- Key Laboratory of Computing and Stochastic, Mathematics (LCSM), (Ministry of Education), School of Mathematics and Statistics, Changsha 410081, P. R. China
| |
Collapse
|
3
|
Hauben M, Rafi M, Abdelaziz I, Hassanzadeh O. Knowledge Graphs in Pharmacovigilance: A Scoping Review. Clin Ther 2024; 46:544-554. [PMID: 38981792 DOI: 10.1016/j.clinthera.2024.06.003] [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: 12/12/2023] [Revised: 05/08/2024] [Accepted: 06/05/2024] [Indexed: 07/11/2024]
Abstract
PURPOSE To critically assess the role and added value of knowledge graphs in pharmacovigilance, focusing on their ability to predict adverse drug reactions. METHODS A systematic scoping review was conducted in which detailed information, including objectives, technology, data sources, methodology, and performance metrics, were extracted from a set of peer-reviewed publications reporting the use of knowledge graphs to support pharmacovigilance signal detection. FINDINGS The review, which included 47 peer-reviewed articles, found knowledge graphs were utilized for detecting/predicting single-drug adverse reactions and drug-drug interactions, with variable reported performance and sparse comparisons to legacy methods. IMPLICATIONS Research to date suggests that knowledge graphs have the potential to augment predictive signal detection in pharmacovigilance, but further research using more reliable reference sets of adverse drug reactions and comparison with legacy pharmacovigilance methods are needed to more clearly define best practices and to establish their place in holistic pharmacovigilance systems.
Collapse
Affiliation(s)
- Manfred Hauben
- Department of Family and Community Medicine, New York Medical College, Valhalla, New York; Truliant Consulting, Baltimore, Maryland
| | - Mazin Rafi
- Department of Statistics, Rutgers University, Piscataway, New Jersey.
| | | | | |
Collapse
|
4
|
Kee G, Kang HJ, Ahn I, Gwon H, Kim Y, Seo H, Choi H, Cho HN, Kim M, Han J, Park S, Kim K, Jun TJ, Kim YH. Are polypharmacy side effects predicted by public data still valid in real-world data? Heliyon 2024; 10:e24620. [PMID: 38304832 PMCID: PMC10831713 DOI: 10.1016/j.heliyon.2024.e24620] [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: 10/24/2023] [Revised: 12/29/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
Background and Objective Although interest in predicting drug-drug interactions is growing, many predictions are not verified by real-world data. This study aimed to confirm whether predicted polypharmacy side effects using public data also occur in data from actual patients. Methods We utilized a deep learning-based polypharmacy side effects prediction model to identify cefpodoxime-chlorpheniramine-lung edema combination with a high prediction score and a significant patient population. The retrospective study analyzed patients over 18 years old who were admitted to the Asan medical center between January 2000 and December 2020 and took cefpodoxime or chlorpheniramine orally. The three groups, cefpodoxime-treated, chlorpheniramine-treated, and cefpodoxime & chlorpheniramine-treated were compared using inverse probability of treatment weighting (IPTW) to balance them. Differences between the three groups were analyzed using the Kaplan-Meier method and Cox proportional hazards model. Results The study population comprised 54,043 patients with a history of taking cefpodoxime, 203,897 patients with a history of taking chlorpheniramine, and 1,628 patients with a history of taking cefpodoxime and chlorpheniramine simultaneously. After adjustment, the 1-year cumulative incidence of lung edema in the patient group that took cefpodoxime and chlorpheniramine simultaneously was significantly higher than in the patient groups that took cefpodoxime or chlorpheniramine only (p=0.001). Patients taking cefpodoxime and chlorpheniramine together had an increased risk of lung edema compared to those taking cefpodoxime alone [hazard ratio (HR) 2.10, 95% CI 1.26-3.52, p<0.005] and those taking chlorpheniramine alone, which also increased the risk of lung edema (HR 1.64, 95% CI 0.99-2.69, p=0.05). Conclusions Validation of polypharmacy side effect predictions with real-world data can aid patient and clinician decision-making before conducting randomized controlled trials. Simultaneous use of cefpodoxime and chlorpheniramine was associated with a higher long-term risk of lung edema compared to the use of cefpodoxime or chlorpheniramine alone.
