1
|
Chen L, Lu Y, Xu J, Zhou B. Prediction of drug's anatomical therapeutic chemical (ATC) code by constructing biological profiles of ATC codes. BMC Bioinformatics 2025; 26:86. [PMID: 40119265 PMCID: PMC11927162 DOI: 10.1186/s12859-025-06102-7] [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: 08/09/2024] [Accepted: 03/04/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND The Anatomical Therapeutic Chemical (ATC) classification system, proposed and maintained by the World Health Organization, is among the most widely used drug classification schemes. Recently, it has become a key research focus in drug repositioning. Computational models often pair drugs with ATC codes to explore drug-ATC code associations. However, the limited information available for ATC codes constrains these models, leaving significant room for improvement. RESULTS This study presents an inference method to identify highly related target proteins, structural features, and side effects for each ATC code, constructing comprehensive biological profiles. Association networks for target proteins, structural features, and side effects are established, and a random walk with restart algorithm is applied to these networks to extract raw associations. A permutation test is then conducted to exclude false positives, yielding robust biological profiles for ATC codes. These profiles are used to construct new ATC code kernels, which are integrated with ATC code kernels from the existing model PDATC-NCPMKL. The recommendation matrix is subsequently generated using the procedures of PDATC-NCPMKL. Cross-validation results demonstrate that the new model achieves AUROC and AUPR values exceeding 0.96. CONCLUSION The proposed model outperforms PDATC-NCPMKL and other previous models. Analysis of the contributions of the newly added ATC code kernels confirms the value of biological profiles in enhancing the prediction of drug-ATC code associations.
Collapse
Affiliation(s)
- Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, People's Republic of China.
| | - Yiwen Lu
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, People's Republic of China
| | - Jing Xu
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, People's Republic of China
| | - Bo Zhou
- School of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, People's Republic of China
| |
Collapse
|
2
|
Zhang W, Tian Q, Cao Y, Fan W, Jiang D, Wang Y, Li Q, Wei XY. GraphATC: advancing multilevel and multi-label anatomical therapeutic chemical classification via atom-level graph learning. Brief Bioinform 2025; 26:bbaf194. [PMID: 40285359 PMCID: PMC12031726 DOI: 10.1093/bib/bbaf194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/06/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
The accurate categorization of compounds within the anatomical therapeutic chemical (ATC) system is fundamental for drug development and fundamental research. Although this area has garnered significant research focus for over a decade, the majority of prior studies have concentrated solely on the Level 1 labels defined by the World Health Organization (WHO), neglecting the labels of the remaining four levels. This narrow focus fails to address the true nature of the task as a multilevel, multi-label classification challenge. Moreover, existing benchmarks like Chen-2012 and ATC-SMILES have become outdated, lacking the incorporation of new drugs or updated properties of existing ones that have emerged in recent years and have been integrated into the WHO ATC system. To tackle these shortcomings, we present a comprehensive approach in this paper. Firstly, we systematically cleanse and enhance the drug dataset, expanding it to encompass all five levels through a rigorous cross-resource validation process involving KEGG, PubChem, ChEMBL, ChemSpider, and ChemicalBook. This effort culminates in the creation of a novel benchmark termed ATC-GRAPH. Secondly, we extend the classification task to encompass Level 2 and introduce graph-based learning techniques to provide more accurate representations of drug molecular structures. This approach not only facilitates the modeling of Polymers, Macromolecules, and Multi-Component drugs more precisely but also enhances the overall fidelity of the classification process. The efficacy of our proposed framework is validated through extensive experiments, establishing a new state-of-the-art methodology. To facilitate the replication of this study, we have made the benchmark dataset, source code, and web server openly accessible.
Collapse
Affiliation(s)
- Wengyu Zhang
- Department of Computer Science, Sichuan University, Chengdu 610065, China
- Department of Computing, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Qi Tian
- Department of Computer Science, Sichuan University, Chengdu 610065, China
| | - Yi Cao
- Department of Computer Science, Sichuan University, Chengdu 610065, China
| | - Wenqi Fan
- Department of Computing, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | | | - Yaowei Wang
- Peng Cheng Laboratory, Shenzhen 518000, China
| | - Qing Li
- Department of Computing, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Xiao-Yong Wei
- Department of Computer Science, Sichuan University, Chengdu 610065, China
- Department of Computing, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| |
Collapse
|
3
|
Messa L, Testa C, Carelli S, Rey F, Jacchetti E, Cereda C, Raimondi MT, Ceri S, Pinoli P. Non-Negative Matrix Tri-Factorization for Representation Learning in Multi-Omics Datasets with Applications to Drug Repurposing and Selection. Int J Mol Sci 2024; 25:9576. [PMID: 39273521 PMCID: PMC11394968 DOI: 10.3390/ijms25179576] [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: 07/12/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
The vast corpus of heterogeneous biomedical data stored in databases, ontologies, and terminologies presents a unique opportunity for drug design. Integrating and fusing these sources is essential to develop data representations that can be analyzed using artificial intelligence methods to generate novel drug candidates or hypotheses. Here, we propose Non-Negative Matrix Tri-Factorization as an invaluable tool for integrating and fusing data, as well as for representation learning. Additionally, we demonstrate how representations learned by Non-Negative Matrix Tri-Factorization can effectively be utilized by traditional artificial intelligence methods. While this approach is domain-agnostic and applicable to any field with vast amounts of structured and semi-structured data, we apply it specifically to computational pharmacology and drug repurposing. This field is poised to benefit significantly from artificial intelligence, particularly in personalized medicine. We conducted extensive experiments to evaluate the performance of the proposed method, yielding exciting results, particularly compared to traditional methods. Novel drug-target predictions have also been validated in the literature, further confirming their validity. Additionally, we tested our method to predict drug synergism, where constructing a classical matrix dataset is challenging. The method demonstrated great flexibility, suggesting its applicability to a wide range of tasks in drug design and discovery.
Collapse
Affiliation(s)
- Letizia Messa
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, 20133 Milan, Italy
| | - Carolina Testa
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, 20133 Milan, Italy
| | - Stephana Carelli
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, 20154 Milan, Italy
- Pediatric Clinical Research Center "Fondazione Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Federica Rey
- Pediatric Clinical Research Center "Fondazione Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Emanuela Jacchetti
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, 20133 Milan, Italy
| | - Cristina Cereda
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, 20154 Milan, Italy
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, 20133 Milan, Italy
| | - Stefano Ceri
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, 20133 Milan, Italy
| | - Pietro Pinoli
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, 20133 Milan, Italy
| |
Collapse
|
4
|
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
|
5
|
Chen L, Xu J, Zhou Y. PDATC-NCPMKL: Predicting drug's Anatomical Therapeutic Chemical (ATC) codes based on network consistency projection and multiple kernel learning. Comput Biol Med 2024; 169:107862. [PMID: 38150886 DOI: 10.1016/j.compbiomed.2023.107862] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/19/2023] [Accepted: 12/17/2023] [Indexed: 12/29/2023]
Abstract
The development and discovery of new drugs is time-consuming and needs lots of human and material resources. Therefore, discovery of novel effects of existing drugs is an important alternative way, which can accelerate the process of designing "new" drugs. The anatomical Therapeutic Chemical (ATC) classification system recommended by World Health Organization (WHO) is a basic research area in this regard. A novel ATC code of an existing drug suggests its novel effects. Some computational models have been proposed, which can predict the drug-ATC code associations. However, their performance is not very high. There still exist spaces for improvement. In this study, a new recommendation system (named PDATC-NCPMKL), which incorporated network consistency projection and multi-kernel learning, was designed to identify drug-ATC code associations. For drugs or ATC codes, several kernels were constructed, which were fused by a multiple kernel learning method and an additional kernel integration scheme. To enhance the performance, the drug-ATC code association adjacency matrix was reformulated by a variant of weighted K nearest known neighbors (WKNKN). The reformulated adjacency matrix, drug and ATC code kernels were fed into network consistency projection to generate the association score matrix. The proposed recommendation system was tested on the ATC codes at the second, third and fourth levels in drug ATC classification system using ten-fold cross-validation. The results indicated that all AUROC and AUPR values were close to or exceeded 0.96. Such performance was higher than some existing computational models. Some additional tests were conducted to prove the utility of adjacency matrix reformulation and to analyze the importance of drug and ATC code kernels.
Collapse
Affiliation(s)
- Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, China.
| | - Jing Xu
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, China.
| | - Yubin Zhou
- Department of Thoracic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
6
|
Gao Z, Ding P, Xu R. IUPHAR review - Data-driven computational drug repurposing approaches for opioid use disorder. Pharmacol Res 2024; 199:106960. [PMID: 37832859 DOI: 10.1016/j.phrs.2023.106960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
Opioid Use Disorder (OUD) is a chronic and relapsing condition characterized by the misuse of opioid drugs, causing significant morbidity and mortality in the United States. Existing medications for OUD are limited, and there is an immediate need to discover treatments with enhanced safety and efficacy. Drug repurposing aims to find new indications for existing medications, offering a time-saving and cost-efficient alternative strategy to traditional drug discovery. Computational approaches have been developed to further facilitate the drug repurposing process. In this paper, we reviewed state-of-the-art data-driven computational drug repurposing approaches for OUD and discussed their advantages and potential challenges. We also highlighted promising repurposed candidate drugs for OUD that were identified by computational drug repurposing techniques and reviewed studies supporting their potential mechanisms of action in treating OUD.
Collapse
Affiliation(s)
- Zhenxiang Gao
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Pingjian Ding
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
7
|
Zhao H, Duan G, Ni P, Yan C, Li Y, Wang J. RNPredATC: A Deep Residual Learning-Based Model With Applications to the Prediction of Drug-ATC Code Association. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:2712-2723. [PMID: 34110998 DOI: 10.1109/tcbb.2021.3088256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The Anatomical Therapeutic Chemical (ATC) classification system, designated by the World Health Organization Collaborating Center (WHOCC), has been widely used in drug screening, repositioning, and similarity research. The ATC classification system assigns different codes to drugs according to the organ or system on which they act and/or their therapeutic and chemical characteristics. Correctly identifying the potential ATC codes for drugs can accelerate drug development and reduce the cost of experiments. Several classifiers have been proposed in this regard. However, they lack of ability to learn basic features from sparsely known drug-ATC code associations. Therefore, there is an urgent need for novel computational methods to precisely predict potential drug-ATC code associations in multiple levels of the ATC classification system based on known associations between drugs and ATC codes. In this paper, we provide a novel end-to-end model, so-called RNPredATC, to predict potential drug-ATC code associations in five ATC classification levels. RNPredATC can extract dense feature vectors from sparsely known drug-ATC code associations and reduce the impact from the degradation problem by a novel deep residual learning. We extensively compare our method with some state-of-the-art methods, including NetPredATC, SPACE, and some multi-label-based methods. Our experimental results show that RNPredATC achieves better performances in five-fold and ten-fold cross validations. Furthermore, the visualization analysis of hidden layers and case studies of predicted associations at the fifth ATC classification level confirm that RNPredATC can effectively identify the potential ATC codes of drugs.