Collapse
Affiliation(s)
- Gaeun Kee
- Department of Information Medicine, Asan Medical Center, 88, Olympicro 43gil, Songpagu, 05505, Seoul, Republic of Korea
| | - Hee Jun Kang
- Division of Cardiology, Asan Medical Center, 88, Olympicro 43gil, Songpagu, 05505, Seoul, Republic of Korea
| | - Imjin Ahn
- Department of Information Medicine, Asan Medical Center, 88, Olympicro 43gil, Songpagu, 05505, Seoul, Republic of Korea
| | - Hansle Gwon
- Department of Information Medicine, Asan Medical Center, 88, Olympicro 43gil, Songpagu, 05505, Seoul, Republic of Korea
| | - Yunha Kim
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympicro 43gil, Songpagu, 05505, Seoul, Republic of Korea
| | - Hyeram Seo
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympicro 43gil, Songpagu, 05505, Seoul, Republic of Korea
| | - Heejung Choi
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympicro 43gil, Songpagu, 05505, Seoul, Republic of Korea
| | - Ha Na Cho
- Department of Information Medicine, Asan Medical Center, 88, Olympicro 43gil, Songpagu, 05505, Seoul, Republic of Korea
| | - Minkyoung Kim
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympicro 43gil, Songpagu, 05505, Seoul, Republic of Korea
| | - JiYe Han
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympicro 43gil, Songpagu, 05505, Seoul, Republic of Korea
| | - Seohyun Park
- Department of Information Medicine, Asan Medical Center, 88, Olympicro 43gil, Songpagu, 05505, Seoul, Republic of Korea
| | - Kyuwoong Kim
- National Cancer Control Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, 10408, Goyang, Republic of Korea
| | - Tae Joon Jun
- Big Data Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympicro 43gil, Songpagu, 05505, Seoul, Republic of Korea
| | - Young-Hak Kim
- Division of Cardiology, Department of Information Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympicro 43gil, Songpagu, 05505, Seoul, Republic of Korea
| |
Collapse
|
5
|
Huang T, Lin KH, Machado-Vieira R, Soares JC, Jiang X, Kim Y. Explainable drug side effect prediction via biologically informed graph neural network. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.26.23290615. [PMID: 37333107 PMCID: PMC10275013 DOI: 10.1101/2023.05.26.23290615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Early detection of potential side effects (SE) is a critical and challenging task for drug discovery and patient care. In-vitro or in-vivo approach to detect potential SEs is not scalable for many drug candidates during the preclinical stage. Recent advances in explainable machine learning may facilitate detecting potential SEs of new drugs before market release and elucidating the critical mechanism of biological actions. Here, we leverage multi-modal interactions among molecules to develop a biologically informed graph-based SE prediction model, called HHAN-DSI. HHAN-DSI predicted frequent and even uncommon SEs of the unseen drug with higher or comparable accuracy against benchmark methods. When applying HHAN-DSI to the central nervous system, the organs with the largest number of SEs, the model revealed diverse psychiatric medications' previously unknown but probable SEs, together with the potential mechanisms of actions through a network of genes, biological functions, drugs, and SEs.
Collapse
Affiliation(s)
- Tongtong Huang
- School of Biomedical Informatics, UTHealth, Houston, TX, United States
| | - Ko-Hong Lin
- School of Biomedical Informatics, UTHealth, Houston, TX, United States
| | - Rodrigo Machado-Vieira
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, UTHealth, Houston, TX, United States
| | - Jair C Soares
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, UTHealth, Houston, TX, United States
| | - Xiaoqian Jiang
- School of Biomedical Informatics, UTHealth, Houston, TX, United States
| | - Yejin Kim
- School of Biomedical Informatics, UTHealth, Houston, TX, United States
| |
Collapse
|
6
|
Li H, Zou L, Kowah JAH, He D, Liu Z, Ding X, Wen H, Wang L, Yuan M, Liu X. A compact review of progress and prospects of deep learning in drug discovery. J Mol Model 2023; 29:117. [PMID: 36976427 DOI: 10.1007/s00894-023-05492-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 02/27/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND Drug discovery processes, such as new drug development, drug synergy, and drug repurposing, consume significant yearly resources. Computer-aided drug discovery can effectively improve the efficiency of drug discovery. Traditional computer methods such as virtual screening and molecular docking have achieved many gratifying results in drug development. However, with the rapid growth of computer science, data structures have changed considerably; with more extensive and dimensional data and more significant amounts of data, traditional computer methods can no longer be applied well. Deep learning methods are based on deep neural network structures that can handle high-dimensional data very well, so they are used in current drug development. RESULTS This review summarized the applications of deep learning methods in drug discovery, such as drug target discovery, drug de novo design, drug recommendation, drug synergy, and drug response prediction. While applying deep learning methods to drug discovery suffers from a lack of data, transfer learning is an excellent solution to this problem. Furthermore, deep learning methods can extract deeper features and have higher predictive power than other machine learning methods. Deep learning methods have great potential in drug discovery and are expected to facilitate drug discovery development.