Collapse
|
8
|
Yi J, Lee S, Lim S, Cho C, Piao Y, Yeo M, Kim D, Kim S, Lee S. Exploring chemical space for lead identification by propagating on chemical similarity network. Comput Struct Biotechnol J 2023; 21:4187-4195. [PMID: 37680266 PMCID: PMC10480321 DOI: 10.1016/j.csbj.2023.08.016] [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: 04/04/2023] [Revised: 08/08/2023] [Accepted: 08/20/2023] [Indexed: 09/09/2023] Open
Abstract
Motivation Lead identification is a fundamental step to prioritize candidate compounds for downstream drug discovery process. Machine learning (ML) and deep learning (DL) approaches are widely used to identify lead compounds using both chemical property and experimental information. However, ML or DL methods rarely consider compound similarity information directly since ML and DL models use abstract representation of molecules for model construction. Alternatively, data mining approaches are also used to explore chemical space with drug candidates by screening undesirable compounds. A major challenge for data mining approaches is to develop efficient data mining methods that search large chemical space for desirable lead compounds with low false positive rate. Results In this work, we developed a network propagation (NP) based data mining method for lead identification that performs search on an ensemble of chemical similarity networks. We compiled 14 fingerprint-based similarity networks. Given a target protein of interest, we use a deep learning-based drug target interaction model to narrow down compound candidates and then we use network propagation to prioritize drug candidates that are highly correlated with drug activity score such as IC50. In an extensive experiment with BindingDB, we showed that our approach successfully discovered intentionally unlabeled compounds for given targets. To further demonstrate the prediction power of our approach, we identified 24 candidate leads for CLK1. Two out of five synthesizable candidates were experimentally validated in binding assays. In conclusion, our framework can be very useful for lead identification from very large compound databases such as ZINC.
Collapse
Affiliation(s)
- Jungseob Yi
- Interdisciplinary Program in Artificial Intelligence, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul, 08826, South Korea
| | - Sangseon Lee
- Institute of Computer Technology, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul, 08826, South Korea
| | - Sangsoo Lim
- School of AI Software Convergence, Dongguk University, Pildong-ro 1-gil, Jung-gu, Seoul, South Korea
| | - Changyun Cho
- Interdisciplinary Program in Bioinformatics, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul, 08826, South Korea
| | - Yinhua Piao
- Department of Computer Science and Engineering, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul, 08826, South Korea
| | - Marie Yeo
- PHARMGENSCIENCE CO., LTD., 216, Dongjak-daero, Seocho-gu, Seoul, 06554, South Korea
| | - Dongkyu Kim
- PHARMGENSCIENCE CO., LTD., 216, Dongjak-daero, Seocho-gu, Seoul, 06554, South Korea
| | - Sun Kim
- Interdisciplinary Program in Artificial Intelligence, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul, 08826, South Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul, 08826, South Korea
- Department of Computer Science and Engineering, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul, 08826, South Korea
- AIGENDRUG CO., LTD., Gwanak-ro 1, Gwanak-gu, Seoul, 08826, South Korea
| | - Sunho Lee
- AIGENDRUG CO., LTD., Gwanak-ro 1, Gwanak-gu, Seoul, 08826, South Korea
| |
Collapse
|
9
|
Singh D, Singh A, Chawla PA. An overview of current strategies and future prospects in drug repurposing in tuberculosis. EXPLORATION OF MEDICINE 2023. [DOI: 10.37349/emed.2023.00125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
A large number of the population faces mortality as an effect of tuberculosis (TB). The line of treatment in the management of TB faces a jolt with ever-increasing multi-drug resistance (DR) cases. Further, the drugs engaged in the treatment of TB are associated with different toxicities, such as renal and hepatic toxicity. Different combinations are sought for effective anti-tuberculosis (anti-TB) effects with a decrease in toxicity. In this regard, drug repurposing has been very promising in improving the efficacy of drugs by enhancement of bioavailability and widening the safety margin. The success in drug repurposing lies in specified binding and inhibition of a particular target in the drug molecule. Different drugs have been repurposed for various ailments like cancer, Alzheimer’s disease, acquired immunodeficiency syndrome (AIDS), hair loss, etc. Repurposing in anti-TB drugs holds great potential too. The use of whole-cell screening assays and the availability of large chemical compounds for testing against Mycobacterium tuberculosis poses a challenge in this development. The target-based discovery of sites has emerged in the form of phenotypic screening as ethionamide R (EthR) and malate synthase inhibitors are similar to pharmaceuticals. In this review, the authors have thoroughly described the drug repurposing techniques on the basis of pharmacogenomics and drug metabolism, pathogen-targeted therapy, host-directed therapy, and bioinformatics approaches for the identification of drugs. Further, the significance of repurposing of drugs elaborated on large databases has been revealed. The role of genomics and network-based methods in drug repurposing has been also discussed in this article.
Collapse
Affiliation(s)
- Dilpreet Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Amrinder Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Pooja A. Chawla
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142001, Punjab, India
| |
Collapse
|
10
|
Sun G, Dong D, Dong Z, Zhang Q, Fang H, Wang C, Zhang S, Wu S, Dong Y, Wan Y. Drug repositioning: A bibliometric analysis. Front Pharmacol 2022; 13:974849. [PMID: 36225586 PMCID: PMC9549161 DOI: 10.3389/fphar.2022.974849] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/12/2022] [Indexed: 11/14/2022] Open
Abstract
Drug repurposing has become an effective approach to drug discovery, as it offers a new way to explore drugs. Based on the Science Citation Index Expanded (SCI-E) and Social Sciences Citation Index (SSCI) databases of the Web of Science core collection, this study presents a bibliometric analysis of drug repurposing publications from 2010 to 2020. Data were cleaned, mined, and visualized using Derwent Data Analyzer (DDA) software. An overview of the history and development trend of the number of publications, major journals, major countries, major institutions, author keywords, major contributors, and major research fields is provided. There were 2,978 publications included in the study. The findings show that the United States leads in this area of research, followed by China, the United Kingdom, and India. The Chinese Academy of Science published the most research studies, and NIH ranked first on the h-index. The Icahn School of Medicine at Mt Sinai leads in the average number of citations per study. Sci Rep, Drug Discov. Today, and Brief. Bioinform. are the three most productive journals evaluated from three separate perspectives, and pharmacology and pharmacy are unquestionably the most commonly used subject categories. Cheng, FX; Mucke, HAM; and Butte, AJ are the top 20 most prolific and influential authors. Keyword analysis shows that in recent years, most research has focused on drug discovery/drug development, COVID-19/SARS-CoV-2/coronavirus, molecular docking, virtual screening, cancer, and other research areas. The hotspots have changed in recent years, with COVID-19/SARS-CoV-2/coronavirus being the most popular topic for current drug repurposing research.
Collapse
Affiliation(s)
- Guojun Sun
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Dashun Dong
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Zuojun Dong
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Qian Zhang
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Hui Fang
- Institute of Information Resource, Zhejiang University of Technology, Hangzhou, China
| | - Chaojun Wang
- Hangzhou Aeronautical Sanatorium for Special Service of Chinese Air Force, Hangzhou, China
| | - Shaoya Zhang
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Shuaijun Wu
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Yichen Dong
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yuehua Wan
- Institute of Information Resource, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
11
|
Zhong C, Ai J, Yang Y, Ma F, Sun W. Small Molecular Drug Screening Based on Clinical Therapeutic Effect. Molecules 2022; 27:molecules27154807. [PMID: 35956770 PMCID: PMC9369618 DOI: 10.3390/molecules27154807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Virtual screening can significantly save experimental time and costs for early drug discovery. Drug multi-classification can speed up virtual screening and quickly predict the most likely class for a drug. In this study, 1019 drug molecules with actual therapeutic effects are collected from multiple databases and documents, and molecular sets are grouped according to therapeutic effect and mechanism of action. Molecular descriptors and molecular fingerprints are obtained through SMILES to quantify molecular structures. After using the Kennard–Stone method to divide the data set, a better combination can be obtained by comparing the combined results of five classification algorithms and a fusion method. Furthermore, for a specific data set, the model with the best performance is used to predict the validation data set. The test set shows that prediction accuracy can reach 0.862 and kappa coefficient can reach 0.808. The highest classification accuracy of the validation set is 0.873. The more reliable molecular set has been found, which could be used to predict potential attributes of unknown drug compounds and even to discover new use for old drugs. We hope this research can provide a reference for virtual screening of multiple classes of drugs at the same time in the future.
Collapse
Affiliation(s)
| | | | | | | | - Wei Sun
- Correspondence: ; Tel.: +86-10-64445826
| |
Collapse
|
12
|
Similarity-Based Method with Multiple-Feature Sampling for Predicting Drug Side Effects. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9547317. [PMID: 35401786 PMCID: PMC8993545 DOI: 10.1155/2022/9547317] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/18/2021] [Accepted: 03/15/2022] [Indexed: 12/23/2022]
Abstract
Drugs can treat different diseases but also bring side effects. Undetected and unaccepted side effects for approved drugs can greatly harm the human body and bring huge risks for pharmaceutical companies. Traditional experimental methods used to determine the side effects have several drawbacks, such as low efficiency and high cost. One alternative to achieve this purpose is to design computational methods. Previous studies modeled a binary classification problem by pairing drugs and side effects; however, their classifiers can only extract one feature from each type of drug association. The present work proposed a novel multiple-feature sampling scheme that can extract several features from one type of drug association. Thirteen classification algorithms were employed to construct classifiers with features yielded by such scheme. Their performance was greatly improved compared with that of the classifiers that use the features yielded by the original scheme. Best performance was observed for the classifier based on random forest with MCC of 0.8661, AUROC of 0.969, and AUPR of 0.977. Finally, one key parameter in the multiple-feature sampling scheme was analyzed.
Collapse
|
13
|
Wang X, Liu M, Zhang Y, He S, Qin C, Li Y, Lu T. Deep fusion learning facilitates anatomical therapeutic chemical recognition in drug repurposing and discovery. Brief Bioinform 2021; 22:6342939. [PMID: 34368838 DOI: 10.1093/bib/bbab289] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 01/17/2023] Open
Abstract
The advent of large-scale biomedical data and computational algorithms provides new opportunities for drug repurposing and discovery. It is of great interest to find an appropriate data representation and modeling method to facilitate these studies. The anatomical therapeutic chemical (ATC) classification system, proposed by the World Health Organization (WHO), is an essential source of information for drug repurposing and discovery. Besides, computational methods are applied to predict drug ATC classification. We conducted a systematic review of ATC computational prediction studies and revealed the differences in data sets, data representation, algorithm approaches, and evaluation metrics. We then proposed a deep fusion learning (DFL) framework to optimize the ATC prediction model, namely DeepATC. The methods based on graph convolutional network, inferring biological network and multimodel attentive fusion network were applied in DeepATC to extract the molecular topological information and low-dimensional representation from the molecular graph and heterogeneous biological networks. The results indicated that DeepATC achieved superior model performance with area under the curve (AUC) value at 0.968. Furthermore, the DFL framework was performed for the transcriptome data-based ATC prediction, as well as another independent task that is significantly relevant to drug discovery, namely drug-target interaction. The DFL-based model achieved excellent performance in the above-extended validation task, suggesting that the idea of aggregating the heterogeneous biological network and node's (molecule or protein) self-topological features will bring inspiration for broader drug repurposing and discovery research.
Collapse
Affiliation(s)
- Xiting Wang
- Life Science School, Beijing University of Chinese Medicine, Beijing, China
| | - Meng Liu
- Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Yiling Zhang
- Beijing University of Chinese Medicine, Beijing, China
| | - Shuangshuang He
- Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Caimeng Qin
- School of Life Sciences, Beijing University of Chinese Medicine and Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yu Li
- Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Tao Lu
- Integrative Medicine Center in School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Kashyap K, Siddiqi MI. Recent trends in artificial intelligence-driven identification and development of anti-neurodegenerative therapeutic agents. Mol Divers 2021; 25:1517-1539. [PMID: 34282519 DOI: 10.1007/s11030-021-10274-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022]
Abstract
Neurological disorders affect various aspects of life. Finding drugs for the central nervous system is a very challenging and complex task due to the involvement of the blood-brain barrier, P-glycoprotein, and the drug's high attrition rates. The availability of big data present in online databases and resources has enabled the emergence of artificial intelligence techniques including machine learning to analyze, process the data, and predict the unknown data with high efficiency. The use of these modern techniques has revolutionized the whole drug development paradigm, with an unprecedented acceleration in the central nervous system drug discovery programs. Also, the new deep learning architectures proposed in many recent works have given a better understanding of how artificial intelligence can tackle big complex problems that arose due to central nervous system disorders. Therefore, the present review provides comprehensive and up-to-date information on machine learning/artificial intelligence-triggered effort in the brain care domain. In addition, a brief overview is presented on machine learning algorithms and their uses in structure-based drug design, ligand-based drug design, ADMET prediction, de novo drug design, and drug repurposing. Lastly, we conclude by discussing the major challenges and limitations posed and how they can be tackled in the future by using these modern machine learning/artificial intelligence approaches.
Collapse
Affiliation(s)
- Kushagra Kashyap
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Central Drug Research Institute (CSIR-CDRI) Campus, Lucknow, India.,Molecular and Structural Biology Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Mohammad Imran Siddiqi
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Central Drug Research Institute (CSIR-CDRI) Campus, Lucknow, India. .,Molecular and Structural Biology Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
| |
Collapse
|
15
|
Zhao H, Zheng K, Li Y, Wang J. A novel graph attention model for predicting frequencies of drug-side effects from multi-view data. Brief Bioinform 2021; 22:6312959. [PMID: 34213525 DOI: 10.1093/bib/bbab239] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/30/2021] [Accepted: 06/04/2021] [Indexed: 12/15/2022] Open
Abstract
Identifying the frequencies of the drug-side effects is a very important issue in pharmacological studies and drug risk-benefit. However, designing clinical trials to determine the frequencies is usually time consuming and expensive, and most existing methods can only predict the drug-side effect existence or associations, not their frequencies. Inspired by the recent progress of graph neural networks in the recommended system, we develop a novel prediction model for drug-side effect frequencies, using a graph attention network to integrate three different types of features, including the similarity information, known drug-side effect frequency information and word embeddings. In comparison, the few available studies focusing on frequency prediction use only the known drug-side effect frequency scores. One novel approach used in this work first decomposes the feature types in drug-side effect graph to extract different view representation vectors based on three different type features, and then recombines these latent view vectors automatically to obtain unified embeddings for prediction. The proposed method demonstrates high effectiveness in 10-fold cross-validation. The computational results show that the proposed method achieves the best performance in the benchmark dataset, outperforming the state-of-the-art matrix decomposition model. In addition, some ablation experiments and visual analyses are also supplied to illustrate the usefulness of our method for the prediction of the drug-side effect frequencies. The codes of MGPred are available at https://github.com/zhc940702/MGPred and https://zenodo.org/record/4449613.
Collapse
Affiliation(s)
- Haochen Zhao
- School of Computer Science and Engineering, Central South University, Changsha 410083, China.,Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
| | - Kai Zheng
- School of Computer Science and Engineering, Central South University, Changsha 410083, China.,Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
| | - Yaohang Li
- Department of Computer Science, Old Dominion University, Norfolk, VA 23529-0001, United States
| | - Jianxin Wang
- School of Computer Science and Engineering, Central South University, Changsha 410083, China.,Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
| |
Collapse
|
16
|
Ding X, Mower J, Subramanian D, Cohen T. Augmenting aer2vec: Enriching distributed representations of adverse event report data with orthographic and lexical information. J Biomed Inform 2021; 119:103833. [PMID: 34111555 DOI: 10.1016/j.jbi.2021.103833] [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: 02/10/2021] [Revised: 05/10/2021] [Accepted: 06/02/2021] [Indexed: 11/29/2022]
Abstract
Adverse Drug Events (ADEs) are prevalent, costly, and sometimes preventable. Post-marketing drug surveillance aims to monitor ADEs that occur after a drug is released to market. Reports of such ADEs are aggregated by reporting systems, such as the Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS). In this paper, we consider the topic of how best to represent data derived from reports in FAERS for the purpose of detecting post-marketing surveillance signals, in order to inform regulatory decision making. In our previous work, we developed aer2vec, a method for deriving distributed representations (concept embeddings) of drugs and side effects from ADE reports, establishing the utility of distributional information for pharmacovigilance signal detection. In this paper, we advance this line of research further by evaluating the utility of encoding orthographic and lexical information. We do so by adapting two Natural Language Processing methods, subword embedding and vector retrofitting, which were developed to encode such information into word embeddings. Models were compared for their ability to distinguish between positive and negative examples in a set of manually curated drug/ADE relationships, with both aer2vec enhancements offering advantages in performances over baseline models, and best performance obtained when retrofitting and subword embeddings were applied in concert. In addition, this work demonstrates that models leveraging distributed representations do not require extensive manual preprocessing to perform well on this pharmacovigilance signal detection task, and may even benefit from information that would otherwise be lost during the normalization and standardization process.
Collapse
Affiliation(s)
- Xiruo Ding
- Department of Biomedical Informatics & Medical Education, University of Washington, Seattle, WA, USA.
| | - Justin Mower
- Department of Computer Science, Rice University, Houston, TX, USA.
| | | | - Trevor Cohen
- Department of Biomedical Informatics & Medical Education, University of Washington, Seattle, WA, USA.
| |
Collapse
|
17
|
Vatansever S, Schlessinger A, Wacker D, Kaniskan HÜ, Jin J, Zhou M, Zhang B. Artificial intelligence and machine learning-aided drug discovery in central nervous system diseases: State-of-the-arts and future directions. Med Res Rev 2021; 41:1427-1473. [PMID: 33295676 PMCID: PMC8043990 DOI: 10.1002/med.21764] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/30/2020] [Accepted: 11/20/2020] [Indexed: 01/11/2023]
Abstract
Neurological disorders significantly outnumber diseases in other therapeutic areas. However, developing drugs for central nervous system (CNS) disorders remains the most challenging area in drug discovery, accompanied with the long timelines and high attrition rates. With the rapid growth of biomedical data enabled by advanced experimental technologies, artificial intelligence (AI) and machine learning (ML) have emerged as an indispensable tool to draw meaningful insights and improve decision making in drug discovery. Thanks to the advancements in AI and ML algorithms, now the AI/ML-driven solutions have an unprecedented potential to accelerate the process of CNS drug discovery with better success rate. In this review, we comprehensively summarize AI/ML-powered pharmaceutical discovery efforts and their implementations in the CNS area. After introducing the AI/ML models as well as the conceptualization and data preparation, we outline the applications of AI/ML technologies to several key procedures in drug discovery, including target identification, compound screening, hit/lead generation and optimization, drug response and synergy prediction, de novo drug design, and drug repurposing. We review the current state-of-the-art of AI/ML-guided CNS drug discovery, focusing on blood-brain barrier permeability prediction and implementation into therapeutic discovery for neurological diseases. Finally, we discuss the major challenges and limitations of current approaches and possible future directions that may provide resolutions to these difficulties.