Collapse
Affiliation(s)
- Huijun Li
- College of Medicine, Guangxi University, Nanning, 530004, China
| | - Lin Zou
- College of Medicine, Guangxi University, Nanning, 530004, China
| | | | - Dongqiong He
- College of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, China
| | - Zifan Liu
- College of Medicine, Guangxi University, Nanning, 530004, China
| | - Xuejie Ding
- College of Medicine, Guangxi University, Nanning, 530004, China
| | - Hao Wen
- College of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, China
| | - Lisheng Wang
- College of Medicine, Guangxi University, Nanning, 530004, China
| | - Mingqing Yuan
- College of Medicine, Guangxi University, Nanning, 530004, China
| | - Xu Liu
- College of Medicine, Guangxi University, Nanning, 530004, China.
| |
Collapse
|
7
|
Fujita K, Masnoon N, Mach J, O’Donnell LK, Hilmer SN. Polypharmacy and precision medicine. CAMBRIDGE PRISMS. PRECISION MEDICINE 2023; 1:e22. [PMID: 38550925 PMCID: PMC10953761 DOI: 10.1017/pcm.2023.10] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 07/05/2024]
Abstract
Precision medicine is an approach to maximise the effectiveness of disease treatment and prevention and minimise harm from medications by considering relevant demographic, clinical, genomic and environmental factors in making treatment decisions. Precision medicine is complex, even for decisions about single drugs for single diseases, as it requires expert consideration of multiple measurable factors that affect pharmacokinetics and pharmacodynamics, and many patient-specific variables. Given the increasing number of patients with multiple conditions and medications, there is a need to apply lessons learned from precision medicine in monotherapy and single disease management to optimise polypharmacy. However, precision medicine for optimisation of polypharmacy is particularly challenging because of the vast number of interacting factors that influence drug use and response. In this narrative review, we aim to provide and apply the latest research findings to achieve precision medicine in the context of polypharmacy. Specifically, this review aims to (1) summarise challenges in achieving precision medicine specific to polypharmacy; (2) synthesise the current approaches to precision medicine in polypharmacy; (3) provide a summary of the literature in the field of prediction of unknown drug-drug interactions (DDI) and (4) propose a novel approach to provide precision medicine for patients with polypharmacy. For our proposed model to be implemented in routine clinical practice, a comprehensive intervention bundle needs to be integrated into the electronic medical record using bioinformatic approaches on a wide range of data to predict the effects of polypharmacy regimens on an individual. In addition, clinicians need to be trained to interpret the results of data from sources including pharmacogenomic testing, DDI prediction and physiological-pharmacokinetic-pharmacodynamic modelling to inform their medication reviews. Future studies are needed to evaluate the efficacy of this model and to test generalisability so that it can be implemented at scale, aiming to improve outcomes in people with polypharmacy.