Collapse
Affiliation(s)
- Sezen Vatansever
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Avner Schlessinger
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Daniel Wacker
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - H. Ümit Kaniskan
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jian Jin
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ming‐Ming Zhou
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Bin Zhang
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
18
|
Al-Taie Z, Liu D, Mitchem JB, Papageorgiou C, Kaifi JT, Warren WC, Shyu CR. Explainable artificial intelligence in high-throughput drug repositioning for subgroup stratifications with interventionable potential. J Biomed Inform 2021; 118:103792. [PMID: 33915273 DOI: 10.1016/j.jbi.2021.103792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 03/26/2021] [Accepted: 04/21/2021] [Indexed: 01/02/2023]
Abstract
Enabling precision medicine requires developing robust patient stratification methods as well as drugs tailored to homogeneous subgroups of patients from a heterogeneous population. Developing de novo drugs is expensive and time consuming with an ultimately low FDA approval rate. These limitations make developing new drugs for a small portion of a disease population unfeasible. Therefore, drug repositioning is an essential alternative for developing new drugs for a disease subpopulation. This shows the importance of developing data-driven approaches that find druggable homogeneous subgroups within the disease population and reposition the drugs for these subgroups. In this study, we developed an explainable AI approach for patient stratification and drug repositioning. Contrast pattern mining and network analysis were used to discover homogeneous subgroups within a disease population. For each subgroup, a biomedical network analysis was done to find the drugs that are most relevant to a given subgroup of patients. The set of candidate drugs for each subgroup was ranked using an aggregated drug score assigned to each drug. The proposed method represents a human-in-the-loop framework, where medical experts use the data-driven results to generate hypotheses and obtain insights into potential therapeutic candidates for patients who belong to a subgroup. Colorectal cancer (CRC) was used as a case study. Patients' phenotypic and genotypic data was utilized with a heterogeneous knowledge base because it gives a multi-view perspective for finding new indications for drugs outside of their original use. Our analysis of the top candidate drugs for the subgroups identified by medical experts showed that most of these drugs are cancer-related, and most of them have the potential to be a CRC regimen based on studies in the literature.
Collapse
Affiliation(s)
- Zainab Al-Taie
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA; Department of Computer Science, College of Science for Women, University of Baghdad, Baghdad, Iraq
| | - Danlu Liu
- Electrical Engineering and Computer Science Department, University of Missouri, Columbia, MO 65211, USA
| | - Jonathan B Mitchem
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA; Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA.
| | - Christos Papageorgiou
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Jussuf T Kaifi
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA
| | - Wesley C Warren
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA; Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Department of Animal Sciences, Bond Life Sciences Center, University of Missouri, 1201 Rollins Street, Columbia, MO 65211, USA
| | - Chi-Ren Shyu
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA; Electrical Engineering and Computer Science Department, University of Missouri, Columbia, MO 65211, USA; Department of Medicine, School of Medicine, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
19
|
Shukla R, Henkel ND, Alganem K, Hamoud AR, Reigle J, Alnafisah RS, Eby HM, Imami AS, Creeden JF, Miruzzi SA, Meller J, Mccullumsmith RE. Signature-based approaches for informed drug repurposing: targeting CNS disorders. Neuropsychopharmacology 2021; 46:116-130. [PMID: 32604402 PMCID: PMC7688959 DOI: 10.1038/s41386-020-0752-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/30/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022]
Abstract
CNS disorders, and in particular psychiatric illnesses, lack definitive disease-altering therapeutics. The limited understanding of the mechanisms driving these illnesses with the slow pace and high cost of drug development exacerbates this issue. For these reasons, drug repurposing - both a less expensive and time-efficient practice compared to de novo drug development - has been a promising strategy to overcome the paucity of treatments available for these debilitating disorders. While empirical drug-repurposing has been a routine practice in clinical psychiatry, innovative, informed, and cost-effective repurposing efforts using big data ("omics") have been designed to characterize drugs by structural and transcriptomic signatures. These strategies, in conjunction with ontological integration, provide an important opportunity to address knowledge-based challenges associated with drug development for CNS disorders. In this review, we discuss various signature-based in silico approaches to drug repurposing, its integration with multiple omics platforms, and how this data can be used for clinically relevant, evidence-based drug repurposing. These tools provide an exciting translational avenue to merge omics-based drug discovery platforms with patient-specific disease signatures, ultimately facilitating the identification of new therapies for numerous psychiatric disorders.
Collapse
Affiliation(s)
- Rammohan Shukla
- Department of Neurosciences, University of Toledo, Toledo, OH, USA.
| | | | - Khaled Alganem
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | | | - James Reigle
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Hunter M Eby
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Ali S Imami
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Justin F Creeden
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Scott A Miruzzi
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Jaroslaw Meller
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Electrical Engineering and Computing Systems, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Informatics, Nicolaus Copernicus University, Torun, Poland
| | - Robert E Mccullumsmith
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
- Neurosciences Institute, ProMedica, Toledo, OH, USA
| |
Collapse
|
20
|
Kanza S, Graham Frey J. Semantic Technologies in Drug Discovery. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11520-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
21
|
Elgart M, Redline S, Sofer T. Machine and Deep Learning in Molecular and Genetic Aspects of Sleep Research. Neurotherapeutics 2021; 18:228-243. [PMID: 33829409 PMCID: PMC8116376 DOI: 10.1007/s13311-021-01014-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2021] [Indexed: 12/11/2022] Open
Abstract
Epidemiological sleep research strives to identify the interactions and causal mechanisms by which sleep affects human health, and to design intervention strategies for improving sleep throughout the lifespan. These goals can be advanced by further focusing on the environmental and genetic etiology of sleep disorders, and by development of risk stratification algorithms, to identify people who are at risk or are affected by, sleep disorders. These studies rely on comprehensive sleep-related data which often contains complex multi-dimensional physiological and molecular measurements across multiple timepoints. Thus, sleep research is well-suited for the application of computational approaches that can handle high-dimensional data. Here, we survey recent advances in machine and deep learning together with the availability of large human cohort studies with sleep data that can jointly drive the next breakthroughs in the sleep-research field. We describe sleep-related data types and datasets, and present some of the tasks in the field that can be targets for algorithmic approaches, as well as the challenges and opportunities in pursuing them.
Collapse
Affiliation(s)
- Michael Elgart
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA USA
- Department of Medicine, Harvard Medical School, Boston, MA USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA USA
- Department of Medicine, Harvard Medical School, Boston, MA USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA USA
- Department of Medicine, Harvard Medical School, Boston, MA USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
| |
Collapse
|
22
|
Yang J, Zhang D, Liu L, Li G, Cai Y, Zhang Y, Jin H, Chen X. Computational drug repositioning based on the relationships between substructure-indication. Brief Bioinform 2020; 22:6032618. [PMID: 33313675 DOI: 10.1093/bib/bbaa348] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/13/2020] [Accepted: 10/30/2020] [Indexed: 12/15/2022] Open
Abstract
At present, computational methods for drug repositioning are mainly based on the whole structures of drugs, which limits the discovery of new functions due to the similarities between local structures of drugs. In this article, we, for the first time, integrated the features of chemical-genomics (substructure-domain) and pharmaco-genomics (domain-indication) based on the assumption that drug-target interactions are mediated by the substructures of drugs and the domains of proteins to identify the relationships between substructure-indication and establish a drug-substructure-indication network for predicting all therapeutic effects of tested drugs through only information on the substructures of drugs. In total, 83 205 drug-indication relationships with different correlation scores were obtained. We used three different verification methods to indicate the accuracy of the method and the reliability of the scoring system. We predicted all indications of olaparib using our method, including the known antitumor effect and unknown antiviral effect verified by literature, and we also discovered the inhibitory mechanism of olaparib toward DNA repair through its specific sub494 (o = C-C: C), as it participates in the low synthesis of the poly subfunction of the apoptosis pathway (hsa04210) by inhibiting the Inositol 1,4,5-trisphosphate receptor(s) (ITPRs) and hydrolyzing poly (ADP ribose) polymerases. ElectroCardioGrams of four drugs (quinidine, amiodarone, milrinone and fosinopril) demonstrated the effect of anti-arrhythmia. Unlike previous studies focusing on the overall structures of drugs, our research has great potential in the search for more therapeutic effects of drugs and in predicting all potential effects and mechanisms of a drug from the local structural similarity.
Collapse
Affiliation(s)
- Jingbo Yang
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Denan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Lei Liu
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Guoqi Li
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Yiyang Cai
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Yan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Hongbo Jin
- Department of Physiology, Harbin Medical University
| | - Xiujie Chen
- College of Bioinformatics Science and Technology, Harbin Medical University
| |
Collapse
|
23
|
Huang L, Luo H, Li S, Wu FX, Wang J. Drug-drug similarity measure and its applications. Brief Bioinform 2020; 22:5956929. [PMID: 33152756 DOI: 10.1093/bib/bbaa265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 02/01/2023] Open
Abstract
Drug similarities play an important role in modern biology and medicine, as they help scientists gain deep insights into drugs' therapeutic mechanisms and conduct wet labs that may significantly improve the efficiency of drug research and development. Nowadays, a number of drug-related databases have been constructed, with which many methods have been developed for computing similarities between drugs for studying associations between drugs, human diseases, proteins (drug targets) and more. In this review, firstly, we briefly introduce the publicly available drug-related databases. Secondly, based on different drug features, interaction relationships and multimodal data, we summarize similarity calculation methods in details. Then, we discuss the applications of drug similarities in various biological and medical areas. Finally, we evaluate drug similarity calculation methods with common evaluation metrics to illustrate the important roles of drug similarity measures on different applications.
Collapse
Affiliation(s)
- Lan Huang
- Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering at Central South University, Hunan, China
| | - Huimin Luo
- School of Computer and Information Engineering at Henan University, Kaifeng, China
| | - Suning Li
- Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Fang-Xiang Wu
- College of Engineering and Department of Computer Sciences, University of Saskatchewan, Saskatoon, Canada
| | - Jianxin Wang
- Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering at Central South University, Hunan, China
| |
Collapse
|
24
|
Feng YH, Zhang SW, Shi JY. DPDDI: a deep predictor for drug-drug interactions. BMC Bioinformatics 2020; 21:419. [PMID: 32972364 PMCID: PMC7513481 DOI: 10.1186/s12859-020-03724-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 08/31/2020] [Indexed: 12/21/2022] Open
Abstract
Background The treatment of complex diseases by taking multiple drugs becomes increasingly popular. However, drug-drug interactions (DDIs) may give rise to the risk of unanticipated adverse effects and even unknown toxicity. DDI detection in the wet lab is expensive and time-consuming. Thus, it is highly desired to develop the computational methods for predicting DDIs. Generally, most of the existing computational methods predict DDIs by extracting the chemical and biological features of drugs from diverse drug-related properties, however some drug properties are costly to obtain and not available in many cases. Results In this work, we presented a novel method (namely DPDDI) to predict DDIs by extracting the network structure features of drugs from DDI network with graph convolution network (GCN), and the deep neural network (DNN) model as a predictor. GCN learns the low-dimensional feature representations of drugs by capturing the topological relationship of drugs in DDI network. DNN predictor concatenates the latent feature vectors of any two drugs as the feature vector of the corresponding drug pairs to train a DNN for predicting the potential drug-drug interactions. Experiment results show that, the newly proposed DPDDI method outperforms four other state-of-the-art methods; the GCN-derived latent features include more DDI information than other features derived from chemical, biological or anatomical properties of drugs; and the concatenation feature aggregation operator is better than two other feature aggregation operators (i.e., inner product and summation). The results in case studies confirm that DPDDI achieves reasonable performance in predicting new DDIs. Conclusion We proposed an effective and robust method DPDDI to predict the potential DDIs by utilizing the DDI network information without considering the drug properties (i.e., drug chemical and biological properties). The method should also be useful in other DDI-related scenarios, such as the detection of unexpected side effects, and the guidance of drug combination.
Collapse
Affiliation(s)
- Yue-Hua Feng
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Jian-Yu Shi
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
25
|
Zhou JP, Chen L, Guo ZH. iATC-NRAKEL: an efficient multi-label classifier for recognizing anatomical therapeutic chemical classes of drugs. Bioinformatics 2020; 36:1391-1396. [PMID: 31593226 DOI: 10.1093/bioinformatics/btz757] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/10/2019] [Accepted: 10/01/2019] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION The anatomical therapeutic chemical (ATC) classification system plays an increasingly important role in drug repositioning and discovery. The correct identification of classes in each level of such system that a given drug may belong to is an essential problem. Several multi-label classifiers have been proposed in this regard. Although they provided satisfactory performance, the feature extraction procedures were still rough. More refined features may further improve the predicted quality. RESULTS In this article, we provide a novel multi-label classifier, called iATC-NRAKEL, to predict drug ATC classes in the first level. To obtain more informative drug features, we employed the drug association information in STITCH and KEGG, which was organized by seven drug networks. The powerful network embedding algorithm, Mashup, was adopted to extract informative drug features. The obtained features were fed into the RAndom k-labELsets (RAKEL) algorithm with support vector machine as the basic classification algorithm to construct the classifier. The 10-fold cross-validation of the benchmark dataset with 3883 drugs showed that the accuracy and absolute true were 76.56 and 74.51%, respectively. The comparison results indicated that iATC-NRAKEL was much superior to all previous reported classifiers. Finally, the contribution of each network was analyzed. AVAILABILITY AND IMPLEMENTATION The codes of iATC-NRAKEL are available at https://github.com/zhou256/iATC-NRAKEL.
Collapse
Affiliation(s)
- Jian-Peng Zhou
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People's Republic of China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People's Republic of China.,Shanghai Key Laboratory of PMMP, East China Normal University, Shanghai 200241, People's Republic of China
| | - Zi-Han Guo
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People's Republic of China
| |
Collapse
|
26
|
Peng Y, Wang M, Xu Y, Wu Z, Wang J, Zhang C, Liu G, Li W, Li J, Tang Y. Drug repositioning by prediction of drug's anatomical therapeutic chemical code via network-based inference approaches. Brief Bioinform 2020; 22:2058-2072. [PMID: 32221552 DOI: 10.1093/bib/bbaa027] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/05/2020] [Accepted: 02/17/2020] [Indexed: 12/17/2022] Open
Abstract
Drug discovery and development is a time-consuming and costly process. Therefore, drug repositioning has become an effective approach to address the issues by identifying new therapeutic or pharmacological actions for existing drugs. The drug's anatomical therapeutic chemical (ATC) code is a hierarchical classification system categorized as five levels according to the organs or systems that drugs act and the pharmacology, therapeutic and chemical properties of drugs. The 2nd-, 3rd- and 4th-level ATC codes reserved the therapeutic and pharmacological information of drugs. With the hypothesis that drugs with similar structures or targets would possess similar ATC codes, we exploited a network-based approach to predict the 2nd-, 3rd- and 4th-level ATC codes by constructing substructure drug-ATC (SD-ATC), target drug-ATC (TD-ATC) and Substructure&Target drug-ATC (STD-ATC) networks. After 10-fold cross validation and two external validations, the STD-ATC models outperformed the SD-ATC and TD-ATC ones. Furthermore, with KR as fingerprint, the STD-ATC model was identified as the optimal model with AUC values at 0.899 ± 0.015, 0.916 and 0.893 for 10-fold cross validation, external validation set 1 and external validation set 2, respectively. To illustrate the predictive capability of the STD-ATC model with KR fingerprint, as a case study, we predicted 25 FDA-approved drugs (22 drugs were actually purchased) to have potential activities on heart failure using that model. Experiments in vitro confirmed that 8 of the 22 old drugs have shown mild to potent cardioprotective activities on both hypoxia model and oxygen-glucose deprivation model, which demonstrated that our STD-ATC prediction model would be an effective tool for drug repositioning.
Collapse
Affiliation(s)
- Yayuan Peng
- East China University of Science and Technology, Shanghai, China
| | - Manjiong Wang
- East China University of Science and Technology, Shanghai, China
| | - Yixiang Xu
- East China University of Science and Technology, Shanghai, China
| | - Zengrui Wu
- East China University of Science and Technology, Shanghai, China
| | - Jiye Wang
- East China University of Science and Technology, Shanghai, China
| | - Chao Zhang
- East China University of Science and Technology, Shanghai, China
| | - Guixia Liu
- East China University of Science and Technology, Shanghai, China
| | - Weihua Li
- East China University of Science and Technology, Shanghai, China
| | - Jian Li
- East China University of Science and Technology, Shanghai, China
| | - Yun Tang
- East China University of Science and Technology, Shanghai, China
| |
Collapse
|
27
|
Rifaioglu AS, Atas H, Martin MJ, Cetin-Atalay R, Atalay V, Doğan T. Recent applications of deep learning and machine intelligence on in silico drug discovery: methods, tools and databases. Brief Bioinform 2019; 20:1878-1912. [PMID: 30084866 PMCID: PMC6917215 DOI: 10.1093/bib/bby061] [Citation(s) in RCA: 261] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 05/25/2018] [Indexed: 01/16/2023] Open
Abstract
The identification of interactions between drugs/compounds and their targets is crucial for the development of new drugs. In vitro screening experiments (i.e. bioassays) are frequently used for this purpose; however, experimental approaches are insufficient to explore novel drug-target interactions, mainly because of feasibility problems, as they are labour intensive, costly and time consuming. A computational field known as 'virtual screening' (VS) has emerged in the past decades to aid experimental drug discovery studies by statistically estimating unknown bio-interactions between compounds and biological targets. These methods use the physico-chemical and structural properties of compounds and/or target proteins along with the experimentally verified bio-interaction information to generate predictive models. Lately, sophisticated machine learning techniques are applied in VS to elevate the predictive performance. The objective of this study is to examine and discuss the recent applications of machine learning techniques in VS, including deep learning, which became highly popular after giving rise to epochal developments in the fields of computer vision and natural language processing. The past 3 years have witnessed an unprecedented amount of research studies considering the application of deep learning in biomedicine, including computational drug discovery. In this review, we first describe the main instruments of VS methods, including compound and protein features (i.e. representations and descriptors), frequently used libraries and toolkits for VS, bioactivity databases and gold-standard data sets for system training and benchmarking. We subsequently review recent VS studies with a strong emphasis on deep learning applications. Finally, we discuss the present state of the field, including the current challenges and suggest future directions. We believe that this survey will provide insight to the researchers working in the field of computational drug discovery in terms of comprehending and developing novel bio-prediction methods.
Collapse
Affiliation(s)
- Ahmet Sureyya Rifaioglu
- Department of Computer Engineering, Middle East Technical University, Ankara, Turkey
- Department of Computer Engineering, İskenderun Technical University, Hatay, Turkey
| | - Heval Atas
- Cancer System Biology Laboratory (CanSyL), Graduate School of Informatics, Middle East Technical University, Ankara, Turkey
| | - Maria Jesus Martin
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL–EBI), Cambridge, Hinxton, UK
| | - Rengul Cetin-Atalay
- Department of Computer Engineering, Middle East Technical University, Ankara, Turkey
| | - Volkan Atalay
- Department of Computer Engineering, Middle East Technical University, Ankara, Turkey
| | - Tunca Doğan
- Cancer System Biology Laboratory (CanSyL), Graduate School of Informatics, Middle East Technical University, Ankara, Turkey and European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL–EBI), Cambridge, Hinxton, UK
| |
Collapse
|
28
|
Yang X, Wang Y, Byrne R, Schneider G, Yang S. Concepts of Artificial Intelligence for Computer-Assisted Drug Discovery. Chem Rev 2019; 119:10520-10594. [PMID: 31294972 DOI: 10.1021/acs.chemrev.8b00728] [Citation(s) in RCA: 413] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Artificial intelligence (AI), and, in particular, deep learning as a subcategory of AI, provides opportunities for the discovery and development of innovative drugs. Various machine learning approaches have recently (re)emerged, some of which may be considered instances of domain-specific AI which have been successfully employed for drug discovery and design. This review provides a comprehensive portrayal of these machine learning techniques and of their applications in medicinal chemistry. After introducing the basic principles, alongside some application notes, of the various machine learning algorithms, the current state-of-the art of AI-assisted pharmaceutical discovery is discussed, including applications in structure- and ligand-based virtual screening, de novo drug design, physicochemical and pharmacokinetic property prediction, drug repurposing, and related aspects. Finally, several challenges and limitations of the current methods are summarized, with a view to potential future directions for AI-assisted drug discovery and design.