Collapse
Affiliation(s)
- Kenji Fujita
- Departments of Clinical Pharmacology and Aged Care, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Nashwa Masnoon
- Departments of Clinical Pharmacology and Aged Care, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - John Mach
- Departments of Clinical Pharmacology and Aged Care, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Lisa Kouladjian O’Donnell
- Departments of Clinical Pharmacology and Aged Care, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Sarah N. Hilmer
- Departments of Clinical Pharmacology and Aged Care, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| |
Collapse
|
8
|
Zhang H, Wang Y, Pan Z, Sun X, Mou M, Zhang B, Li Z, Li H, Zhu F. ncRNAInter: a novel strategy based on graph neural network to discover interactions between lncRNA and miRNA. Brief Bioinform 2022; 23:6747810. [PMID: 36198065 DOI: 10.1093/bib/bbac411] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/04/2022] [Accepted: 08/23/2022] [Indexed: 12/14/2022] Open
Abstract
In recent years, many studies have illustrated the significant role that non-coding RNA (ncRNA) plays in biological activities, in which lncRNA, miRNA and especially their interactions have been proved to affect many biological processes. Some in silico methods have been proposed and applied to identify novel lncRNA-miRNA interactions (LMIs), but there are still imperfections in their RNA representation and information extraction approaches, which imply there is still room for further improving their performances. Meanwhile, only a few of them are accessible at present, which limits their practical applications. The construction of a new tool for LMI prediction is thus imperative for the better understanding of their relevant biological mechanisms. This study proposed a novel method, ncRNAInter, for LMI prediction. A comprehensive strategy for RNA representation and an optimized deep learning algorithm of graph neural network were utilized in this study. ncRNAInter was robust and showed better performance of 26.7% higher Matthews correlation coefficient than existing reputable methods for human LMI prediction. In addition, ncRNAInter proved its universal applicability in dealing with LMIs from various species and successfully identified novel LMIs associated with various diseases, which further verified its effectiveness and usability. All source code and datasets are freely available at https://github.com/idrblab/ncRNAInter.
Collapse
Affiliation(s)
- Hanyu Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Yunxia Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ziqi Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiuna Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bing Zhang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Zhaorong Li
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Honglin Li
- School of Computer Science and Technology, East China Normal University, Shanghai 200062, China.,Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai 200237, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
9
|
Lin S, Zhang G, Wei DQ, Xiong Y. DeepPSE: Prediction of polypharmacy side effects by fusing deep representation of drug pairs and attention mechanism. Comput Biol Med 2022; 149:105984. [PMID: 35994933 DOI: 10.1016/j.compbiomed.2022.105984] [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: 06/22/2022] [Revised: 07/17/2022] [Accepted: 08/14/2022] [Indexed: 11/18/2022]
Abstract
Polypharmacy (multiple use of drugs) is an effective strategy for combating complex or co-existing diseases. However, a major consequence of polypharmacy is a higher risk of adverse side effects due to drug-drug interactions, which are rare and observed in relatively small clinical testing. Thus, identification of polypharmacy side effects remains challenging. Here, we propose a deep learning-based method, DeepPSE, to predict polypharmacy side effects in an end-to-end way. DeepPSE is composed of two main modules. First, multiple types of neural networks are constructed and fused to learn the deep representation of a drug pair. Second, the encoder block of transformer that includes self-attention mechanism is built to get latent features, which are further fed into the fully connected layer to predict polypharmacy side effects of drug pairs. Further, DeepPSE is compared with five baseline or state-of-the-art methods on a benchmark dataset of 964 types of polypharmacy side effects across 63473 drug pairs. Experimental results demonstrate that DeepPSE achieves better performance than that of all five methods. The source codes and data are available at https://github.com/ShenggengLin/DeepPSE.
Collapse
Affiliation(s)
- Shenggeng Lin
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Guangwei Zhang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510275, 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, Zhongguancun Scientific Park, Meixi, Nayang, Henan, 473006, China; Peng Cheng National Laboratory, Vanke Cloud City Phase I Building 8, Xili Street, Nanshan District, Shenzhen, Guangdong, 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, China.
| |
Collapse
|
10
|
Liu Q, Yao E, Liu C, Zhou X, Li Y, Xu M. M2GCN: multi-modal graph convolutional network for modeling polypharmacy side effects. APPL INTELL 2022. [DOI: 10.1007/s10489-022-03839-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
11
|
Yao J, Sun W, Jian Z, Wu Q, Wang X. Effective knowledge graph embeddings based on multidirectional semantics relations for polypharmacy side effects prediction. Bioinformatics 2022; 38:2315-2322. [PMID: 35176135 DOI: 10.1093/bioinformatics/btac094] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/25/2022] [Accepted: 02/15/2022] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION Polypharmacy is the combined use of drugs for the treatment of diseases. However, it often shows a high risk of side effects. Due to unnecessary interactions of combined drugs, the side effects of polypharmacy increase the risk of disease and even lead to death. Thus, obtaining abundant and comprehensive information on the side effects of polypharmacy is a vital task in the healthcare industry. Early traditional methods used machine learning techniques to predict side effects. However, they often make costly efforts to extract features of drugs for prediction. Later, several methods based on knowledge graphs are proposed. They are reported to outperform traditional methods. However, they still show limited performance by failing to model complex relations of side effects among drugs. RESULTS To resolve the above problems, we propose a novel model by further incorporating complex relations of side effects into knowledge graph embeddings. Our model can translate and transmit multidirectional semantics with fewer parameters, leading to better scalability in large-scale knowledge graphs. Experimental evaluation shows that our model outperforms state-of-the-art models in terms of the average area under the ROC and precision-recall curves. AVAILABILITY AND IMPLEMENTATION Code and data are available at: https://github.com/galaxysunwen/MSTE-master.
Collapse
Affiliation(s)
- Junfeng Yao
- School of Informatics, Xiamen University, Xiamen, Fujian 361005, China.,Institute of Artificial Intelligence, Xiamen University, Xiamen, Fujian 361005, China.,Key Laboratory of Digital Protection and Intelligent Processing of Intangible Cultural Heritage of Fujian and Taiwan Ministry of Culture and Tourism, Xiamen University, Xiamen, Fujian 361005, China
| | - Wen Sun
- School of Informatics, Xiamen University, Xiamen, Fujian 361005, China
| | - Zhongquan Jian
- School of Informatics, Xiamen University, Xiamen, Fujian 361005, China.,Institute of Artificial Intelligence, Xiamen University, Xiamen, Fujian 361005, China
| | - Qingqiang Wu
- School of Informatics, Xiamen University, Xiamen, Fujian 361005, China.,Institute of Artificial Intelligence, Xiamen University, Xiamen, Fujian 361005, China.,Key Laboratory of Digital Protection and Intelligent Processing of Intangible Cultural Heritage of Fujian and Taiwan Ministry of Culture and Tourism, Xiamen University, Xiamen, Fujian 361005, China
| | - Xiaoli Wang
- School of Informatics, Xiamen University, Xiamen, Fujian 361005, China
| |
Collapse
|
12
|
Zhu B, Xu Y, Zhao P, Yiu SM, Yu H, Shi JY. NNAN: Nearest Neighbor Attention Network to Predict Drug–Microbe Associations. Front Microbiol 2022; 13:846915. [PMID: 35479616 PMCID: PMC9035839 DOI: 10.3389/fmicb.2022.846915] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Many drugs can be metabolized by human microbes; the drug metabolites would significantly alter pharmacological effects and result in low therapeutic efficacy for patients. Hence, it is crucial to identify potential drug–microbe associations (DMAs) before the drug administrations. Nevertheless, traditional DMA determination cannot be applied in a wide range due to the tremendous number of microbe species, high costs, and the fact that it is time-consuming. Thus, predicting possible DMAs in computer technology is an essential topic. Inspired by other issues addressed by deep learning, we designed a deep learning-based model named Nearest Neighbor Attention Network (NNAN). The proposed model consists of four components, namely, a similarity network constructor, a nearest-neighbor aggregator, a feature attention block, and a predictor. In brief, the similarity block contains a microbe similarity network and a drug similarity network. The nearest-neighbor aggregator generates the embedding representations of drug–microbe pairs by integrating drug neighbors and microbe neighbors of each drug–microbe pair in the network. The feature attention block evaluates the importance of each dimension of drug–microbe pair embedding by a set of ordinary multi-layer neural networks. The predictor is an ordinary fully-connected deep neural network that functions as a binary classifier to distinguish potential DMAs among unlabeled drug–microbe pairs. Several experiments on two benchmark databases are performed to evaluate the performance of NNAN. First, the comparison with state-of-the-art baseline approaches demonstrates the superiority of NNAN under cross-validation in terms of predicting performance. Moreover, the interpretability inspection reveals that a drug tends to associate with a microbe if it finds its top-l most similar neighbors that associate with the microbe.