Collapse
Affiliation(s)
- Xin Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| | - Yifei Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| | - Ryan Byrne
- ETH Zurich , Department of Chemistry and Applied Biosciences , Vladimir-Prelog-Weg 4 , CH-8093 Zurich , Switzerland
| | - Gisbert Schneider
- ETH Zurich , Department of Chemistry and Applied Biosciences , Vladimir-Prelog-Weg 4 , CH-8093 Zurich , Switzerland
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| |
Collapse
|
29
|
Gazerani P. Identification of novel analgesics through a drug repurposing strategy. Pain Manag 2019; 9:399-415. [DOI: 10.2217/pmt-2018-0091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The identification of new indications for approved or failed drugs is a process called drug repositioning or drug repurposing. The motivation includes overcoming the productivity gap that exists in drug development, which is a high-cost–high-risk process. Repositioning also includes rescuing drugs that have safely entered the market but have failed to demonstrate sufficient efficiency for the initial clinical indication. Considering the high prevalence of chronic pain, the lack of sufficient efficacy and the safety issues of current analgesics, repositioning seems to be an attractive approach. This review presents example of drugs that already have been repositioned and highlights new technologies that are available for the identification of additional compounds to stimulate the curiosity of readers for further exploration.
Collapse
Affiliation(s)
- Parisa Gazerani
- Biomedicine, Department of Health Science & Technology, Aalborg University, Frederik Bajers Vej 3 B, 9220 Aalborg East, Denmark
| |
Collapse
|
30
|
Park K. A review of computational drug repurposing. Transl Clin Pharmacol 2019; 27:59-63. [PMID: 32055582 PMCID: PMC6989243 DOI: 10.12793/tcp.2019.27.2.59] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/23/2019] [Accepted: 06/24/2019] [Indexed: 12/21/2022] Open
Abstract
Although sciences and technology have progressed rapidly, de novo drug development has been a costly and time-consuming process over the past decades. In view of these circumstances, ‘drug repurposing’ (or ‘drug repositioning’) has appeared as an alternative tool to accelerate drug development process by seeking new indications for already approved drugs rather than discovering de novo drug compounds, nowadays accounting for 30% of newly marked drugs in the U.S. In the meantime, the explosive and large-scale growth of molecular, genomic and phenotypic data of pharmacological compounds is enabling the development of new area of drug repurposing called computational drug repurposing. This review provides an overview of recent progress in the area of computational drug repurposing. First, it summarizes available repositioning strategies, followed by computational methods commonly used. Then, it describes validation techniques for repurposing studies. Finally, it concludes by discussing the remaining challenges in computational repurposing.
Collapse
Affiliation(s)
- Kyungsoo Park
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
31
|
Zhao K, So HC. Drug Repositioning for Schizophrenia and Depression/Anxiety Disorders: A Machine Learning Approach Leveraging Expression Data. IEEE J Biomed Health Inform 2019; 23:1304-1315. [DOI: 10.1109/jbhi.2018.2856535] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
32
|
Zhou X, Dai E, Song Q, Ma X, Meng Q, Jiang Y, Jiang W. In silico drug repositioning based on drug-miRNA associations. Brief Bioinform 2019; 21:498-510. [DOI: 10.1093/bib/bbz012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/14/2018] [Accepted: 01/11/2019] [Indexed: 02/06/2023] Open
Abstract
Abstract
Drug repositioning has become a prevailing tactic as this strategy is efficient, economical and low risk for drug discovery. Meanwhile, recent studies have confirmed that small-molecule drugs can modulate the expression of disease-related miRNAs, which indicates that miRNAs are promising therapeutic targets for complex diseases. In this study, we put forward and verified the hypothesis that drugs with similar miRNA profiles may share similar therapeutic properties. Furthermore, a comprehensive drug–drug interaction network was constructed based on curated drug-miRNA associations. Through random network comparison, topological structure analysis and network module extraction, we found that the closely linked drugs in the network tend to treat the same diseases. Additionally, the curated drug–disease relationships (from the CTD) and random walk with restarts algorithm were utilized on the drug–drug interaction network to identify the potential drugs for a given disease. Both internal validation (leave-one-out cross-validation) and external validation (independent drug–disease data set from the ChEMBL) demonstrated the effectiveness of the proposed approach. Finally, by integrating drug-miRNA and miRNA-disease information, we also explain the modes of action of drugs in the view of miRNA regulation. In summary, our work could determine novel and credible drug indications and offer novel insights and valuable perspectives for drug repositioning.
Collapse
Affiliation(s)
- Xu Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Enyu Dai
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Qian Song
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Xueyan Ma
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Qianqian Meng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Yongshuai Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, P. R. China
| | - Wei Jiang
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, P. R. China
| |
Collapse
|
33
|
Extractive stripping voltammetry at carbon paste electrodes for determination of biologically active organic compounds. MONATSHEFTE FUR CHEMIE 2019. [DOI: 10.1007/s00706-018-2346-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
34
|
Shi JY, Li JX, Mao KT, Cao JB, Lei P, Lu HM, Yiu SM. Predicting combinative drug pairs via multiple classifier system with positive samples only. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2019; 168:1-10. [PMID: 30527128 DOI: 10.1016/j.cmpb.2018.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 10/24/2018] [Accepted: 11/12/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND AND OBJECTIVE Due to the synergistic effects of drugs, drug combination is one of the effective approaches for treating complex diseases. However, the identification of drug combinations by dose-response methods is still costly. It is promising to develop supervised learning-based approaches to predict potential drug combinations on a large scale. Nevertheless, these approaches have the inadequate utilization of heterogeneous features, which causes the loss of information useful to classification. Moreover, they have an intrinsic bias, because they assume unknown drug pairs as non-combinations, of which some could be real drug combinations in practice. METHODS To address above issues, this work first designs a two-layer multiple classifier system (TLMCS) to effectively integrate heterogeneous features involving anatomical therapeutic chemical codes of drugs, drug-drug interactions, drug-target interactions, gene ontology of drug targets, and side effects. To avoid the bias caused by labelling unknown samples as negative, it then utilizes the one-class support vector machines, (which requires no negative instance and only labels approved drug combinations as positive instances), as the member classifiers in TLMCS. Last, both a 10-fold cross validation (10-CV) and a novel prediction are performed to validate the performance of TLMCS. RESULTS The comparison with three state-of-the-art approaches under 10-CV exhibits the superiority of TLMCS, which achieves the area under the receiver operating characteristic curve = 0.824 and the area under the precision-recall curve = 0.372. Moreover, the experiment under the novel prediction demonstrates its ability, where 9 out of the top-20 predicted combinative drug pairs are validated by checking the published literature. Furthermore, for each of the newly-validated drug combinations, this work analyses the combining mode of the member drugs and investigates their relationship in terms of drug targeting pathways. CONCLUSIONS The proposed TLMCS provides an effective framework to integrate those heterogeneous features and is trained by only positive samples such that the bias of taking unknown drug pairs as negative samples can be avoided. Furthermore, its results in the novel prediction reveal five types of drug combinations and three types of drug relationships in terms of pathways.
Collapse
Affiliation(s)
- Jian-Yu Shi
- School of Life Science, Northwestern Polytechnical University, China.
| | - Jia-Xin Li
- School of Life Science, Northwestern Polytechnical University, China.
| | - Kui-Tao Mao
- School of Computer Science, Northwestern Polytechnical University, China.
| | - Jiang-Bo Cao
- School of Life Science, Northwestern Polytechnical University, China.
| | - Peng Lei
- Department of Chinese Medicine, Shaanxi Provincial People's Hospital, China.
| | - Hui-Meng Lu
- School of Life Science, Northwestern Polytechnical University, China.
| | - Siu-Ming Yiu
- Department of Computer Science, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
35
|
Zhao X, Chen L, Lu J. A similarity-based method for prediction of drug side effects with heterogeneous information. Math Biosci 2018; 306:136-144. [PMID: 30296417 DOI: 10.1016/j.mbs.2018.09.010] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/22/2018] [Accepted: 09/25/2018] [Indexed: 12/25/2022]
Abstract
Drugs can produce intended therapeutic effects to treat different diseases. However, they may also cause side effects at the same time. For an approved drug, it is best to detect all side effects it can produce. Otherwise, it may bring great risks for pharmaceuticals companies as well as be harmful to human body. It is urgent to design quick and reliable identification methods to detect the side effects for a given drug. In this study, a binary classification model was proposed to predict drug side effects. Different from most previous methods, our model termed the pair of drug and side effect as a sample and convert the original problem to a binary classification problem. Based on the similarity idea, each pair was represented by five features, each of which was derived from a type of drug property. The strong machine learning algorithm, random forest, was adopted as the prediction engine. The ten-fold cross-validation on five datasets with different negative samples indicated that the proposed model yielded a good performance of Matthews correlation coefficient around 0.550 and AUC around 0.8492. In addition, we also analyzed the contribution of each drug property for construction of the model. The results indicated that drug similarity in fingerprint was most related to the prediction of drug side effects and all drug properties gave less or more contributions.
Collapse
Affiliation(s)
- Xian Zhao
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People's Republic of China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People's Republic of China; Shanghai Key Laboratory of PMMP, East China Normal University, Shanghai 200241, People's Republic of China.
| | - Jing Lu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, People's Republic of China
| |
Collapse
|
36
|
Yella JK, Yaddanapudi S, Wang Y, Jegga AG. Changing Trends in Computational Drug Repositioning. Pharmaceuticals (Basel) 2018; 11:E57. [PMID: 29874824 PMCID: PMC6027196 DOI: 10.3390/ph11020057] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/01/2018] [Accepted: 06/02/2018] [Indexed: 12/12/2022] Open
Abstract
Efforts to maximize the indications potential and revenue from drugs that are already marketed are largely motivated by what Sir James Black, a Nobel Prize-winning pharmacologist advocated-"The most fruitful basis for the discovery of a new drug is to start with an old drug". However, rational design of drug mixtures poses formidable challenges because of the lack of or limited information about in vivo cell regulation, mechanisms of genetic pathway activation, and in vivo pathway interactions. Hence, most of the successfully repositioned drugs are the result of "serendipity", discovered during late phase clinical studies of unexpected but beneficial findings. The connections between drug candidates and their potential adverse drug reactions or new applications are often difficult to foresee because the underlying mechanism associating them is largely unknown, complex, or dispersed and buried in silos of information. Discovery of such multi-domain pharmacomodules-pharmacologically relevant sub-networks of biomolecules and/or pathways-from collection of databases by independent/simultaneous mining of multiple datasets is an active area of research. Here, while presenting some of the promising bioinformatics approaches and pipelines, we summarize and discuss the current and evolving landscape of computational drug repositioning.