Collapse
Affiliation(s)
- Bei Zhu
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Yi Xu
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Pengcheng Zhao
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Siu-Ming Yiu
- Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Hui Yu
- School of Computer Science, Northwestern Polytechnical University, Xi’an, China
- *Correspondence: Hui Yu,
| | - Jian-Yu Shi
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- Jian-Yu Shi,
| |
Collapse
|
13
|
Pang S, Zhang Y, Song T, Zhang X, Wang X, Rodriguez-Patón A. AMDE: a novel attention-mechanism-based multidimensional feature encoder for drug-drug interaction prediction. Brief Bioinform 2021; 23:6489100. [PMID: 34965586 DOI: 10.1093/bib/bbab545] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/31/2021] [Accepted: 11/26/2021] [Indexed: 11/14/2022] Open
Abstract
The properties of the drug may be altered by the combination, which may cause unexpected drug-drug interactions (DDIs). Prediction of DDIs provides combination strategies of drugs for systematic and effective treatment. In most of deep learning-based methods for predicting DDI, encoded information about the drugs is insufficient in some extent, which limits the performances of DDIs prediction. In this work, we propose a novel attention-mechanism-based multidimensional feature encoder for DDIs prediction, namely attention-based multidimensional feature encoder (AMDE). Specifically, in AMDE, we encode drug features from multiple dimensions, including information from both Simplified Molecular-Input Line-Entry System sequence and atomic graph of the drug. Data experiments are conducted on DDI data set selected from Drugbank, involving a total of 34 282 DDI relationships with 17 141 positive DDI samples and 17 141 negative samples. Experimental results show that our AMDE performs better than some state-of-the-art baseline methods, including Random Forest, One-Dimension Convolutional Neural Networks, DeepDrug, Long Short-Term Memory, Seq2seq, Deepconv, DeepDDI, Graph Attention Networks and Knowledge Graph Neural Networks. In practice, we select a set of 150 drugs with 3723 DDIs, which are never appeared in training, validation and test sets. AMDE performs well in DDIs prediction task, with AUROC and AUPRC 0.981 and 0.975. As well, we use Torasemide (DB00214) as an example and predict the most likely drug to interact with it. The top 15 scores all have been reported with clear interactions in literatures.
Collapse
Affiliation(s)
- Shanchen Pang
- College of Computer Science and Technology, China University of Petroleum, Qingdao 266580, China
| | - Ying Zhang
- College of Computer Science and Technology, China University of Petroleum, Qingdao 266580, China
| | - Tao Song
- College of Computer Science and Technology, China University of Petroleum, Qingdao 266580, China.,Department of Artificial Intelligence, Faculty of Computer Science, Polytechnical University of Madrid, Campus de Montegancedo, Boadilla del Monte 28660, Madrid, Spain
| | - Xudong Zhang
- College of Computer Science and Technology, China University of Petroleum, Qingdao 266580, China
| | - Xun Wang
- College of Computer Science and Technology, China University of Petroleum, Qingdao 266580, China.,China High Performance Computer Research Center, Institute of Computer Technology, Chinese Academy of Science, Beijing, 100190 Beijing, China
| | - Alfonso Rodriguez-Patón
- Department of Artificial Intelligence, Faculty of Computer Science, Polytechnical University of Madrid, Campus de Montegancedo, Boadilla del Monte 28660, Madrid, Spain
| |
Collapse
|
14
|
Zhang S, Wang J, Pei L, Liu K, Gao Y, Fang H, Zhang R, Zhao L, Sun S, Wu J, Song B, Dai H, Li R, Xu Y. Interpretability analysis of one-year mortality prediction for stroke patients based on deep neural network. IEEE J Biomed Health Inform 2021; 26:1903-1910. [PMID: 34714758 DOI: 10.1109/jbhi.2021.3123657] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Clinically, physicians collect the benchmark medical data to establish archives for a stroke patient and then add the follow up data regularly. It has great significance on prognosis prediction for stroke patients. In this paper, we present an interpretable deep learning model to predict the one-year mortality risk on stroke. We design sub-modules to reconstruct features from original clinical data that highlight the dissimilarity and temporality of different variables. The model consists of Bidirectional Long Short-Term Memory (Bi-LSTM), in which a novel correlation attention module is proposed that takes the correlation of variables into consideration. In experiments, datasets are collected clinically from the department of neurology in a local AAA hospital. It consists of 2,275 stroke patients hospitalized in the department of neurology from 2014 to 2016. Our model achieves a precision of 0.9414, a recall of 0.9502 and an F1-score of 0.9415. In addition, we provide the analysis of the interpretability by visualizations with reference to clinical professional guidelines.
Collapse
|