Collapse
Affiliation(s)
- Jaswanth K Yella
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way MLC 7024, Cincinnati, OH 45229, USA.
| | - Suryanarayana Yaddanapudi
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way MLC 7024, Cincinnati, OH 45229, USA.
| | - Yunguan Wang
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way MLC 7024, Cincinnati, OH 45229, USA.
| | - Anil G Jegga
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way MLC 7024, Cincinnati, OH 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
- Department of Computer Science, University of Cincinnati College of Engineering, Cincinnati, OH 45219, USA.
| |
Collapse
|
37
|
Sylvestre E, Bouzillé G, Chazard E, His-Mahier C, Riou C, Cuggia M. Combining information from a clinical data warehouse and a pharmaceutical database to generate a framework to detect comorbidities in electronic health records. BMC Med Inform Decis Mak 2018; 18:9. [PMID: 29368609 PMCID: PMC5784648 DOI: 10.1186/s12911-018-0586-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 01/05/2018] [Indexed: 11/25/2022] Open
Abstract
Background Medical coding is used for a variety of activities, from observational studies to hospital billing. However, comorbidities tend to be under-reported by medical coders. The aim of this study was to develop an algorithm to detect comorbidities in electronic health records (EHR) by using a clinical data warehouse (CDW) and a knowledge database. Methods We enriched the Theriaque pharmaceutical database with the French national Comorbidities List to identify drugs associated with at least one major comorbid condition and diagnoses associated with a drug indication. Then, we compared the drug indications in the Theriaque database with the ICD-10 billing codes in EHR to detect potentially missing comorbidities based on drug prescriptions. Finally, we improved comorbidity detection by matching drug prescriptions and laboratory test results. We tested the obtained algorithm by using two retrospective datasets extracted from the Rennes University Hospital (RUH) CDW. The first dataset included all adult patients hospitalized in the ear, nose, throat (ENT) surgical ward between October and December 2014 (ENT dataset). The second included all adult patients hospitalized at RUH between January and February 2015 (general dataset). We reviewed medical records to find written evidence of the suggested comorbidities in current or past stays. Results Among the 22,132 Common Units of Dispensation (CUD) codes present in the Theriaque database, 19,970 drugs (90.2%) were associated with one or several ICD-10 diagnoses, based on their indication, and 11,162 (50.4%) with at least one of the 4878 comorbidities from the comorbidity list. Among the 122 patients of the ENT dataset, 75.4% had at least one drug prescription without corresponding ICD-10 code. The comorbidity diagnoses suggested by the algorithm were confirmed in 44.6% of the cases. Among the 4312 patients of the general dataset, 68.4% had at least one drug prescription without corresponding ICD-10 code. The comorbidity diagnoses suggested by the algorithm were confirmed in 20.3% of reviewed cases. Conclusions This simple algorithm based on combining accessible and immediately reusable data from knowledge databases, drug prescriptions and laboratory test results can detect comorbidities. Electronic supplementary material The online version of this article (10.1186/s12911-018-0586-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emmanuelle Sylvestre
- INSERM, U1099, F-35000, Rennes, France. .,Université de Rennes 1, LTSI, F-35000, Rennes, France. .,CHU Rennes, CIC Inserm 1414, F-35000, Rennes, France. .,CHU Rennes, Centre de Données Cliniques, F-35000, Rennes, France.
| | - Guillaume Bouzillé
- INSERM, U1099, F-35000, Rennes, France.,Université de Rennes 1, LTSI, F-35000, Rennes, France.,CHU Rennes, CIC Inserm 1414, F-35000, Rennes, France.,CHU Rennes, Centre de Données Cliniques, F-35000, Rennes, France
| | - Emmanuel Chazard
- Département de Santé Publique, Université de Lille EA 2694, CHU Lille, F-59000, Lille, France
| | - Cécil His-Mahier
- INSERM, U1099, F-35000, Rennes, France.,Université de Rennes 1, LTSI, F-35000, Rennes, France.,CHU Rennes, CIC Inserm 1414, F-35000, Rennes, France.,CHU Rennes, Centre de Données Cliniques, F-35000, Rennes, France
| | - Christine Riou
- INSERM, U1099, F-35000, Rennes, France.,Université de Rennes 1, LTSI, F-35000, Rennes, France.,CHU Rennes, CIC Inserm 1414, F-35000, Rennes, France.,CHU Rennes, Centre de Données Cliniques, F-35000, Rennes, France
| | - Marc Cuggia
- INSERM, U1099, F-35000, Rennes, France.,Université de Rennes 1, LTSI, F-35000, Rennes, France.,CHU Rennes, CIC Inserm 1414, F-35000, Rennes, France.,CHU Rennes, Centre de Données Cliniques, F-35000, Rennes, France
| |
Collapse
|
38
|
Chen L, Liu T, Zhao X. Inferring anatomical therapeutic chemical (ATC) class of drugs using shortest path and random walk with restart algorithms. Biochim Biophys Acta Mol Basis Dis 2017; 1864:2228-2240. [PMID: 29247833 DOI: 10.1016/j.bbadis.2017.12.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 12/01/2017] [Accepted: 12/12/2017] [Indexed: 01/02/2023]
Abstract
The anatomical therapeutic chemical (ATC) classification system is a widely accepted drug classification scheme. This system comprises five levels and includes several classes in each level. Drugs are classified into classes according to their therapeutic effects and characteristics. The first level includes 14 main classes. In this study, we proposed two network-based models to infer novel potential chemicals deemed to belong in the first level of ATC classification. To build these models, two large chemical networks were constructed using the chemical-chemical interaction information retrieved from the Search Tool for Interactions of Chemicals (STITCH). Two classic network algorithms, shortest path (SP) and random walk with restart (RWR) algorithms, were executed on the corresponding network to mine novel chemicals for each ATC class using the validated drugs in a class as seed nodes. Then, the obtained chemicals yielded by these two algorithms were further evaluated by a permutation test and an association test. The former can exclude chemicals produced by the structure of the network, i.e., false positive discoveries. By contrast, the latter identifies the most important chemicals that have strong associations with the ATC class. Comparisons indicated that the two models can provide quite dissimilar results, suggesting that the results yielded by one model can be essential supplements for those obtained by the other model. In addition, several representative inferred chemicals were analyzed to confirm the reliability of the results generated by the two models. This article is part of a Special Issue entitled: Accelerating Precision Medicine through Genetic and Genomic Big Data Analysis edited by Yudong Cai & Tao Huang.
Collapse
Affiliation(s)
- Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People's Republic of China.
| | - Tao Liu
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People's Republic of China.
| | - Xian Zhao
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People's Republic of China
| |
Collapse
|
39
|
Shi JY, Li JX, Gao K, Lei P, Yiu SM. Predicting combinative drug pairs towards realistic screening via integrating heterogeneous features. BMC Bioinformatics 2017; 18:409. [PMID: 29072137 PMCID: PMC5657064 DOI: 10.1186/s12859-017-1818-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
Background Drug Combination is one of the effective approaches for treating complex diseases. However, determining combinative drug pairs in clinical trials is still costly. Thus, computational approaches are used to identify potential drug pairs in advance. Existing computational approaches have the following shortcomings: (i) the lack of an effective integration of heterogeneous features leads to a time-consuming training and even results in an over-fitted classifier; and (ii) the narrow consideration of predicting potential drug combinations only among known drugs having known combinations cannot meet the demand of realistic screenings, which pay more attention to potential combinative pairs among newly-coming drugs that have no approved combination with other drugs at all. Results In this paper, to tackle the above two problems, we propose a novel drug-driven approach for predicting potential combinative pairs on a large scale. We define four new features based on heterogeneous data and design an efficient fusion scheme to integrate these feature. Moreover importantly, we elaborate appropriate cross-validations towards realistic screening scenarios of drug combinations involving both known drugs and new drugs. In addition, we perform an extra investigation to show how each kind of heterogeneous features is related to combinative drug pairs. The investigation inspires the design of our approach. Experiments on real data demonstrate the effectiveness of our fusion scheme for integrating heterogeneous features and its predicting power in three scenarios of realistic screening. In terms of both AUC and AUPR, the prediction among known drugs achieves 0.954 and 0.821, that between known drugs and new drugs achieves 0.909 and 0.635, and that among new drugs achieves 0.809 and 0.592 respectively. Conclusions Our approach provides not only an effective tool to integrate heterogeneous features but also the first tool to predict potential combinative pairs among new drugs.
Collapse
Affiliation(s)
- Jian-Yu Shi
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Jia-Xin Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Ke Gao
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Peng Lei
- Department of Chinese Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Siu-Ming Yiu
- Department of Computer Science, the University of Hong Kong, Hong Kong, China.
| |
Collapse
|
40
|
Bolgár B, Antal P. VB-MK-LMF: fusion of drugs, targets and interactions using variational Bayesian multiple kernel logistic matrix factorization. BMC Bioinformatics 2017; 18:440. [PMID: 28978313 PMCID: PMC5628496 DOI: 10.1186/s12859-017-1845-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/21/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Computational fusion approaches to drug-target interaction (DTI) prediction, capable of utilizing multiple sources of background knowledge, were reported to achieve superior predictive performance in multiple studies. Other studies showed that specificities of the DTI task, such as weighting the observations and focusing the side information are also vital for reaching top performance. METHOD We present Variational Bayesian Multiple Kernel Logistic Matrix Factorization (VB-MK-LMF), which unifies the advantages of (1) multiple kernel learning, (2) weighted observations, (3) graph Laplacian regularization, and (4) explicit modeling of probabilities of binary drug-target interactions. RESULTS VB-MK-LMF achieves significantly better predictive performance in standard benchmarks compared to state-of-the-art methods, which can be traced back to multiple factors. The systematic evaluation of the effect of multiple kernels confirm their benefits, but also highlights the limitations of linear kernel combinations, already recognized in other fields. The analysis of the effect of prior kernels using varying sample sizes sheds light on the balance of data and knowledge in DTI tasks and on the rate at which the effect of priors vanishes. This also shows the existence of "small sample size" regions where using side information offers significant gains. Alongside favorable predictive performance, a notable property of MF methods is that they provide a unified space for drugs and targets using latent representations. Compared to earlier studies, the dimensionality of this space proved to be surprisingly low, which makes the latent representations constructed by VB-ML-LMF especially well-suited for visual analytics. The probabilistic nature of the predictions allows the calculation of the expected values of hits in functionally relevant sets, which we demonstrate by predicting drug promiscuity. The variational Bayesian approximation is also implemented for general purpose graphics processing units yielding significantly improved computational time. CONCLUSION In standard benchmarks, VB-MK-LMF shows significantly improved predictive performance in a wide range of settings. Beyond these benchmarks, another contribution of our work is highlighting and providing estimates for further pharmaceutically relevant quantities, such as promiscuity, druggability and total number of interactions.
Collapse
Affiliation(s)
- Bence Bolgár
- Department of Measurement and Information Systems, Budapest University of Technology and Economics, Magyar tudósok krt. 2., Budapest, 1117 Hungary
| | - Péter Antal
- Department of Measurement and Information Systems, Budapest University of Technology and Economics, Magyar tudósok krt. 2., Budapest, 1117 Hungary
| |
Collapse
|
41
|
Olson T, Singh R. Predicting anatomic therapeutic chemical classification codes using tiered learning. BMC Bioinformatics 2017; 18:266. [PMID: 28617230 PMCID: PMC5471942 DOI: 10.1186/s12859-017-1660-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The low success rate and high cost of drug discovery requires the development of new paradigms to identify molecules of therapeutic value. The Anatomical Therapeutic Chemical (ATC) Code System is a World Health Organization (WHO) proposed classification that assigns multi-level codes to compounds based on their therapeutic, pharmacological and chemical characteristics as well as the in-vivo sites(s) of activity. The ability to predict ATC codes of compounds can assist in creation of high-quality chemical libraries for drug screening and in applications such as drug repositioning. We propose a machine learning architecture called tiered learning for prediction of ATC codes that relies on the prediction results of the higher levels of the ATC code to simplify the predictions of the lower levels. RESULTS The proposed approach was validated using a number of compounds in both cross-validation and test setting. The validation experiments compared chemical descriptors, initialization methods and classification algorithms. The prediction accuracy obtained with tiered learning was found to be either comparable or better than that of established methods. Additionally, the experiments demonstrated the generalizability of the tiered learning architecture, in that its use was found to improve prediction rates for a majority of machine learning algorithms when compared to their stand-alone application. CONCLUSION The basis of our approach lies in the observation that anatomical-therapeutic biological activity of certain types typically precludes activities of many other types. Thus, there exists a characteristic distribution of the ATC codes, which can be leveraged to limit the search-space of possible codes that can be ascribed at a particular level once the codes at the preceding levels are known. Tiered learning utilizes this observation to constrain the learning space for ATC codes at a particular level based on the ATC code at higher levels. This simplifies the prediction and allows for improved accuracy.
Collapse
Affiliation(s)
- Thomas Olson
- Department of Computer Science, San Francisco State University, San Francisco, CA, USA
| | - Rahul Singh
- Department of Computer Science, San Francisco State University, San Francisco, CA, USA. .,Center for Discovery and Innovation in Parasitic Diseases, University of California, San Diego, CA, USA.
| |
Collapse
|
42
|
Fu X, Mervin LH, Li X, Yu H, Li J, Mohamad Zobir SZ, Zoufir A, Zhou Y, Song Y, Wang Z, Bender A. Toward Understanding the Cold, Hot, and Neutral Nature of Chinese Medicines Using in Silico Mode-of-Action Analysis. J Chem Inf Model 2017; 57:468-483. [DOI: 10.1021/acs.jcim.6b00725] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Xianjun Fu
- School
of Information Management, Shandong University of Traditional Chinese Medicine, 250355 Jinan, China
- Centre
for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Lewis H. Mervin
- Centre
for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Xuebo Li
- School
of Information Management, Shandong University of Traditional Chinese Medicine, 250355 Jinan, China
| | - Huayun Yu
- College
of TCM, Shandong University of Traditional Chinese Medicine, 250355 Jinan, China
| | - Jiaoyang Li
- School
of Information Management, Shandong University of Traditional Chinese Medicine, 250355 Jinan, China
| | - Siti Zuraidah Mohamad Zobir
- Centre
for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Azedine Zoufir
- Centre
for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Yang Zhou
- School
of Information Management, Shandong University of Traditional Chinese Medicine, 250355 Jinan, China
| | - Yongmei Song
- School
of Information Management, Shandong University of Traditional Chinese Medicine, 250355 Jinan, China
| | - Zhenguo Wang
- School
of Information Management, Shandong University of Traditional Chinese Medicine, 250355 Jinan, China
| | - Andreas Bender
- Centre
for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
43
|
Generating Gene Ontology-Disease Inferences to Explore Mechanisms of Human Disease at the Comparative Toxicogenomics Database. PLoS One 2016; 11:e0155530. [PMID: 27171405 PMCID: PMC4865041 DOI: 10.1371/journal.pone.0155530] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/29/2016] [Indexed: 12/20/2022] Open
Abstract
Strategies for discovering common molecular events among disparate diseases hold promise for improving understanding of disease etiology and expanding treatment options. One technique is to leverage curated datasets found in the public domain. The Comparative Toxicogenomics Database (CTD; http://ctdbase.org/) manually curates chemical-gene, chemical-disease, and gene-disease interactions from the scientific literature. The use of official gene symbols in CTD interactions enables this information to be combined with the Gene Ontology (GO) file from NCBI Gene. By integrating these GO-gene annotations with CTD’s gene-disease dataset, we produce 753,000 inferences between 15,700 GO terms and 4,200 diseases, providing opportunities to explore presumptive molecular underpinnings of diseases and identify biological similarities. Through a variety of applications, we demonstrate the utility of this novel resource. As a proof-of-concept, we first analyze known repositioned drugs (e.g., raloxifene and sildenafil) and see that their target diseases have a greater degree of similarity when comparing GO terms vs. genes. Next, a computational analysis predicts seemingly non-intuitive diseases (e.g., stomach ulcers and atherosclerosis) as being similar to bipolar disorder, and these are validated in the literature as reported co-diseases. Additionally, we leverage other CTD content to develop testable hypotheses about thalidomide-gene networks to treat seemingly disparate diseases. Finally, we illustrate how CTD tools can rank a series of drugs as potential candidates for repositioning against B-cell chronic lymphocytic leukemia and predict cisplatin and the small molecule inhibitor JQ1 as lead compounds. The CTD dataset is freely available for users to navigate pathologies within the context of extensive biological processes, molecular functions, and cellular components conferred by GO. This inference set should aid researchers, bioinformaticists, and pharmaceutical drug makers in finding commonalities in disease mechanisms, which in turn could help identify new therapeutics, new indications for existing pharmaceuticals, potential disease comorbidities, and alerts for side effects.
Collapse
|
44
|
New strategy for drug discovery by large-scale association analysis of molecular networks of different species. Sci Rep 2016; 6:21872. [PMID: 26912056 PMCID: PMC4766474 DOI: 10.1038/srep21872] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/02/2016] [Indexed: 12/13/2022] Open
Abstract
The development of modern omics technology has not significantly improved the efficiency of drug development. Rather precise and targeted drug discovery remains unsolved. Here a large-scale cross-species molecular network association (CSMNA) approach for targeted drug screening from natural sources is presented. The algorithm integrates molecular network omics data from humans and 267 plants and microbes, establishing the biological relationships between them and extracting evolutionarily convergent chemicals. This technique allows the researcher to assess targeted drugs for specific human diseases based on specific plant or microbe pathways. In a perspective validation, connections between the plant Halliwell-Asada (HA) cycle and the human Nrf2-ARE pathway were verified and the manner by which the HA cycle molecules act on the human Nrf2-ARE pathway as antioxidants was determined. This shows the potential applicability of this approach in drug discovery. The current method integrates disparate evolutionary species into chemico-biologically coherent circuits, suggesting a new cross-species omics analysis strategy for rational drug development.
Collapse
|
45
|
Liu Z, Guo F, Wang Y, Li C, Zhang X, Li H, Diao L, Gu J, Wang W, Li D, He F. BATMAN-TCM: a Bioinformatics Analysis Tool for Molecular mechANism of Traditional Chinese Medicine. Sci Rep 2016; 6:21146. [PMID: 26879404 PMCID: PMC4754750 DOI: 10.1038/srep21146] [Citation(s) in RCA: 522] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 01/19/2016] [Indexed: 02/05/2023] Open
Abstract
Traditional Chinese Medicine (TCM), with a history of thousands of years of clinical practice, is gaining more and more attention and application worldwide. And TCM-based new drug development, especially for the treatment of complex diseases is promising. However, owing to the TCM's diverse ingredients and their complex interaction with human body, it is still quite difficult to uncover its molecular mechanism, which greatly hinders the TCM modernization and internationalization. Here we developed the first online Bioinformatics Analysis Tool for Molecular mechANism of TCM (BATMAN-TCM). Its main functions include 1) TCM ingredients' target prediction; 2) functional analyses of targets including biological pathway, Gene Ontology functional term and disease enrichment analyses; 3) the visualization of ingredient-target-pathway/disease association network and KEGG biological pathway with highlighted targets; 4) comparison analysis of multiple TCMs. Finally, we applied BATMAN-TCM to Qishen Yiqi dripping Pill (QSYQ) and combined with subsequent experimental validation to reveal the functions of renin-angiotensin system responsible for QSYQ's cardioprotective effects for the first time. BATMAN-TCM will contribute to the understanding of the "multi-component, multi-target and multi-pathway" combinational therapeutic mechanism of TCM, and provide valuable clues for subsequent experimental validation, accelerating the elucidation of TCM's molecular mechanism. BATMAN-TCM is available at http://bionet.ncpsb.org/batman-tcm.
Collapse
Affiliation(s)
- Zhongyang Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100850, China.,National Center for Protein Sciences Beijing, Beijing 102206, China
| | - Feifei Guo
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Yong Wang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chun Li
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xinlei Zhang
- Beijing Genestone Technology Ltd., Beijing 100085, China
| | - Honglei Li
- Beijing Genestone Technology Ltd., Beijing 100085, China
| | - Lihong Diao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100850, China.,National Center for Protein Sciences Beijing, Beijing 102206, China
| | - Jiangyong Gu
- Beijing National Laboratory for Molecular Sciences, State Key Lab of Rare Earth Material Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Wei Wang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Dong Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100850, China.,National Center for Protein Sciences Beijing, Beijing 102206, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100850, China.,National Center for Protein Sciences Beijing, Beijing 102206, China
| |
Collapse
|